Open access peer-reviewed chapter - ONLINE FIRST

Neurological Impact of Type I Interferon Dysregulation

Written By

Alessio Mylonas

Submitted: 13 June 2024 Reviewed: 24 June 2024 Published: 31 July 2024

DOI: 10.5772/intechopen.1006023

Rare Neurodegenerative Disorders - New Insights IntechOpen
Rare Neurodegenerative Disorders - New Insights Edited by Liam Chen

From the Edited Volume

Rare Neurodegenerative Disorders - New Insights [Working Title]

Liam Chen

Chapter metrics overview

50 Chapter Downloads

View Full Metrics

Abstract

Type I interferons are a class of potent and tightly regulated cytokines important for antiviral and anti-tumoural innate and adaptive immunity. Dysregulated production can have serious neurologic consequences as exemplified in a family of rare diseases called type I interferonopathies. Interferonopathies represent a group of genetically determined conditions characterised by upregulated type I interferon production causing a spectrum of neuroinflammatory and systemic manifestations. This chapter delves into the historical discovery of type I interferons, their role in innate immunity, and the subsequent identification of interferonopathies placing emphasis on the mechanisms of neurologic dysfunction that often dominate the clinical picture. The insights gained from studying these rare diseases offer valuable lessons for neurodegenerative and neuropsychiatric conditions which demonstrate considerable overlap with interferonopathies, underscoring the broader significance of type I interferons in more common neurologic diseases. Relevant therapeutic strategies targeting this pathway are discussed, emphasising the need for brain-penetrant approaches.

Keywords

  • type I interferon
  • IFNα
  • IFNβ
  • IFNAR
  • Type I interferonopathies
  • Aicardi-Goutières syndrome
  • Alzheimer’s disease
  • Down’s syndrome
  • Systemic lupus erythematosus
  • Neurolupus
  • Interferon therapies
  • Inflammaging
  • Traumatic brain injury
  • HIV/AIDS-associated neurocognitive disorders
  • major depression
  • anifrolumab
  • anti-IFNAR1
  • CXCL10
  • CXCR3

1. Introduction

Type I interferonopathies, or simply interferonopathies, are a family of rare autoinflammatory diseases typified by overt sustained activation of type I interferon signalling. In their most characteristic appearance, interferonopathies are a form of neuroinflammatory disease with a genetic origin. In recent years, type I interferons have been implicated in an increasing number of neurologic diseases, raising the question of whether a better understanding of interferonopathies can enhance our knowledge of neurodegeneration mechanisms.

In this chapter, interferonopathies and the type I interferon pathway are introduced, and they are contextualised with our current knowledge of the biology and the underlying immunologic mechanisms. This is then examined in relation to the continuously expanding spectrum of neuropsychiatric diseases induced by interferon dysfunction. While interferonopathies are rare monogenic diseases, dysregulation of the type I interferon pathway has important implications for more common polygenic diseases. Our understanding of this pathway, of these rare interferonopathies, and of the more common interferon dysfunction diseases that they have parallels with, has allowed for great advances in the development of interferon as a therapeutic target with potentially great future clinical implications.

1.1 Historical perspective

Type I interferonopathies are a recent concept encompassing a diverse family of diseases. The term was first introduced by Yanick Crow in 2011 who proposed a unifying notion for several Mendelian diseases that share clinical and molecular features and, importantly, which involve type I interferon dysregulation [1]. Yet the history of type I interferonopathies dates all the way back to the early 1980s, when the deleterious effects of interferons were first observed and hypothesised.

The discovery of type I interferons is a fascinating story of scientific serendipity and perseverance. Interferons have been studied since the mid-1950s, when famous Alick Isaacs and Jean Lindenmann reported the discovery of a soluble factor that could interfere with the replication of influenza virus which they named “interferon” [2, 3]. Initially, the two microbiologists were studying the influenza virus and the resulting cytopathic activity in chick cells and remarked that some cells were more resistant to the infection and importantly that this resistance could be transferred. While fraught with resistance and scepticism from the research community, and hampered by technological limitations, this discovery paved the way for unprecedented therapeutic advances. Years later, it was discovered that many different interferons exist, with broadly overlapping induction mechanisms, functions, and biological effects. Importantly, it was discovered that exogenous delivery could be leveraged for therapeutic benefit in patients.

The first human use of interferons occurred in 1973. A non-purified interferon preparation was delivered intranasally to healthy volunteers later exposed to rhinovirus, demonstrating antiviral immunity in humans for the first time [4]. Aside from their antiviral function, anti-tumour activities were described [5] paving the way for the first antitumour use in humans [6] and the first approval of recombinant IFNα therapy for hairy cell leukaemia in 1986. Later, immunoregulatory effects were also unintentionally discovered in multiple sclerosis (MS), an autoimmune disease, reasoned to be triggered by viral infections [7]. Today, type I interferons, the very same subtype that Isaacs and Lindenmann had first discovered, are used in the treatment of many viral infections, cancers, and even multiple sclerosis. They have also become essential for our understanding of the molecular and cellular basis of innate immunity and host-pathogen interactions. In parallel, as their use increased in the clinics, it very quickly became clear that they also induced psychocognitive effects [8, 9, 10], and that they may also induce de novo autoimmunity [11].

In 1978, Ion Gresser, a pioneer of research on the antiviral and antitumoral effects of interferons, demonstrated the counterintuitive benefits of IFNα-neutralising immunoglobulins during acute disease caused by infection [12]. This important observation was the first of many that would contrast the beneficial and detrimental of effects of type I interferons [13] and set the tone for the field for years to come. In many respects, this finding also foretold the great medical advances that would later be anti-cytokine therapies, first with anti-TNF in 1992 [14] and today with the first approved anti-interferon signalling therapies [15].

Around the same time, Jean Aicardi and Françoise Goutières reported eight children with severe early onset encephalopathy, characterised by calcification of the basal ganglia, white matter abnormalities, and calcification of the basal ganglia [16], but the underlying pathomechanism remained unclear. Fuelled by great interest in viral immunity, and the effects of interferons, Aicardi and colleagues investigated the cerebrospinal fluid and sera of these patients, and found that they had high levels of IFNα in both, yet could not attribute a viral aetiology to this [17]. It was many years later that Yanick Crow, Thomas Lindahl, and colleagues attributed to the Aicardi-Goutieres syndrome (AGS) to loss-of-function mutations of the TREX1 gene encoding a DNA exonuclease [18] and of the RNASEH2A, RNASEH2B, and RNASEH2C genes encoding subunits of the RNA endonuclease RNASEH2 [19]. AGS would later be recognised as the archetypical type I interferonopathy, and one of many which are still being discovered (Figure 1).

Figure 1.

Chronological overview of type I interferon-related discoveries. From the discovery of the virus-induced “interference” factors in 1957, type I interferons have been centre-stage for therapeutic development, both as exogenous administration, as disease biomarker, and ultimately as anti-cytokine therapy. From the first in human use of interferons first as an antiviral in 1973 and later for the treatment of MS and tumours in 1981, to the first approval of recombinant IFNα in 1986, type I interferons have provided numerous clinical benefits for patients across different disease areas. In the space of 16 years, interferons had gone from discovery to clinical use, a remarkable achievement for the time. Concurrently, reports of psychocognitive side effects were starting to appear from 1980 onwards, a testament to the immediacy of the effect in the CNS of these highly bioactive cytokines. It quickly became apparent that interferon therapy could lead to numerous other side effects such as SLE, and its involvement in autoinflammatory and autoimmune diseases was thereby discovered. The first description of Aicardi-Goutières Syndrome 1984, prior to the genomics revolution of the 2000s, would lead to the discovery of a new and expanding disease family called “interferonopathies”. Despite the first preclinical evidence of the benefits of anti-interferon biologic use in 1978, it would take 43 years to the first approval of an anti-interferon signalling therapy, a full 28 years after the first anti-cytokine biologic therapy. The list of potential indications that could benefit from targeted inhibition of the type I interferon pathway is still being determined with ongoing clinical trials and case studies across different autoinflammatory and autoimmune diseases.

1.2 Definition and classification of interferonopathies

Few cohort studies have so far been published, and many aspects are derived from case reports. Although type I interferonopathies are defined by similar underlying mechanisms, they are a family of diseases with distinct genetic alterations and a classification has been formulated based on clinical and genetic features (Table 1). Broadly, these can be sub-classified as monogenic interferonopathies and non-monogenic type I interferon dysfunction diseases, forming classical interferonopathies, brain vasculopathies, and one autoimmune disease.

FamilyDiseaseGenes affectedClinical characteristicsNeurologic symptoms
Classical interferonopathiesAGSTREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, IFIH1, DNASE2, RNU7-1, LSM11Intracranial calcification, skin inflammation, hepatosplenomegaly, thrombocytopenia, elevated liver enzymes, cerebrospinal fluid lymphocytosisProgressive encephalopathy, microcephaly, spasticity, dystonia, seizures, cognitive impairment, white matter abnormalities
USP18 loss of functionUSP18AGS, pseudo-TORCH syndromeMicrocephaly, intracranial calcifications, brain atrophy, seizures, developmental delay, hearing loss
ISG15 loss of functionISG15AGS, dermatologic symptoms, increased susceptibility to mycobacterial infectionsSeizures, developmental delay, microcephaly, ataxia, hypotonia, dystonia, spasticity, leukoencephalopathy
SPENCDACP5Short stature, spinal involvement, dermatologic manifestationsIntracranial calcifications, spasticity, developmental delay
PRAASPSMB4, PSMB8, PSMB9, PSMB10, PSMB12, PSMA3, PSMG2, POMPRecurrent fevers, nodular erythema, pernio-like rash, joint contractures, severe inflammationBrain atrophy, encephalopathy, basal ganglia calcification
SAVISTING1Skin lesions, interstitial lung disease, pulmonary hypertensionCerebral vasculitis, ischemic stroke, intracranial hemorrhage
SMSRIGI, IFIH1Aortic and mitral valve calcifications, dental dysplasia, osteoporosis, muscle weakness, delayed growthHeadache, mood disorders, psychosis, seizures, stroke, neuropathy, myelopathy
Brain vasculopathiesRVCLTREX1Vision loss, neurological involvement, kidney, liver, gastrointestinal, thyroid, and bone diseaseBrain lesions, strokes, brain atrophy, dementia, headache, dizziness, seizures, paralysis of cranial nerves, cerebral infarcts and haemorrhage
Interferon-associated TMAUnknownEndothelial dysfunction, microangiopathic hemolytic anemia, microvascular ischemia, kidney injury, hypertensionHeadache, mental confusion, loss of cognitive function, loss of memory, slowing of speech, hemiparesis
Kohlmeier Degos diseaseUnknownPapules with porcelain-white center and red border, gastrointestinal complications, neurological symptomsHeadache, vision loss, diplopia, papilledema, partial loss of vision, shortness of breath, chest pain, epilepsy, thickening of pericardium
Autoimmune diseasesSLETREX1, DNASE1, DNASE1L3, PRDM1, IRF5, IRF7, STAT4, TNFAIP3, TNFSF4, C1QA, C1QB, C1QCFever, rash, arthritis, serositis, nephritis, cytopenias, antinuclear antibodiesNeuropsychiatric manifestations, such as headache, mood disorders, psychosis, seizures, stroke, neuropathy, myelopathy2

Table 1.

Type I interferonopathies associated with neurological dysfunction.

AGS: Aicardi-Goutières Syndrome; USP18: Ubiquitin specific peptidase 18; ISG15: Interferon-stimulated gene 15; SPENCD: Spondyloenchondrodysplasia; PRAAS: Proteasome-associated autoinflammatory syndromes; SAVI: STING-associated vasculopathy with onset in infancy; SMS: Singleton-Merten syndrome; RVCL: Retinal vasculopathy with cerebral leukodystrophy; TMA: thrombotic microangiopathy; SLE: Systemic lupus erythematosus.

Interferonopathies are a monogenic family of diseases caused by mutations in genes involved in the recognition, production, or regulation of type I interferons. These mutations lead to nucleic acid or metabolite accumulation, activating cytosolic nucleic acid sensors. These sensors then induce the production and secretion of type I interferons, which bind to their receptors on the cell surface and initiate a signalling cascade that involves the phosphorylation and nuclear translocation of transcription factors. They thereby activate expression of hundreds of interferon-stimulated genes (ISGs), which mediate the antiviral, immunomodulatory, and inflammatory effects of type I interferons.

Interferonopathies can be classified into six main diseases, based on the clinical presentation and the genetic defect: (1) Aicardi-Goutières syndrome (AGS), (2) USP18 and ISG15 loss-of-function diseases, (3) spondyloenchondrodysplasia (SPENCD), (4) proteasome-associated autoinflammatory syndromes (PRAAS), (5) STING-associated vasculopathy with onset in infancy (SAVI), and (6) Singleton-Merten syndrome (SMS).

AGS is the most common among type I interferonopathies and is characterised by early-onset encephalopathy, calcification of the basal ganglia, leukodystrophy, skin lesions, and systemic inflammation. It is caused by mutations in genes encoding nucleases, such as TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR1, or genes involved in nucleic acid metabolism, such as RNASEH2, IFIH1, and ADAR2. These mutations impair the degradation or editing of endogenous nucleic acids, leading to the accumulation of self-DNA or RNA, which activate the cGAS-STING or RIG-I-MDA5 pathways, respectively. Gain-of-function mutations in STAT2 have also been identified [20, 21, 22, 23], bypassing nucleic acid metabolism steps and highlighting the complexity of AGS. USP18 and ISG15 loss-of-function diseases share important similarities and display neuropathology similar to AGS. SPENCD and PRAAS are less well studied, and the mechanism by which type I interferons are induced is undefined, though patients also display basal ganglia calcifications.

Among the typically non encephalitogenic type I interferonopathies, SAVI is perhaps the better studied. It is caused by gain-of-function mutations in STING1 (previously TMEM173), encoding the stimulator of interferon genes (STING) protein. STING is a key adaptor protein that activates type I interferon signalling in response to cytosolic DNA. It is diagnosed by genetic testing and by blood measurements of interferon response. Unlike AGS, systemic, rather than principally CNS-centred, symptoms are more pronounced, and often quite disparate. Early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease have been described, with rare cases of accompanying alopecia [24]. The skin lesions typically affect the fingers, toes, ears, and nose and can lead to ulceration, necrosis, and sometimes amputation. The lung disease manifests as progressive fibrosis, respiratory failure, and pulmonary hypertension. What is surprising is that, often these symptoms can occur without simultaneous involvement of both skin and lungs. Yet, upon imaging, occasionally intracerebral calcifications are also observed and can aid diagnosis. Only a handful of cases have been reported so far; thus, many questions remain as to the differential clinical signs. SMS, while also a classical interferonopathy, presents with a yet different array of symptoms, including skin inflammation, calcifications of the aorta, and progressive osteoporosis. Neurologic symptoms have also been reported though they are not thought as the main clinical sign.

With more attentive clinical investigations, additional rare type I interferonopathies are being discovered. It is important to note that not all of them present with overt neurological signs and often certain subtypes will include patients with severe psychocognitive disease while others display only systemic signs of disease. The underlying cause for this is largely unknown. Throughout the rest of the chapter, special emphasis will be put on encephalopathic diseases.

1.3 Overview of type I interferon pathway

Being a complex and tightly regulated network of molecular interactions, this pathway mediates cellular responses to infections, as well as endogenous nucleic acids when recognised as danger signals. The pathway can be divided into three overarching stages: (1) sensing, (2) signalling, and (3) feedback regulation (Figure 2).

Figure 2.

Type I interferon signalling regulation and pathway. Simplified summary of triggering molecules, associated sensors, type I interferons produced, receptor-mediated signalling, and generic interferon-responses.

Sensing refers to the recognition of pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs) by pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), and cGAS-STING. These receptors are expressed in different cellular compartments, such as the plasma membrane (TLR1, -2, -4, -5, -6), endosomes (TLR3, -7, -8, -9), cytosol (MDA-5, RIG-I, cGAS-STING, ZBP1, etc.), or nucleus (IFI16, hnRNPA2B1, cGAS in some contexts, etc.), and can detect different types of nucleic acids, such as viral or bacterial DNA or RNA, or self-DNA or RNA. Upon ligand binding, these receptors activate downstream signalling pathways that converge on the activation of interferon regulatory transcription factor 3 (IRF3) and 7 (IRF7), the master transcription factors of type I interferons [25].

Signalling initiates when binding to its cognate receptor is achieved, triggering the induction of downstream pathways and amplification of type I interferon production. IRF3 and IRF7 translocate to the nucleus and bind to the interferon-stimulated response elements (ISREs) in the promoters of type I interferon genes, such as IFNA1, IFNA2, and IFNB1, and induce their transcription, translation, and secretion. They then bind to their receptor, the interferon α/β receptor (IFNAR) formed of two chains: IFNAR1 and IFNAR2. This signalling can take place on the same or neighbouring cells and activate the Janus kinase (JAK) signal transducer and activator of transcription (STAT) pathway, which involves the phosphorylation and nuclear translocation of STAT1 and -2. These STATs form a complex with IRF9, called ISGF3, which binds to the ISREs in the promoters of ISGs, such as MX1, OAS1, and ISG15, and induce their transcription and expression. ISGs mediate the antiviral, immunomodulatory, and inflammatory effects of type I interferons. Other non-canonical pathways independent of JAK-STAT are also known to be activated in a cell- or context-specific manner, and signalling is mediated through PI3K-mTOR, NF-κB, and MAPK and can have broad outcomes [26, 27, 28].

Feedback regulation is initiated simultaneously following signalling, triggering both positive and negative modulation mechanisms for the pathway. Positive feedback loops involve the induction of IRF7, which enhances the expression of more type I interferons and ISGs, creating a feedforward loop that amplifies the response. Negative feedback loops involve the induction of suppressors of cytokine signalling (SOCS), protein inhibitors of activated STATs (PIAS), and ubiquitin-specific proteases (USPs), which inhibit the JAK-STAT pathway, or the induction of tripartite motif-containing proteins (TRIMs), which degrade IRF3 and IRF7, or the induction of microRNAs, such as miR-146a and miR-155, which silence certain mRNAs which are key components of the pathway. Ultimately, type I interferons are cytokines that play a crucial role in the innate immune response, but also act at the interface of adaptive immunity [29, 30, 31] and, as such, need to be tightly regulated. When its signalling is aberrantly activated or sustained, it can lead to a variety of clinical manifestations, ranging from autoinflammatory syndromes to autoimmune disorders.

Advertisement

2. Neurologic disease in type I interferonopathies

Type I interferonopathies are a group of rare monogenic disorders characterised by constitutive activation of type I interferon signalling, leading to chronic inflammation and multiorgan damage. Severe interferonopathies, and typically the most frequent ones, are first and foremost neuroinflammatory disorders. The most common clinical signs of type I interferonopathies are neurological, affecting the central and peripheral nervous systems. These include developmental delay, intellectual disability, seizures, spasticity, ataxia, dystonia, neuropathy, and hearing loss. The exact incidence and prevalence of type I interferonopathies are for the most part unknown, but estimates place incidence between 1:10,000 and 1:1,000,000 [32, 33, 34]. While the exact percentage of each type I interferonopathy among the total cases is also uncertain, the most common is Aicardi-Goutières syndrome (AGS).

2.1 Aicardi-Goutières syndrome

AGS is an interferonopathy, typically exemplified as a neuroinflammatory encephalopathy resembling congenital viral infection. By most accounts, AGS is the most common of the interferonopathies, and the better studied [34]. Clinically, AGS can present as neonatal-onset AGS or late-onset. Neonatal-onset can often be mistaken for a transplacental-acquired infection. When evidence for an obvious infection is lacking, common practice is that AGS should be considered. Symptoms during the first few weeks of life include slowed cognitive growth, abnormal movements, ataxia, and epileptic seizures, as well as systemic signs of infection including persistent fever. Late-onset AGS is challenging as symptoms may occur several months later alongside otherwise normal infantile development. Slowed growth of head circumference, spasticity, and weakness may sometimes be readily apparent. Imaging in both early and late onset forms often reveals microcephaly, intracranial calcification, leukoencephalopathy, necrosis, vasculopathy with aneurysms, infarcts, and sometimes discernible haemorrhage [35]. On a histopathological level, acute demyelinating lesions reveal the extent of neurological damage, especially in late-onset patients, with diffuse demyelination reminiscent of acute demyelinating encephalomyelitis [36]. Microangiopathy is also a common occurrence [37, 38, 39, 40]. Calcium deposits form around the blood vessels, and it is thought that this is attributed to excessive cell senescence and apoptosis [41].

Type I interferons are found consistently in CSF and serum at the neonatal stage [17, 42]. As interferons display high bioactivity reflected by a short half-life, they are notoriously difficult to detect, and in the early days, cytopathic protection assays were performed. Simply, CSF from patients was found to contain sufficient interferons to avert toxicity associated with vesicular stomatitis virus challenge in a recipient cell line. Simple experiments first gave clues as to which interferon subtypes were present in patients with AGS, pinpointing IFNα as the principal culprit. Leaps in bioassay technologies have allowed detection down to 0.1 femtograms, confirming on average 1000-fold increase in IFNα compared to healthy individuals [42, 43]. Analysis of the largest AGS cohort to date reveals that significantly higher levels of IFNα in CSF are consistently found in earlier onset, more severe, disease supporting the notion of a detrimental role of type I interferons that is dose dependent [44]. Intriguingly, this is not observed consistently in serum [44] which supports that the production is central rather than systemic in AGS. Importantly, interferon-response gene expression has allowed the definition of an interferon-score, which is now a widely used, highly sensitive, rapid, cheap, and specific interferon metric [42, 45, 46, 47, 48, 49, 50].

Mutations in nine genes have been identified to this day, all of which are involved in nucleic acid detection and metabolism. Overall, loss-of-function mutations lead to deficiencies in the clearance of nucleic acids, while the gain-of-function mutations cause overt sensing of nucleic acids. Mutations in TREX1 [18], RNASEH2A, RNASEH2B, RNASEH2C [19], SAMHD1 [51], and ADAR1 [52] result in the accumulation of nucleic acids, sometimes derived from endogenous retroviral elements, which result in potent activation of nucleic acid sensors. In particular, TREX1 is an important exonuclease, whose inactivation leads to accumulated DNAs in the cytoplasm that trigger overexpression of type I interferons [18] through the cGAS-STING pathway [53, 54]. Beyond AGS, mutations have been described in systemic lupus erythematosus (SLE) [55] and familial chilblain lupus (FCL) [56], linking together AGS and lupus. Similarly, mutations in RNASEH2A/B/C, which form a protein complex that degrades RNA:DNA hybrids and excises incorrectly inserted ribonucleotide monophosphates during DNA replication, lead to DNA damage and enhanced generation of byproducts of DNA repair [57]. Accumulated DNA repair metabolites in the cytoplasm caused by RNASEH2 mutants stimulate the cGAS-STING pathway, resulting in AGS [58]. Lastly, mutations in LSM11 and RNU7-1, which encode two components of the canonical replication-dependent histone pre-mRNA processing complex, cause impaired processing of mRNAs [59]. This leads to the release of cGAS from nucleosomes, thus binding to proximal nuclear DNA, and activates the cGAS-STING-TBK1 pathway inducing the expression of type I interferons [59]. Lastly, mutations in ADAR1 cause aberrant activation of nucleic acid sensors MDA5 and ZBP1 through enhancement of recognition of endogenous retroviral elements and causing uncontrolled type I interferon production [60, 61, 62]. These mechanisms exemplify the importance of accurate regulation of endogenous nucleic acid sensing and how their loss-of-function inevitably results in overt type I interferon production. Similar aberrant responses can also happen with gain-of-function mutations. Mutations in IFIH1 have resulted in overactivity of its gene product, MDA5, causing it to bind to RNA more avidly [63, 64]. This mimics excessive nucleic acid sensing and also results in type I interferon overproduction. While the genetic mutations and genes affected are numerous, mechanisms of AGS are related to aberrant nucleic acid sensing and defective DNA or RNA degradation. Mutations that cause dysfunctional negative regulation also exist, leading to other encephalopathic interferonopathies with substantial clinical overlap.

2.2 Other encephalopathic interferonopathies

Though rare, many other encephalopathic type I interferonopathies have been identified. Loss-of-function mutations in ISG15 cause disease similar to AGS and SPENCD including calcifications of the cerebral basal ganglia and type I interferon signatures [65] but can also cause dermatologic disease as often seen with interferonopathies [66]. It is recognised that ISG15, itself an interferon-response gene, is important for limiting type I interferon production, and that disease in ISG15 deficiency is systemic, but the exact contribution to disease from within the brain is undetermined. Interestingly, it is through its interaction with USP18, mutations of which are also associated with AGS [67], that ISG15 can limit type I interferon signalling by displacing JAK1 from IFNAR [68, 69]. Importantly, in vivo loss of USP18 has been studied in rodents, and it was found to cause unabated – of otherwise tonic self-limiting – type I interferon signalling and to lead to the generation of over phagocytic microglia causing white matter damage and neurodegeneration [70].

Rare mutations in a lysosomal protein encoded by ACP5, also cause a type I interferonopathy called Spondyloenchondrodysplasia (SPENCD). It is characterised by skeletal dysplasia typified by vertebral abnormalities and metaphyseal lesions of the long bones. Concurrently, brain calcifications with neurological impairment, and high type I interferon signatures, reminiscent of classical AGS, are also observed [71, 72]. Little is known about the pathological mechanism, yet these mutations result in a highly penetrant monogenic systemic lupus erythematosus (SLE) manifestation [73, 74, 75], suggestive of a broader relationship between type I interferonopathies and lupus.

Advertisement

3. Encephalopathic non-monogenic type I interferonopathies

Systemic lupus erythematosus (SLE) is a disease driven by type I interferons and characterised by neuropsychiatric involvement. Interestingly, many overlapping psychocognitive disturbances are also observed as side effects from exogenous interferon therapies.

3.1 Neuropsychiatric involvement in lupus

Systemic lupus erythematosus is the historical and immunological archetype autoimmune disease, affecting multiple systems including the CNS. It is estimated that almost four million people worldwide live with SLE [76]. More than half of SLE patients exhibit some neuropsychiatric manifestation [77, 78] though the exact prevalence and list of symptoms describing it are debatable [79, 80]. It is associated with more severe cases as demonstrated by a threefold higher mortality rate than SLE patients without obvious neuropsychiatric affliction [78, 79]. Confusional state, anxiety, cognitive dysfunction, mood disorder, and psychosis are some common manifestations of neuropsychiatric lupus, delineating a group of syndromes [81]. Further evidence of neuronal involvement in lupus comes from increased neurofilament light (NfL) in both plasma and CSF [82, 83, 84]. NfL is a bonafide marker of neuronal damage that gets released and drained into the CSF, which pours out to the circulation, proportionally to the extent of axonal damage [85]. In SLE, NfL is further increased in patients with obvious neuropsychiatric involvement [83, 84] and correlates to neurocognitive and motor function [86].

SLE and AGS display some important commonalities that ultimately define both diseases as interferonopathies. Variants in TREX1, which cause AGS, also increase the risk of SLE [55, 87, 88], and particularly neuropsychiatric lupus [87]. Therefore, it is not surprising that type I interferons are strongly implicated in SLE [42, 89] and have been found to be pathogenic since targeting either IFNα [90, 91] or IFNAR signalling [92, 93] ameliorates disease progression. Higher IFNα in serum is also a strong predictor of mortality and correlates with neurological manifestations [94]. IFNα is also highly produced in CSF [43, 95, 96, 97], though it is unclear whether higher production in serum or CSF could inform on the central versus systemic source of type I interferon production [42, 43, 96]. As of yet, it is unknown whether targeting this pathway will be beneficial for neuropsychiatric lupus, but preclinical evidence suggests this.

Studying neuropsychiatric symptoms in mouse models of lupus is possible. However, similar to human studies, this aspect has been less investigated than its systemic disease counterpart. A lupus-prone mouse model overexpressing Tlr7 among other genes and exhibiting high peripheral type I interferon signatures [98, 99], also develop strikingly elevated interferon signatures in CNS [100]. Importantly, these mice exhibit anxiety and fatigue similar to clinical symptoms in humans. The response seems spatially restricted in high intensity patches across the entire brain, affecting predominantly microglia but also neurons and oligodendrocytes [100]. This is also seen in a different lupus-prone mouse model, where induced interferon-responses accompanying microglial activation [101] suggest conserved pathology affecting the CNS. Additionally, exogenous administration of IFNα precipitates lupus pathology [102, 103] and neurologic disorder including anxiety, depression, and cognitive impairment.

The mechanisms by which type I interferons cause or aggravate development of neuropsychiatric symptoms in SLE have not been exhaustively investigated. One proposed mechanism is through modulation of neuroactive metabolites. SLE patients with cognitive dysfunction are found to have increased quinolinic acid [104, 105, 106], to kynurenic acid ratios [107]. Quinolinic acid is an NMDAR agonist causing glutamatergic excitotoxicity [108, 109] and more so when favoured over kynurenic acid [110]. This metabolite balance dysregulation concurrently correlates significantly with type I interferon-response [111]. Quinolinic acid is metabolised from tryptophan and through indoleamine 2,3-dioxygenase (IDO) [112], an interferon-inducible enzyme known to mediate neurobehavioural alterations [113]. In SLE patients, IDO is induced in a type I interferon-specific manner [114]. This is paralleled by an increase in kynureine to tryptophan ratio in the circulation as IDO metabolises tryptophan into kynureine [107, 111, 114]. Loss of serotonin is also reasoned to be affected in a similar way. Serotonin is an important neurotransmitter that is also metabolised from tryptophan [115] and which is deregulated in human SLE [114, 116] and in the hippocampus of lupus-prone mice [117]. Increased IDO in SLE can explain the reduction of serotonin observed in the periphery [114], and it is arguable that a similar mechanism may take place in the CNS though this has yet to be demonstrated in SLE.

Overall, while a lot of what is known about the pathological mechanisms of neuropsychiatric SLE are inspired from findings from AGS, clinical evidence is highly suggestive that the type I interferon pathway may be therapeutically relevant for the neuropsychiatric manifestations as well as well as for systemic disease. The associations between neuropsychiatric lupus and AGS, and the teachings it has provided, are a testament to how rare monogenic neuroinflammatory disorders can provide aetiological insights into the pathogenesis of more common polygenic disorders. Importantly, more learnings can also be achieved from clinical experience with exogenous IFNα and IFNβ therapies.

3.2 Exogenous interferon treatment

Type I interferons have been used in the clinics to treat different diseases for many years, including hairy cell leukaemia, chronic myeloid leukaemia, hepatitis C virus infection, melanoma, multiple sclerosis, systemic mastocytosis, chronic hepatitis B virus infection renal cell carcinoma, and Kaposi’s sarcoma. Despite being used at refined therapeutic doses, psychocognitive side effects are common and have been well documented from 40 years of clinical experience with these cytokines [118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134]. Common side-effects in the short term include confusion, headaches, fatigue, myalgia, flu-like symptoms, and psycho-emotional disturbance. Longer treatment regimens often result in dementia-like outcomes [135, 136, 137, 138, 139, 140]. Furthermore, high dose versus low dose IFNα therapy much exacerbated signs and incidence rates [141], indicative of a dose-dependent effect.

Depression is the most common occurrence. Incidence among both IFNα- or IFNβ-treated patients is estimated to be 50%, even with optimised dosages, and prophylactic antidepressant therapy is often initiated pre-emptively [123, 128] though its true efficacy is still debated [142, 143]. Major depressive episodes are also commonplace and have been reported in as many as 30% of HCV patients treated [126, 133]. Resting state fMRI performed in a cohort of 22 patients with hepatitis C infection pre- and post-peripheral therapeutic dosing of IFNα revealed rapid and profound changes in the brain [144]. The altered brain functional network and reduced global connectivity efficiency correlated well with changes in anxiety, fatigue, confusion, and mood, reinforcing the immediateness of changes to neuronal networks. Importantly, while the majority of cases of IFNα-induced depression will achieve remission following discontinuation or end of therapy, this can take up to 3 years [125, 130, 133] indicating long-lasting neural changes. These figures may be underestimated due to the lack of long-term follow-up after treatment cessation [130, 133].

Assessing the adverse impact of IFNβ therapy on multiple sclerosis (MS) has been challenging, as MS itself leads to cognitive dysfunction [145]. Cognitive dysfunction in MS also correlates with depression and fatigue [146, 147], adding a further layer of complexity. In comparison, fingolimod is a superior therapy for MS, providing better outcomes for cognitive decline compared to IFNβ [148, 149, 150, 151, 152, 153]. Fingolimod causes immunosuppression as well as neuroprotection [154], an advantage over IFNβ which, specifically in MS, only exerts immunosuppression. It is therefore challenging to separate any neurotoxic effects associated with IFNβ. As new therapies are slowly making their way to more common clinical use in MS [155], more careful personalised medicine approaches can be achieved reducing long-term sequelae for patients.

The impact of type I interferons on psychocognitive states involves a complex, understudied, and multifactorial process involving changes in neuroactive metabolite balance, potentially hormonal dysfunction, brain microvascular dysfunction, and induction of psychoactive chemokines. As previously discussed, they potently induce IDO, which catalyses a key metabolic reaction that leads to the loss of serotonin and kynurenic acid, favouring the excitotoxic quinolinic acid [107, 113, 156]. Interferon therapies have also been found to induce thyroid dysfunction, ranging the entire spectrum of hypothyroidism, hyperthyroidism, and thyroiditis. Clinical thyroid disorders are frequently associated with psychiatric symptomatology [157, 158, 159, 160]. The exact prevalence is difficult to determine as few studies evaluate thyroid status, but it may be as low as 11% and high as 45% [161, 162, 163, 164, 165]. While thyroid hormones are known to exert biochemical effects on the brain, the bulk of the research conducted is descriptive rather than mechanistic. No significant correlation could be easily discerned between thyroid hormones and development of major depression induced by IFNα [161], perhaps suggesting parallel pathways. Nevertheless, given the systematic thyroid hormone imbalance induction by IFNα therapy, perhaps some degree of mood dysfunction may be attributable to it, though research is for now lacking.

Interferons can also directly act on the brain. In mice, it was found that peripherally administered IFNβ was sufficient to induce depressive symptoms [166], and that IFNAR activation on brain endothelial cells was responsible for the depressive symptoms. Activation of the BBB’s endothelial cell IFNAR pathway results in downregulation of adherens and tight junction transcripts causing endothelial dysfunction and exemplified by BBB leakage. Evidence of this is also perhaps apparent from the clinics. Thrombotic microangiopathy (TMA) is another serious side effect of interferon therapies [167, 168, 169] requiring immediate discontinuation and critical care. These events are associated with higher dose interferon therapy, and to result in leaky microvasculature accompanied by perivascular immune cell infiltration and narrowing of the endothelial cell lumen in human patients and in mice [168]. Chemokines CCL2 and CXCL10 are strongly induced by interferons in brain endothelial cells [166], providing a plausible explanation for the microvascular changes and infiltration. Of note, it is the microangiopathy that is the dominant brain characteristic, as also observed in neuropsychiatric lupus [170], and thrombosis is instead a far less prominent feature [168]. As not all microvessel damage is visible by MRI, it is unclear whether subclinical cerebral vascular damage is happening throughout all interferon-treated patients, thus widely contributing to the psychocognitive dysfunction induced by type I interferons. Lastly, activation of CXCR3 signalling in neurons by brain endothelial cell-derived CXCL10 elicits changes in synaptic plasticity responsible for the depressive phenotype in vivo and in vitro causing weakened synaptic long-term potentiation in hippocampus [166, 171].

Genetic associations between type I interferon-induced depression and gene variants exist, but knowledge is for now limited. One study linked polymorphisms in cyclooxygenase 2 (COX2) or phospholipase A2 (PLA2) with a more than three-fold increased risk of developing depression [172]. Intriguingly, an association in patients with major depression not induced by interferon therapy could also be made, suggesting some similar underlying mechanisms [172]. In line with this observation, IFNα-induced depressive symptoms could be mitigated in mice treated with non-steroidal anti-inflammatory drugs (NSAIDs) inhibiting COX1/COX2 [173]. This is an important consideration in light of reported anti-depressive effects of NSAIDs [174, 175] and of enhanced efficacy when used in combination with anti-depressants [176].

Interestingly, type I interferon signatures can also be found in major depression not induced by interferon therapy [177, 178]. Prototypical ISGs such as MXs, OASs, IFITs, ADAR, and CXCL10 [179] are found upregulated in the circulation of patients. It is unclear whether type I interferons might be causative; however, this would argue for a detrimental role in major depression. Depression is also suggested as a trigger for Alzheimer’s disease and cognitive decline [180], raising important additional implications. In one study, an association was found between the Apolipoprotein E ε4 (APOE4) allele, which confers greater risk of Alzheimer’s, and higher incidence of interferon-induced neuropsychiatric symptoms [181], suggesting a link between type I interferons, depression, and Alzheimer’s disease.

The neurocognitive impact of exogenous therapy with type I interferons is well-recognised, and despite systemic administration routes and refined dosages, patients exhibit often debilitating and dangerous psychocognitive side-effects. While anxiety and depression can be partially mitigated, psychomotor dysfunction, fatigue, and confusion are difficult to alleviate [140]. Over the years, the standard of care for many of these indications is moving away from IFNα an IFNβ therapies, both as first-line therapy and often entirely. The advent of more efficacious, and safer direct-acting and all-oral antivirals [182, 183], checkpoint blockade inhibitors [184], and immunomodulating therapies [155, 185] has allowed a consistent phasing out of interferons in clinical care.

Advertisement

4. Translation to more common neurodegenerative diseases

There is growing interest in understanding neuroinflammation in rare diseases because it represents a promising and translatable therapeutic target, also for more common neurodegenerative disorders. Neuroinflammation is recognised as a key hallmark of diseases such as Alzheimer’s and Parkinson’s disease. While the role of type I interferons in these diseases remains to be fully understood, several lines of evidence suggest their implication in their disease process.

4.1 Alzheimer’s disease

Alzheimer’s disease (AD) is the most common cause of dementia, affecting more than 50 million individuals worldwide. It is characterised by gradual cognitive decline and behavioural changes due to chronic neurodegeneration, and predominant impairment of anterograde episodic memory. Individuals over the age of 65 are most commonly affected, representing 90 to 95% of all AD cases. Overall, age is the strongest risk factor for AD suggesting that the ageing process is strongly implicated [186]. Early-onset AD (EOAD) affects individuals below the age of 65 and is known to be caused by one of a handful mutations. The discovery that certain mutations in the amyloid precursor protein APP gene [187, 188] or the presenilin 1 [189] and 2 [189, 190] (PSEN1 and PSEN2) genes, encoding the enzymes cleaving amyloid peptides, cause EOAD has galvanised the field and concomitantly allowed headway in the understanding of late-onset AD (LOAD). As aggregated misfolded amyloid β-containing extracellular plaques are a major pathologic hallmark of AD, the understanding of the amyloid misfolding and aggregation processes have led to major strides for elucidating the pathology. This is also thanks to the generation of mouse models with abnormalities in APP processing mimicking human EOAD [191]. After decades of therapeutics research, and at the time of writing, two biologics targeting amyloid deposits have been approved for therapy following modest but positive trial outcomes [192, 193]: aducanumab and lecanemab.

Subsequent work has identified and proposed many other components importantly implicated in AD. One such component is the protein tau. Tau, like Aβ, has a propensity to misfold, aggregate, and spread, and mutations in its gene, MAPT, have been identified in familial inherited tauopathies such as frontotemporal lobar degeneration with tau inclusions (FTLD-tau) [194]. Hyperphosphorylation is another key feature of tau and a marker of severity of neuronal pathology [195, 196]. Intracellular tau protein-containing neurofibrillary tangles are found in brain regions related to clinical symptoms and to better correlate with pathology than amyloid burden [197, 198], leading to believe that it may be directly implicated in the pathogenesis [199].

Aside from APP, PSEN1 and PSEN2, and MAPT, APOE is the most important risk factor for AD [200, 201, 202]. Certain alleles substantially increase risk (E4/E4: 12-fold increase) while others confer resistance (E2/E2: 2.5-fold decrease), while others are the common variants (E3/E3: no impact). The impact that APOE has on AD is likely even more intricate. An R136 mutation was identified in an individual carrying an autosomal dominant mutation in PSEN1, but which was protected from developing EOAD [203]. Named after the city in New Zealand where it was discovered, the Christchurch mutation confers resistance to AD by reducing tau pathology [204]. The genetics of EOAD are complex, but they have allowed many advances in our understanding of AD. Genetic research of LOAD also revealed new facets of AD, especially the involvement of the immune system.

Neuroinflammation is increasingly being recognised as an important component in the pathogenesis of AD. Genome-wide association studies performed on LOAD have implicated the immune system. Loci mapped to genes hypothesised to carry out immune, or at least microglial, function-related roles have been proposed such as ABI2, ACE, ADAM10, ADAMTS1, BIN1, CD2AP, CD33, CLU, CR1, HLA-DRB1, HLA-DRB5, IL34, MEF2C, MINK1, MTHFR, PCG2, PILRA, SHARPIN, SPI1, SORL1, TOMM40, and TREM2 [205, 206, 207, 208, 209]. Although more than 84 loci have been identified, they may not represent the full spectrum of risk variants for LOAD. As studies become larger and better powered for discovery of subtle associations, more are likely to be added to this list [209]. TREM2 is an important risk variant [210, 211] with consistently strong association and susceptibility risk, currently estimated to three- to four-fold increase [212]. One heterozygous R47H variant represents the highest risk for AD aside from familial mutations and APOE alleles, and has therefore been studied extensively despite sometimes contradictory results [213, 214].

AD is a multifactorial disease with complex cellular interactions that culminate in neuronal cell death. Numerous mouse models have allowed studying the pathogenesis of AD at the molecular and cellular level. Coupled with the advent of single-cell transcriptomic technologies [215], they have permitted a better understanding of the cellular states during AD and have allowed characterisation of virtually all brain cell types. Across AD models, microglia cell types have been identified and characterised according to their transcriptomes and consistently classified as “homeostatic”, “disease associated microglia” or “DAM”, and “type I interferon-responsive microglia” or “IRM” [216, 217, 218, 219, 220, 221]. A lot of excitement was generated from research performed describing the DAM subsets in mice as they are believed to define a distinctive neurodegeneration microglial cluster [222, 223]. However, a clear and consistent DAM cluster has been difficult to pinpoint in humans [224]. IRM have been described in human AD, and the subset is found to be associated to microglia exhibiting endolysosomal dysfunction, cytoplasmic dsDNA, and activated morphology [224]. This same IRM subset also carries significant differential expression of genes associated with genetic risk for AD, such as APP, APOE, GRN, CD33, and C4A and hence is reasoned to be a putative target for therapeutic intervention [224]. While it is now clear that clues relating to presence of type I interferons are seen throughout human AD [224, 225, 226, 227, 228, 229], its role can only be studied using mouse models.

Similar to human AD, type I interferon signatures can be found across different AD models of either amyloidosis, tauopathy, or combinations of the two [217, 220, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238] and despite differences in affected brain areas, temporal and cognitive pathology evolution, and disease mechanisms, suggesting conserved induction pathways. Importantly, genetic loss of IFNAR or targeting via monoclonal antibody is protective for the overall progression [228, 229, 232]. In an APP/PS1 mouse model, loss of IFNAR signalling leads to significant amelioration of the spatial memory deficits, and inhibition of inflammation and microgliosis markers, while astrocytes exhibit activation potentially as a compensatory mechanism [232]. These observations were accompanied by complete loss of interferon-signalling, as expected, but also of IFNα expression hinting at a potentially self-sustained pathway in AD [232]. Similarly, blockade of IFNAR signalling by delivery of a monoclonal antibody to the cerebral ventricles, thus bypassing the BBB, leads to significant reduction in microglial activation [228]. IFNAR signalling is similarly detrimental in tauopathy models [237]. Loss of IFNAR causes strikingly reduced tau hyperphosphorylation and inflammatory cytokine and chemokine production in vivo and in vitro stimulation of neurons with IFNα or IFNβ exacerbates tau hyperphosphorylation and seeded tau aggregation [237]. Furthermore, tau triggers the generation of interferon-responsive oligodendrocytes in vivo, as evidenced by single cell analyses [239], though their contribution to disease remains to be elucidated. Further evidence that tau drives disease through type I interferons comes from deeper investigations of the protective effects of the Christchurch APOE mutation [240]. Mice carrying tau mutations and the human E3/E3 Christchurch mutation were protected from tau pathology by loss of cGAS-STING-induced type I interferon production. While the complete mechanism is still unclear, microglia carrying the protective mutation had suppressed production interferons [240]. Other mutations commonly found in AD were also linked to type I interferons. Concurrent loss of function of TREM2 in tauopathy and amyloidosis AD-prone mice causes exacerbation of the type I interferon signatures, along with more pronounced tau aggregation, hyperphosphorylation, and neurodegeneration [220]. This is paralleled by findings in individuals carrying TREM2 variants R47H and R62H, which have a strongly increased AD risk, and concurrent enhancement of type I interferon signatures compared to TREM2 common variants [241, 242]. It is still not demonstrated whether these AD associated variants exacerbate pathology through IFNAR signalling, though mouse models of tau pathology with TREM2 R47H-carrying variants recapitulate enhanced IRM [234], and this is through an increased responsiveness to triggers of type I interferon production [241].

The induction of type I interferons in AD is thought to be mediated by an amyloid-facilitated nucleic acid recognition process. Nucleic acids are found on and around amyloid plaques in human and mouse models [228, 243, 244, 245, 246], which is probably linked to charge complementarity. Microglia, due to their phagocytic activity, actively take up oligomeric amyloids containing nucleic acids, which then signal to nucleic acid sensors leading to production of type I interferons [228]. Neurons are also capable of producing IFNα and IFNβ in response to Aβ peptide stimulation directly and without exogenous nucleic acids, and to do so via MyD88 and IRF7 [247], raising the prospect of a parallel pathway of mitochondrial or nuclear nucleic acid release following cell damage. cGAS-STING is also found to be expressed and induced in AD-prone mice across different neuronal cell types and is likely a key nucleic acid sensor mediating the production of type I interferons in response to amyloids [248, 249] but possibly not the only one.

Clearance of nucleic acids, whether following uptake or whether released subsequent to nuclear or mitochondrial damage, is an important process. Genetic loss of these mechanisms can result in severe diseases, including type I interferonopathies. Phospholipase D3 (PLD3) is an exonuclease that degrades mitochondrial DNA limiting exaggerated TLR9 [250, 251] and cGAS-STING [251, 252] responses which result in the induction of type I interferon production. Importantly, rare variants in PLD3 have been discovered to increase risk of AD [253, 254], and potentially in EOAD [255] suggesting that defective nucleic acid nuclease activity could be implicated in AD and cause aberrant triggering of type I interferons. PLD3 is indeed found deregulated in AD [256, 257] and accumulated in neuritic plaques [257], suggesting a defective intracellular exonuclease function. Another protein involved in clearance of nucleic acids is 2’,5’-oligoadenylate synthetase 1 (OAS1). OAS1 is a type I interferon-inducible protein which, through its interaction with RNaseL, is known to degrade dsRNA and limit viral infection but to also limit sensing of intracellular RNAs [258, 259]. While the role of this interferon-inducible protein in the pathogenesis of AD still not known, risk variants have been identified suggesting that it may be implicated in the disease process [260, 261].

As for how type I interferons mediate neurodegeneration in AD, there are multiple mechanisms demonstrated so far. For one, Aβ stimulated IFNAR deficient glia produce significantly less or no inflammatory cytokines, including IFNα and IFNβ, compared to IFNAR sufficient glia, and transfer of conditioned media to neuronal cultures demonstrates a neurotoxic activity that requires IFNAR signalling [232, 262]. One way by which IFNβ has been proposed to be directly neurotoxic [263, 264] is through mitochondrial destabilisation [265]. Furthermore, IFNβ induced by nucleic acid recognition also cause further upregulation and hyperactivity of DNA sensors in microglia [266], suggestive of a self-propelled activation loop. Type I interferons also cause inhibition of Aβ peptide phagocytosis by microglia, suggesting that decreased clearance of reactive amyloid species could also exacerbate neuroinflammatory outcomes [262]. In vivo, IFNAR signalling activation leads to complement-driven elimination of synapses [228, 229], an effect that seems entirely driven by microglia, and likely mostly ones near plaques and co-expressing CLEC7 and AXL [229].

Ageing is the most important risk factor for AD. Type I interferons have been associated with brain ageing [267, 268] and are therefore thought to also impact AD through processes in common with ageing. One identified process is through inhibition of the transcription factor Myocyte-specific enhancer factor 2C (MEF2C) [233, 249, 268]. MEF2C is involved in neuronal signalling, differentiation, and integrating memory formation [269, 270, 271]. Moreover, polymorphisms of the MEF2C locus have been identified in AD repeatedly across studies and cohorts [205, 272, 273, 274]. Type I interferons cause an inhibition of MEF2C during normal ageing which results in deteriorating cognitive function including learning and spatial memory and exacerbated neuroinflammation [268]. In AD-prone mouse models, both amyloid- [233] and tauopathy-based [249], type I interferons caused the downregulation of MEF2C leading to increased microglial activation, increased synaptic loss, and cognitive dysfunction.

However, the detrimental effects of IFNAR signalling in AD are likely not mediated solely by microglia. Neuronal, but not microglial, IFNAR signalling contributes to amyloid plaque formation in AD-prone mice, a mechanism suggested to be mediated by the interferon-induced transmembrane protein 3 (IFITM3) [229]. IFITM3 is an interferon-inducible protein that binds to the γ-secretase complex responsible for Aβ cleavage from APP and enhances its activity [275]. It is overexpressed in AD as well as mouse models, in line with the overexpression of other type I interferon stimulated genes, and loss of function leads to striking reduction in plaque density [275] supportive of the observation that IFNAR signalling in neurons participates in amyloid plaque deposition [229]. Certain variants of IFITM3 are also significantly associated with cognitive decline, amyloid and tau burden and brain atrophy [276], though it remains to be demonstrated how these variants affect the amyloid processing-associated function of IFITM3. Neurons also respond directly Aβ by producing IFNα and IFNβ through MyD88-IRF7, a pathway which sensitised to concurrent neurotoxicity [247].

Evidence points to a predominantly detrimental role of type I interferons in BBB integrity. Interferon signatures in brain endothelial cells are elevated in AD-prone mice [277]. IFNβ directly increases endothelial cell permeability to large molecules, downregulating intercellular cadherins [277] and suggesting a direct consequence on BBB leakiness. Evidence is lacking in vivo, however, and it remains to be fully elucidated whether this can be replicated in endothelial cell targeted IFNAR-deficient AD-prone animals. This is especially important as there is contrasting evidence indicating that systemic IFNβ therapy in the context of MS participates in restoring BBB integrity [278]. It is unclear whether this is an indirect effect on the BBB via immunoregulation or a direct effect on endothelial cells. Different models of cerebrovascular damage [279, 280] and infection [281, 282], both of which trigger local type I interferon production by endothelial cells via cGAS-STING, demonstrate a detrimental role on BBB permeability. Thus, elucidating the role of type I interferon signalling according to the disease context and elucidating whether local and systemic effects may have opposing functions is important in understanding AD-associated BBB.

Type I interferon-induced CXCL10 has also been implicated in dementia associated with TAR DNA binding protein-43 (TDP-43) pathology. TDP-43 is an RNA interacting protein whose function is regulation of splicing, trafficking, and stabilisation of RNA [283]. Its normal function is lost in about 50% of frontotemporal dementia (FTD), a progressive neurodegenerative disease characterised by neuronal intranuclear and cytoplasmic inclusions [284, 285]. Loss of the normal function of TDP-43 triggers the production of type I interferons via dysregulation of normal RNA sequestration causing accumulation of dsRNA leading to activation of RIG-I [286] and by destabilisation of mitochondria causing mtDNA release and triggering of cGAS-STING [287]. This results in neurodegeneration in vitro and in vivo [286, 287, 288], and one proposed mechanism is through type I interferon-induction of CXCL10 and signalling to hippocampal presynaptic terminals overexpressing CXCR3 [288]. This triggers sustained neuronal hyperactivity and memory deficits in vivo [288]. It is not yet known what exactly causes upregulation of CXCR3 in neurons in the context of TDP-43 pathology, but clues of a TDP-43-type I interferon-CXCL10 pathway can be seen in human [287, 289, 290, 291]. It is noteworthy that TDP-43 pathology is also observed in about one-third of AD cases [292, 293] suggesting overlapping mechanisms with AD potentially via this identified axis.

Another CXCL10-driven mechanism involves CD8 T-cell infiltration [294, 295, 296]. In recent years, there has been new appreciation for the role of T cells in AD [297, 298]. Evidence now suggests that a CXCR3-CXCL10 axis exists for the recruitment of CD8 T cells, exacerbating neuronal damage and possibly also directly contribute to interferon signalling enhancement, and that this activity converges through the IFNAR1 [296]. The exact role of CD8 T cells in vivo in AD remains for now still elusive [296, 299], yet interferon-responsive CD8 T cells seem to be a common feature among amyloid [296] and tauopathy [300] models of AD. While microglia far outnumber CD8 T-cells in the brain, both in human AD and models, more work will be needed to discriminate between the type I interferon-driven recruitment and activation of CD8 T cells through CXCR3-CXCL10 mechanisms, and the type II interferon-mediated recruitment self-enhancement.

4.2 Down’s syndrome

Down’s syndrome (DS) occurs when an individual has an extra copy of chromosome 21, a phenomenon called trisomy 21. It is characterised by intellectual disability and developmental delay, and a striking incidence of early-onset AD (EOAD). The notable increase in life expectancy of individuals with DS has revealed dementia and other co-occurring neurological and immunological conditions. By age 40, half of the individuals develop AD, this increases to 77% by age 60, and virtually all develop AD by age 70 [301, 302]. Individuals also display important immune dysregulation [303, 304], with notable neuroinflammation [305] and resembling an interferonopathy [306, 307]. A higher prevalence of depression is also seen in DS, and this is associated more strongly with dementia [301, 302]. In addition, basal ganglia calcification is also found in 10–45% of individuals, [308, 309, 310, 311, 312, 313, 314] an observation that is not fully understood but thought to be attributed to an accelerated ageing process. While it was expected from a clinical perspective that DS ageing is accelerated, as evidenced by accelerated hair greying, decreased skin flexibility, and premature death among others, epigenetic age of both brain tissue and blood is confirmed from a molecular marker standpoint [315]. The epigenetic clock used to objectively assess ageing is based on a quantitative assessment of DNA methylation [316]. As a measure of ageing, it stands out among existing epigenetic clocks also because of its impressive predictive ability for time-to-death which further validates the approach [317, 318]. The ageing process is significantly accelerated circulating cells in DS, a process called immunosenescence [319], and this is further accentuated in brain [315].

Chromosome 21 carries ca. 200 genes, triplication of which causes DS and accompanying disease. Deconvolution of the specific role of each gene in DS is difficult. Triplication of APP gene is attributed as the main reason for EOAD, including accompanying senile plaque deposition paralleling genetic AD [320]. The IFNAR1 and IFNAR2 genes which form the two active chains of the functional IFNAR are also found on chromosome 21. Complicating interpretation, however, the interferon gamma receptor 2 (IFNGR2) chain and the interleukin 10 receptor beta subunit (IL10RB) genes are also found on chromosome 21, one of the two heterodimers of the type II and type III receptors, respectively, and whose signalling overlaps substantially with type I interferon signalling. Nevertheless, much research has been carried out on the role of interferons in DS.

Firstly, cells isolated from DS individuals are more reactive to exogenous IFNα or IFNβ treatment demonstrating that the addition of an additional fully functional set of IFNAR receptors leads to an expected increased signalling [304, 306, 321, 322]. Remarkably, signalling downstream of IFNAR is not overcome by negative regulation, and no desensitisation is apparent [304]. However, it is argued that inappropriate and mistimed responses are likely to occur [322]. Importantly, IFNAR triplication also has consequences in vivo in patients. DS individuals display significant correlation between IFNAR1 and inflammation markers including CRP and inflammatory gene signatures [323] as well as of the interferon-response signatures [307, 323]. Furthermore, systemic interferon signatures correlate with cardiovascular severity and depression [307] suggesting that increased interferon signalling associates with a more severe phenotype. While the increased interferon-response and dependence on JAK/STAT signalling seems obvious considering what precedes, the mechanism responsible for triggering of increased type I interferon production remains elusive. One hypothesis is that the aneuploidy state itself fundamentally triggers type I interferons via cGAS-STING signalling due to accumulation of dsDNA in the cytosol [324]. This provides a rationale for the mechanism of triggering type I interferon production in DS, though this remains to be specifically demonstrated.

To investigate mechanistically the role of type I interferons in DS much work has been performed using engineered mice that mimic the trisomy of chromosome 21. Early experiments showed overall amelioration of the phenotype by neutralisation of a cocktail of anti-interferon antibodies with DS mimicking mice available at the time [325]. Currently, numerous DS models exist, but the Dp16 model is often deemed superior. It is trisomic for the entire mouse chromosome 16 region, which includes approximately 113 orthologs found in human chromosome 21, and importantly excludes any orthologs of genes not found on human chromosome 21 [326]. While the same difficulty in distinguishing the contribution of each of type I, II, and III interferons exists in this DS mouse model as it does in human, it is found that the IFNAR receptor is overexpressed among all three interferon receptors and widely expressed in immune cells [323], mimicking the human condition [304]. Importantly, loss of the interferon receptor locus leads to spatial memory rescue and accompanying normalisation of synaptogenesis and dopamine receptor signalling [323].

Taken together, this research suggests a mild form of interferonopathy in DS [306]. It is important to better understand the clinical evolution of disease and define the pathogenic mechanisms to better guide clinical decisions. The predictable chronological sequence of DS allows for the rigorous investigation of the events preceding amyloid deposition and eventual neurodegeneration, making DS an important condition to further elucidate neurodegenerative diseases such as AD.

4.3 Inflammaging

Cell senescence is perhaps the most evident hallmark of ageing. Over the past 10 years, chronic low-grade inflammation has become recognised as an important addition to the hallmarks of ageing in a process dubbed “inflammaging” [327, 328]. Brain ageing, however, remains a challenging field and most research is performed using animal models.

To elucidate the molecular culprits of brain ageing, Michal Schwartz and colleagues performed transcriptomics profiling of various organs from aged mice and found selective upregulation of type I interferon signalling in the choroid plexus [267]. The choroid plexus is a critical structure in the brain that serves as a hub for neurovascular communication and as principal producer of CSF [329]. It would follow that consequences in the choroid plexus may affect wider brain activity. What the group found is that, not only are type I interferons overexpressed, they are also responsible for negatively affecting cognitive function and hippocampal neurogenesis during ageing. This was confirmed by other groups that found IFNα levels in CSF to significantly positively correlate with ageing-induced memory deficits in mice [231]. Blocking IFNAR signalling by delivering function-neutralising antibodies into the CSF via intracerebroventricular administration reduces interferon signalling in the choroid plexus. This leads to the restoration of brain-derived neurotrophic factor (BDNF) and insulin-like growth factor (IGF1). It also inhibits microgliosis and astrocytosis in the hippocampus, demonstrating the extensive harmful effects of ageing-induced type I interferons in the brain [267]. Importantly, type I interferons and interferon responses are also seen in human, in elderly individuals [267], but more research is needed to characterise the ageing responses in the choroid plexus and the consequences for neurocognitive and memory processes. Other lines of evidence suggest that interferon signalling in the choroid plexus has implications for neurodegeneration. During the COVID-19 pandemic it was noted that patients that exhibited severe respiratory symptoms also developed neuropsychiatric [330], neurocognitive [331, 332], and fatigue [333] symptoms at a significantly higher rate. It is suggested that cognitive dysfunction caused by COVID-19 may also be linked to aberrant type I interferon production in the choroid plexus [334] as evidenced by chronic interferon-responses during severe COVID-19 [335]. AD patients also exhibit ageing-related inflammation in the choroid plexus [336], which is also paralleled in a mouse model of AD [231], though more work is needed to define whether type I interferons are involved in ageing-associated AD changes in the choroid plexus.

Exactly how type I interferons participate in the aberrant process of inflammaging remains to be fully elucidated. One proposed mechanism is through suppression of anti-oxidative factors. In mice, mitochondrial instability due to old age leads to type I interferon responses via mitochondrial DNA release and activation of cGAS-STING [337, 338, 339, 340]. The induced type I interferons then counteract NRF2, a potent antioxidant, leading to accumulated ROS and oxidative stress across different organs including heart, liver, kidney [339], and lung [341]. Although this mechanism has not been specifically demonstrated in the brain, studies in the retina and CNS indicate a similar ageing-related mechanism induced by interferons that leads to ROS [340]. Furthermore, cGAS-STING-type I interferon signalling also leads to anaemia by increasing inflammatory monocyte expansion and haemophagocytosis [339], something that could contribute to low grade chronic brain hypoxia [342].

Another mechanism proposed is linked to the senescence process. Senescence describes the abnormal state of permanent cell-cycle arrest, typically in response to stress or damage, and which is ultimately responsible for ageing [343]. Senescent cells also exhibit a senescence-associated secretory phenotype (SASP), releasing cytokines, chemokines and other molecules that can lead to tissue degeneration and a decline in organ function. Work performed to discover ageing-associated processes in the brain found that type I interferon signatures are specifically enriched across all neuronal cell types during ageing in mice, marking a unique signature capable of resolving chronological age [344]. This includes neural stem cells, a cell type vital for neuron renewal, maintenance, and repair, which also exhibited type I interferon signatures with its top gene being Ifi27 [344]. Sophisticated work has been performed to understand how a senescent state occurs. Retrotransposable elements (RTEs), remnants from ancient retroviruses that have integrated permanently into the genome, become more active during cell senescence. This intriguing process is also believed to be a strong contributor to somatic retrotransposition in the brain, a process important for neuronal somatic mosaicism [345, 346, 347]. It is found that senescent cells downregulate TREX1, leading to accumulation of RTEs which then triggers cGAS-STING and strong induction of type I interferons [348, 349]. This explains the interferon signatures found in senescent cells and during ageing, and is therefore a great marker, but evidence suggests that they are also responsible for triggering senescence and ageing [268] through cGAS-STING [350]. Type I interferons are notorious for promoting cell cycle arrest and inhibit proliferation [258, 351], an antiviral tactic meant to slow down viral spread. They are also potent inducers of chemokines and other cytokines. Hence, it stands to reason that chronic overexposure may also itself participate in the senescence process. Indeed, microglia from DS have been found to enter a senescence programme along with type I interferon overexpression, and remarkably inhibition of this signalling prevents development of senescence [352]. Similar processes have been observed in the senescence of haematopoietic and germinal stem cells [353], as well as hepatic stellate cells [348], indicating a potential universal mechanism. In the brain, a mechanism whereby microglia are responsible for triggering senescence in neurons and other glial cells is now hypothesised to be a major driving force behind cognitive impairment both in ageing, AD, and during brain injury [354, 355, 356]. One proposed mechanism of type I interferon-driven ageing of microglia is through the downregulation of the protective and inflammation-limiting transcription factor MEF2C [268]. MEF2C is found to be implicated in brain development and neuropsychiatric disorders [357, 358]. Chronic production of IFNβ in the brain causes downregulation of MEF2C, as does ageing, and in so doing inhibits a resilience to ageing-induced [268] and disease-induced cognitive decline [249]. Lastly, two recognised pro-ageing SASPs which promote decreased hippocampal neurogenesis, synaptic plasticity, impaired learning and memory and increased microgliosis, are CCL11 and CCL2 [359, 360]. There is evidence demonstrating that type I interferons are inducers of CCL2 and CCL11 [ 166, 171, 361, 362], though it remains to be demonstrated whether type I interferons promote ageing and senescence also through CCL2 and CCL11.

4.4 Brain trauma

Traumatic brain injury (TBI) was once thought to cause static neurological damage. Yet, research now indicates that it can set off a chain reaction leading to ongoing neurodegeneration, widespread damage outside mechanically injured sites such as the hippocampus [363] and the onset of dementia. It is common for individuals to suffer prolonged cognitive deterioration, which can be attributed, to some extent, to the emergence of dementia and AD following concussive injury [364]. Large cohort and meta-analyses have attributed a 1.5-4 fold relative fold risk increase in dementia following TBI depending on number of concussive events, and time following the event [365, 366]. This effect is exacerbated in the elderly, as both functional outcome and survival are significantly worse in elderly individuals, making age one of the more reliable prognostic factor [367, 368, 369]. Older individuals also represent the highest proportion of TBI events [370], adding further credence to the need for enhanced care for the advanced age groups. Progressive neurodegeneration due to TBI may account for 5–15% of all cases [371], suggesting that understanding the pathological mechanism of TBI may lead to better treatments and a staggering reduction of incident neurodegenerative diseases.

Neuroinflammation is known to cause secondary injury progression following TBI, potentially leading to long-term sequelae and progressive neurodegeneration [372, 373]. Much effort has been made to characterise the inflammatory environment of TBI in carefully controlled conditions, and to investigate the contribution of identified pathways to the long-term outcomes. For this purpose, preclinical research in the mouse has been instrumental, providing a broadly homogenous induction and allowing for experimental therapies, a feat that is difficult to achieve in emergency medicine settings involving humans. Multiple lines of evidence have now identified the type I interferon pathway to be strongly implicated, both by spatial [374], single cell [375, 376, 377, 378, 379], microdissected tissue [380], as well as bulk [280, 381, 382] transcriptomic analyses. While expression of interferon-response genes is widespread across microglia, meningeal macrophages, astrocytes, and oligodendrocytes, most evidence points to microglia and macrophages as the main producer of type I interferons following injury. Importantly, activation of the type I interferon pathway is detrimental to the resolution process, as consistently evidenced across genetic ablation models and through intervention by monoclonal antibody treatments [382, 383, 384]. Pharmacologic inhibition or genetic ablation of cGAS-STING also cause abrogation of type I interferon signalling and confers neuroprotection and accelerated neurocognitive amelioration [379, 385, 386, 387, 388] suggesting that aberrant DNA sensing through microglial STING may be a critical trigger for sustained and pathogenic inflammation following TBI. Activation of transposable elements as well as mitochondrial DNA release is found to trigger activation of cGAS-STING in this context [377, 389]. Other sources of nucleic acids, as well as nucleic acid sensors, are also likely to be involved. Neutrophil extracellular traps (NETs) have been found to be important in triggering neuroinflammation in TBI [390] and are triggers for TLR9 and type I interferon production [391].

Consistent evidence of the implication of this pathway is also seen during human TBI, despite high heterogeneity compared to preclinical models. Expression of STING1 is significantly upregulated following trauma, both at the site of injury and at the contralateral side [385] confirming its potential implication in the detrimental neuroinflammatory process. Upregulation of IFNβ, but not IFNα, is also seen within the first 6 h post TBI in patients that succumbed to the trauma [383], though it remains to be determined whether this partiality for IFNβ is maintained during the secondary, chronic neuroinflammation. Furthermore, nucleic triggers for the cGAS-STING pathway and for other nucleic acid sensors are highly enriched following TBI. Cell-free nucleic acids have been found in enormous amounts in both CSF [392] and plasma [393], and correlate with severity of trauma and outcome following injury. The extent to which nucleic acids pour from the brain to the CSF and circulation is a testament to the excessive cell death and accompanying nucleic acid release, and highlights the quantities that must be present in parenchyma during the acute event and therafter. With such excess of type I interferon triggering molecules, it is not surprising that interferon responses are also apparent, evidenced from human single-nuclei transcriptional analyses [377]. Lastly, the more severe outcomes seen in elderly individuals may be paralleled by enhanced persistence of type I interferon signalling and worsened outcomes observed in aged mice [378, 380, 394] and may provide a model for uncovering age-related determinants of interferon-driven chronic neurodegeneration following TBI.

Mediating the pathogenic activities of the cGAS-STING-type I interferon pathway is likely to involve multiple downstream events. Evidence suggests that effector T-cell infiltration via CXCL10 is one important component, whereby infiltrating CXCR3+ Th1 cells cause white matter injury, promoting anxiety and depressive neurocognitive changes [395]. Neuron loss in the hippocampus could be attributed to sustained chronic IFNβ [384], yet exact mechanisms remain to be elucidated. NOX2, a subunit of NADPH important for the production of ROS, is highly neurotoxic during TBI [396, 397, 398] and particularly in the hippocampus. Loss of IFNβ significantly attenuates NOX2 [384], implicating the type I interferon pathway in pathogenic ROS production. Neutrophils, which are important producers of ROS, can in fact be instigated to produce ROS following IFNα stimulation [399]. One possibility is that type I interferons exert control over ROS production via infiltrating neutrophils, which are generally regarded as detrimental in a sterile injury context in the brain [400]. Adding further weight to this hypothesis is the observation that neutrophil infiltration is significantly decreased in microglial STING deficient animals during TBI [387]. However, neutrophil infiltration is thought to be a self-limiting event, which may not carry over during the secondary, chronic neurodegeneration phase. Thus, another possibility is that interferons carry ROS production over through chronic phases via exerted control over IDO [156]. IDO is known to be induced by type I interferons in microglia and to result in production of ROS in response to infection [156, 401]. Thus, in the context of unabated type I interferon signalling, microglial damage of neurons may be mediated by IDO-dependent ROS production.

TBI induces strong immediate damage and a neuroinflammatory response that is detrimental to the long-term recovery via secondary injury processes. Although complex and multifaceted, the neuroinflammatory response is characterised by type I interferon signalling, albeit not exclusively. Yet, blockade of the pathway in preclinical models and the parallels seen in human suggest that targeting this pathway in human may hamper the aberrant neuroinflammatory process and may improve neurocognitive outcomes by reducing neurodegenerative processes.

4.5 HIV/AIDS-associated neurocognitive disorders

Human Immunodeficiency Virus (HIV) is the devastating virus that causes acquired immunodeficiency syndrome (AIDS), and which affects an estimated 39 million people worldwide. HIV uses the CD4 surface protein and either the CXCR4 or CCR5 receptors to gain entry into cells, thereby infecting them, and killing them in the replication process [402, 403]. Over time, this causes systemic immunosuppression as CD4 T cells, dendritic cells, monocytes, and macrophages become depleted. Due to advancements in the standard of care, which relies on antiretroviral therapy (ART), AIDS has become a secondary concern as viral replication is inhibited and infected individuals live longer lives without developing immunosuppression. While viral replication is effectively hampered, infection persists.

HIV/AIDS-associated neurocognitive disorders (HAND) are a common occurrence among people with HIV. Meta-analyses suggest a combined prevalence of 50% among all infected individuals, presenting with at least one neurocognitive sign among all seropositive individuals. Of those, cognitive-motor disorder (60%), major depression (15-40%), and delirium (17%) [404] are some of the neurologic complications of HIV. The prevailing theory is that infected cells serve as a ‘Trojan horse,’ transporting HIV to the brain, and causing infection of microglia [405], and astrocytes by cell-to-cell transfer [406, 407]. Remarkably, while ART can reduce the prevalence of HAND, incidence remains abnormally high and a concern for virtually all HIV patients, a fact probably explained by increased longevity [408, 409]. This suggests that neurocognitive decline in HIV patients despite controlled viral load may have similar mechanisms as age-related cognitive decline.

A complete picture of the pathological mechanism behind neurocognitive disease in HIV is lacking. Its pathogenesis is attributed to persisting viral load in the brain, unabated neuroinflammation, and neuronal loss due to pro-inflammatory cytokines. Microglia and brain infiltrating macrophages serve as the main reservoirs following HIV neuroinvasion, typically within two weeks after infection. Gene expression profiling of brain samples from seropositive individuals that had neurocognitive impairment reveals strong type I interferon-response genes such as OASs, IFITs, IFITMs, and CXCL10 across multiple studies [410, 411, 412, 413, 414]. While this is not surprising for a viral infection, uncontrolled inflammation can become detrimental causing neuroinflammation and neurocognitive impairments [415]. This is exemplified in mice overexpressing IFNα in the brain, which are both protected from viral encephalitis, but also develop progressive neurodegeneration as a consequence of persistent neuroinflammation [416]. Importantly, IFNAR signalling is required for neurocognitive decline and neuroinflammation due to HIV, in vivo [417, 418]. Recognition is thought to occur via stimulation of nucleic acid sensors TLR7 and TLR9 [419], IFI16 and STING [420], RIG-I [421], all converging on type I interferon production. This is paralleled in HIV infected individuals as IFNα is detectable in CSF [422] and correlating with viral load [423, 424] as well as NfL [425] providing further credence to the neurotoxic effect of sustained type I interferon production in brain following HIV infection.

Evidence also exists that while ART may inhibit viral replication, neuroinflammation remains unabated. In a brain organoid model, HIV infection leads to upregulation of type I interferon, interferon-signatures, and of other inflammatory mediators in microglia, the main infected cell in brain, causing elevated inflammatory outcomes also in non-microglial cells [426]. This further reflects the extent of damaging responses to HIV in cells not directly infected. Importantly, while ART causes ablation of the expression of HIV proteins, microglia become a persistent reservoir of HIV [405] exemplified by continuous production of interferon-response CXCL10 and chemokine CCL2 despite ART [426] suggesting that intracellular viral recognition and response sustain a replication-independent inflammatory response. Consolidating the observation that ART does not abolish HAND incidence is the fact that, while it suppresses viral replication, it does not affect latent virus [427]. This is reflected by the persistence of interferon-response gene expression in patients undergoing ART [413]. Hence, it is reasoned that the microglial viral reservoir alone may be sufficient to perpetuate neuroinflammation and HAND, even at low or undetectable viraemic loads. It is important to note that not all ART display the same CNS penetrance. The structure of the BBB coupled with organised efflux mechanisms block or severely limit the access of ART to this reservoir and argues for the development of new generation ART with improved brain penetrance [428, 429].

Beyond HIV, there may be parallels to other viral infections. Cognitive deficits have been recorded in patients recovering from COVID-19 following SARS-CoV-2 infection [430, 431, 432, 433]. The virus is known to infect the choroid plexus as viral entry factors including the receptor ACE2 are highly expressed by its epithelial cells [335, 434]. The interferon signatures [335] were found to parallel what is seen in neurodegenerative diseases [216, 217, 218, 219, 220, 221] as well as choroid plexus during ageing [267], lending to the hypothesis of a type I interferon-associated cognitive dysfunction pathway in COVID-19 [334].

Advertisement

5. Mechanisms of neurologic dysfunction

Research on the rare type I interferonopathies, which typically present in infants, has been challenging. Case studies and small patient cohorts account for most of our knowledge of this family of diseases. Understanding the mechanisms by which type I interferon causes neurologic disease can have substantial impact on treatment of interferonopathies. Type I interferons are also implicated in other non-mendelian diseases which are often accompanied by a predominant neurological component. Therefore, a better mechanistic understanding could have important implications for more common neurological diseases. The following section describes some of the better-known effects in the brain, linking together different disorders and emerging concepts, and highlights some of the remaining open questions.

5.1 Local source of interferons in the brain

Type I interferons can be produced by almost every cell in the CNS, despite lacking dedicated – or “professional” – producers of these cytokines. Being most prominently produced in response to nucleic acids, expression patterns of nucleic acid sensors therefore dictate for the most part whether any specific cell type produces interferons. Some, basally express virtually all nucleic acid sensors, such as microglia, whereas others, such as astrocytes, express a more specialised subset of sensors.

Astrocytes have been proposed as a major producer and responder in AGS caused by TREX1 or RNASEH2 mutations [435, 436, 437]. Immunohistochemical staining of postmortem brain sections revealed that astrocytes were the main producers of IFNα and CXCL10, a typically interferon-responsive chemokine. Mutations in the genes associated with AGS cause aberrant accumulation of DNA which becomes the trigger for production in astrocytes [438, 439]. Moreover, chronic exposure to IFNα was found to cause aberrant activation of astrocytes making them reactive, while simultaneously inducing gene expression changes reminiscent of AGS [436].

Microglia are thought of as the main producer in more common neurodegenerative diseases. In Alzheimer’s disease, both Aβ oligomers [232, 247, ] and amyloid-associated nucleic acids [228, 229], as well as soluble tau [237, 239, 240, 440] are capable of triggering production from microglia. This is also the case for HIV-associated neurocognitive disorders, which is less surprising given that microglia are the main CNS reservoir for HIV [405].

Neurons and oligodendrocytes can also produce type I interferons, albeit to a lesser extent than astrocytes and microglia. They have been found to express TLR3 and to be capable of producing IFNβ in response to dsRNAs [441, 442]. Neurons also express cGAS-STING and respond to mtDNA by triggering the production of IFNβ [287]. They are also found to express TLR9 [443, 444], TLR8 [445], TLR7 [446, 447], RIG-I [448], though evidence of their involvement in production of interferons remains for now limited.

Brain vascular endothelial cells form an important direct interface between the brain parenchyma and the circulation. The cGAS-STING pathway plays an important role in vascular endothelial cells, as exemplified by the observation that mutations in the STING1 gene cause overt production of type I interferons leading to vasculopathy [449]. Brain endothelial cells produce type I interferons [282, 450] and they are known to be able to do so via STING [281, 451], via RIG-I [452, 453], or also in response to TNF through IRF1 [454]. While more research is needed on the endothelial cell contribution to type I interferon production specifically in the brain, convincing evidence exists that they are capable of it.

In addition to nucleic acid sensors, surface pattern recognition receptors are also expressed by cells in the brain. It is well-described that TLR2 and TLR4 respond to disparate triggers including viral proteins, protein aggregates, and bacterial lipids initiating production of pro-inflammatory cytokines. This includes type I interferons which are produced through TLR2-MyD88 and TLR4-TRIF [455, 456]. TLR2 and TLR4 are known to be expressed on microglia, astrocytes, and brain endothelial cells, and neurons are described to express TLR4. It is important to note that induction of type I interferons is also mediated through pathways other than nucleic acid sensing during neuroinflammation.

Direct evidence for type I interferon production in vivo is difficult to obtain. High, ubiquitous expression of the IFNAR causes low bioavailability of these cytokines, but also results in interferon-related signatures that are powerful surrogate markers. Despite this, technologies that allow the direct detection of IFNα and IFNβ have enabled the quantification of significant increases in the CSF of HAND, neurolupus, and AGS patients, suggesting high continuous CNS production. Ultimately, which cells in the CNS produce type I interferons is often context and disease dependent. Another important consideration is that some nucleic acid sensors can also be induced by type I interferons themselves, thereby initiating a chain reaction of production which can be self-sustained for as long as stimuli are present. In some cases, production is initiated in the periphery causing multiorgan diseases with sometimes prominent CNS involvement.

5.2 Systemic source of interferon

Interferons produce systemically are also capable of signalling to the brain [457]. In mice, it was demonstrated that delivery of systemic interferons stimulates the induction of interferon-response genes in brain parenchymal cells [458], and that microglia become strongly activated and to upregulate complement alongside classical pro-inflammatory interferon-responses [459].

The exact mechanisms by which peripheral type I interferons signal to the brain are not fully understood. From research on the cross-talk between peripheral inflammation and the CNS, it is reasoned this can happen in any of four different ways [460]: (1) passively across the BBB and through brain regions called circumventricular organs, which are devoid of a BBB and highly permeable to permit rapid communication between CNS and circulation [457, 458], (2) through induction of BBB leakiness via downregulation of adherens and tight junctions [166, 277, 450], (3) by active uptake through the choroid plexus endothelial cells [166, 267], or (4) through cell migration across the BBB, such as activated monocytes attracted to a microglial CCL2 gradient [361] which may carry type I interferon signalling. Though less is known about the latter mechanism, it is thought that this is a primary mechanism contributing to depression [460, 461, 462].

The most obvious example of systemic production of type I interferons is lupus. In SLE, plasmacytoid dendritic cells (pDCs) and monocytes are thought to be primary sources [463, 464, 465, 466, 467] however, despite being a systemic disease, it is difficult to posit that there is no production in the brain.

Type I interferons form an important innate immune defence barrier, and are, as such, capable of being produced by all cell types, depending on the inflammatory context. They can be produced in response to viral or bacterial infections, or in response to damage associated signals, either locally in the brain or systemically, and to freely traverse the BBB. It is probable that interferon production in both the circulation and the CNS concomitantly contribute to neuroinflammation, even when the source appears predominantly either systemic or central.

5.3 Cellular responses to interferon in the brain

While sources of interferon can be various, all type I interferon signalling converges through a single surface receptor, the IFNAR. This ubiquitously expressed dimeric receptor, formed of an IFNAR1 and an IFNAR2 chain, allows for a wide range of cellular responses, affecting various cell types. Microglia, astrocytes, neurons, and endothelial cells are known to respond strongly and in specific ways.

Microglia assume a hyper-ramified morphology [468], a typical change signifying activation and immune surveillance mechanisms. Microgliosis is strongly induced by type I interferons, and across neuroinflammation models, whether AD, AGS, or lupus, it is dependent on IFNAR signalling. Concurrently, increased processes and complexity are also induced, which typically signify increased synaptic pruning. Short-lived proliferation and apoptosis following long-term stimulation are also observed. Lastly, their antigen presentation capacities are enhanced by upregulation of MHC I and II genes and of activation markers CD68, CD40, CD80, and CD86 [228, 459, 468, 469, 470]. This is further exemplified in single cell studies across human AD and mouse models, where interferon-response signatures overlap with antigen processing and presentation genes.

Astrocytes also respond to type I interferons, though their interferon-response gene expression is less pronounced than that of microglia [469]. It also seems that, in AD models, astrocytosis is not dependent on interferons. This is in contrast to AGS, where astrocytes are thought of as the primary involved cell type [436]. Intriguingly, like microglia, they also upregulate genes related to the antigen presentation machinery, though they are not classically thought of as APCs. Yet, reactive astrocytes display antigen presentation characteristics, as well as astrocytic toxicity markers.

Endothelial cells of the brain vasculature are major responders to type I interferons. They respond by producing chemokines, allowing cell infiltration, and succumbing to apoptosis leading to vascular dysfunction. An AGS mouse model driven by astrocyte promoter-dependent production of IFNα recapitulates microangiopathy, perivascular T-cell infiltration, perivascular calcification, and capillary calibre and formation of aneurysms seen in human patients [44]. Importantly, endothelial cell-specific ablation of IFNAR causes near-complete rescue of cerebral vascular disease [44], suggesting that endothelial cells are the principal responders in AGS and a major target for type I interferon-driven neuroinflammation. Endothelial cells exposed to type I interferons display reduced mobility and invasion [471], in line with their cancer inhibiting properties [472]. In fact, IFNAR signalling inhibits vascular endothelial cell growth factor (VEGF)-induced proliferation [452] highlighting the anti-angiogenic potency of these cytokines. It was shown that CXCL10 can inhibit endothelial cell proliferation in a CXCR3-dependent [473] and -independent [474] pathway. Interferon-inducible IFI35 is also described as an important inhibitor of endothelial cell proliferation and migration [475], indicating that multiple parallel pathways exist. Further accentuating the importance of type I interferons in endothelial cell biology is the surprising discovery of basal interferon-response expressing endothelial cells across different organs including the brain [476] suggesting a regulatory and homeostatic role of tonic type I interferon signalling, though research is needed to characterise these cells and their roles.

5.4 Immune cell infiltration

Certain chemokines such as CXCL9, CXCL10, and CXCL11 are known to be preferentially interferon-inducible, while others, such as CCL2, CCL5, and CCL20, are also induced by interferons but not exclusively. While microglia also display important chemokine production in response to interferons, vascular endothelial cells are particularly suited to the task by virtue of their position at the interface between tissue and circulation. Chemokine gradients and presentation at the lumen side allows for efficient recruitment of immune cells expressing cognate chemokine receptors. In brain endothelial cells, IFNβ treatment causes upregulation and production of CXCL9, -10, -11, CCL2, and -5 which are also able to signal in the brain parenchyma [166]. They bind CXCR3, CCR2, and CCR5 which are predominantly expressed on T-cells, NK cells, monocytes, and dendritic cells, among others.

It has been reported that IFNβ can also inhibit monocyte infiltration by downregulation of adhesion molecules during experimental autoimmune encephalomyelitis (EAE) [477], a model of MS, supporting the anti-inflammatory characteristics observed in MS. It is unclear whether this is disease- or tissue-context specific, as subcutaneous administration of IFNβ in MS patients causes upregulation of the aforementioned chemokines accompanied by extensive T-cell and macrophage perivascular infiltrates [478]. IFNα overexpressing mice show extensive perivascular infiltrates in the brain [416, 479] which depend specifically on endothelial cell IFNAR signalling [44]. Despite having elevated interferon-responses, ADAR1 mutation-carrying mice do not, display perivascular infiltrates [480], while a different nuclease deficiency, RNASET2, leads to IFNAR-driven T-cell and monocyte infiltration [481]. In the context of brain injury, IFNAR-signalling enhances T-cell and monocyte recruitment [382]. This suggests that type I interferon-driven immune cell infiltration is context dependent and requires more research to fully elucidate the underlying differences that set the final outcomes apart.

5.5 Vascular dysfunction

As a consequence of inhibition of normal endothelial cell proliferation and function, it is not surprising that type I interferons can also cause pathogenic vascular dysfunction. IFNAR signalling in endothelial cells specifically causes development of BBB disruption, microangiopathy, calcification, neuron loss and premature death in an AGS mouse model [45]. There is also clinical evidence supporting this observation. Exogenous interferon therapy has been associated with thrombotic microangiopathy in a dose dependent manner [168, 482]. Type I interferons cause abnormal brain vascular morphology, displaying smaller size microvasculature, sections of widened vessel formation, and microaneurysms. Specifically, vascular lumen narrowing is seen diffusely across the brain [168]. The interferon-inducible IFITM1 protein has been discovered to be implicated in the formation of stable vascular lumen during angiogenesis through stabilisation of endothelial cell-to-cell interactions [483]. While loss of IFITM1 leads to abnormal vessel formation, it is for now unknown whether upregulation can also disturb endothelial cell-to-cell contacts resulting in inappropriate vascular lumen formation and dysfunction.

Certain contexts, such as viral infection, can greatly inhibit normal function and healing until viral stimuli are cleared. In endothelial cells, it is found that viral sensing through MDA-5 triggers IFNβ production. This thereby blocks vascular repair, angiogenesis, and BBB restoration following injury, causing a failure to recover normal neurological function [280, 484]. It is likely that even milder neurovascular damage, in the presence of overactive interferon signalling, can cause similar outcomes. In a mouse model of AD, IFNβ correlates with BBB disruption and contributes to it by downregulating adherens junctions and tight junctions causing leakiness [277]. Both IFNβ and IFNAR1 expression is markedly increased in vascular endothelial cells in vivo compared to non-AD-prone mice, indicating an induced hyper-sensitivity to type I interferons which is likely caused in some form by amyloids. While Aβ can trigger microglia to produce type I interferons by multiple mechanisms, it is unknown whether similar mechanisms take place in endothelial cells. Importantly, upregulation of IFNAR can be a key sensitisation step that needs further research. Cerebral amyloid angiopathy (CAA) is a condition where amyloids deposit along the walls of cerebral blood vessels [485]. This causes microhaemorrhaging which further precipitates cognitive decline [486, 487]. Furthermore, current generation anti-amyloid therapies are known to cause amyloid-related imaging abnormalities (ARIA), essentially aggravated haemorrhaging induced by the amyloid targeting therapies. It is for now unknown whether there is a direct link between CAA and type I interferon signalling, and whether they participate in the induction of ARIA.

Following BBB disruption, leakage of blood products containing DAMPs of various sources gain access to the brain parenchyma and trigger damage sensors. One such activator is fibrin, a powerful trigger of CD11b/CD18 surface heterodimers expressed on microglia and which is gaining emerging interest as the full extent of its involvement in CNS diseases and neurodegeneration [488]. During AD progression, mice lacking fibrinogen, a plasma protein required to create fibrin, show reduced gliosis, neuronal damage and cognitive decline and suggest that vascular damage synergises with amyloid pathology [489]. The type I interferon pathway is found to be strongly triggered by fibrin [490] indicating that a leaky BBB can also directly trigger this neuroinflammatory pathway and suggesting a self-sustained feedforward mechanism of interferon-BBB leakage-interferon. The exact trigger for type I interferon production, whether indirect or direct, remains to be elucidated – whether indirectly through induction of DNA release following mitochondrial or cell damage, or directly through a potentially novel pathway. Yet, if not properly regulated, this is likely a mechanism through which neurodegeneration-inducing neuroinflammatory events can propagate throughout the brain.

As for how exactly type I interferons cause calcifying microangiopathy, this is still an open question. One hypothesis is that subclinical disruption of microvessels may be sufficient to cause continuous deposition of calcium along vessel walls. Evidence of direct induction of calcification also exists in vitro, as IFNα precipitates calcification at concentrations in the range found in CSF patients [491]. Finally, it is thought that cell senescence is linked to calcification, but it is unclear which one causes the other and how [41]. While type I interferons are known to cause both calcification and cell senescence, it remains to be discovered how this mechanism is mediated and whether through senescence.

5.6 Phagocytosis and synaptic pruning

Overall, type I interferons have been found to both inhibit appropriate microglial removal of aggregated proteins and debris, but also to enhance synaptic engulfment and neuronal elimination.

While in the context of AD it is still debated whether amyloid plaque formation is truly detrimental by enhancing neuronal network disruption or whether it renders reactive amyloid peptides unreactive in extracellular clumps, amyloid plaque deposition is generally accepted as a sign of exhausted phagocytosis. In vitro, both IFNα and IFNβ significantly reduce phagocytosis in a dose-dependent manner [262]. Furthermore, IFNAR signalling interferes with normal Aβ phagocytosis. This coincides with an increased pro-inflammatory state including increased cytokine production. This suggests that type I interferon drives mitochondria away from a phagocytic clean-up function towards a hyper-reactive pro-inflammatory state.

In vivo, there are some contradictory results, however [229, 262]. In APP/PS1 mice, plaque deposition seems grossly unaltered [262] whereas a different group could demonstrate decreased plaques [229]. Importantly, this was observed in mice lacking IFNAR specifically in non-microglial cells, but not in mice lacking microglial IFNAR. This seems to suggest that the phagocytic activity against plaques in microglia may be driven by unalterable genetic drivers which can, at best, be modulated by changes in neuronal interferon-inducible IFITM3. Likely, these results do not rule out clearance of reactive oligomeric amyloids, which may act by decreasing the overall inflammatory state. These results cannot rule out a possible effect in human LOAD where genetic drivers for amyloid deposition should be other than aberrant processing of amyloids.

Complicating matters further is the observation that type I interferons upregulate phagocytic markers [492] and promote synaptic pruning leading to neurodegeneration [228, 229, 233, 249, 352, 493]. Synapses are specialised junctions through which neurons signal to each other. They consist of three parts: the presynaptic terminal of the signalling neuron, the synaptic cleft, and the postsynaptic terminal of the target cell. Synaptophysin is a membrane glycoprotein present in presynaptic vesicles and is involved in the regulation of neurotransmitter release. Postsynaptic density protein 95 (PSD-95) is a scaffolding protein located in the postsynaptic density of excitatory synapses and it plays a critical role in anchoring and clustering neurotransmitter receptors and other signalling complexes at the synaptic membrane. Synaptic pruning mechanisms occur via phagocytic microglia and astrocytes in a complement-dependent mechanism [494, 495]. During synaptic pruning, certain synapses are tagged for removal by complement proteins such as C1q and C3. These proteins bind to the synapses and mark them for elimination. Microglia express complement receptors that recognise these tagged synapses. Once bound, microglia can engulf and digest the synaptic material, effectively pruning the synapse. This mechanism ensures the refinement of neural circuits and is essential for proper brain development and function. Dysregulation of this process leads, however, to neurodegenerative diseases. Type I interferons are found to induce complement-dependent synaptic pruning across multiple neurodegeneration models, but also in normal healthy development [496, 497] suggesting that this is a conserved mechanism and that it requires delicate regulation. Inducing the expression of complement components, such as C1q, C3, and C4, to mark synapses for elimination is perhaps only part of the mechanism, and it is still unclear whether interferons can also enhance the activity of complement receptors binding to complement-opsonized synapses, such as CR3 and CR4, and facilitate their engulfment.

Overall, type I interferons activate microglia to become reactive and pro-inflammatory, and to poise them for complement-driven synaptic pruning and neuronal engulfment, while rendering them unable to clean up reactive aggregated proteins. Whether this is a result of task overload, or a preferential outcome due to a switch in signalling remains to be determined and the underlying mechanisms discovered.

5.7 Mediating neuroinflammation-induced neurodegeneration

TBI is perhaps the most typical display of neuroinflammation features [498, 499, 500] which are associated with development of dementia [501, 502]. The neurodegeneration process persists long after the concussive event where reactive microglia morphology and upregulation of CD68 can be observed years after injury along with white matter atrophy [503]. Single nuclei RNA sequencing reveals an interferon signature in oligodendrocytes [377], the specialised glial cell type forming the myelin sheath typical of the white matter and allowing the efficacious saltatory nerve conduction between neurons.

These observations have been corroborated in bona fide mouse models of TBI which have been used to expand on the mechanistic link between brain injury, type I interferon signalling, reactive microglia, and neurodegeneration. Sustained type I interferon signatures are found strongly upregulated during TBI in both microglia and astrocytes, alongside genes related to glial reactivity [374]. Reactive microglia morphology and CD68 expression were also found to be induced by type I interferons directly in vivo [228]. Importantly, inactivation of IFNAR leads to reduced neuronal loss and protection of white matter resulting in improved neurocognitive function [382, 383]. STING-deficient animals lose most of the type I interferon signatures [385], hence the trigger itself can be speculated to be massive release of nucleic acids from damaged cells. Remarkably, neuronal STING-derived IFNβ alone is sufficient for promoting neuroinflammation [384, 395], in part through a decrease in NOX2, an important inflammatory mediator of post-traumatic neurodegeneration [396, 398]. IFNβ also initiates a CXCL10-driven white matter injury process, through CXCR3-dependent and -independent neurotoxic activities, and through induction of Th1 cell infiltration [395]. Yet, this does not account for the sustained interferon production succeeding acute trauma. Perhaps type I interferons cause a feed-forward loop of aberrant neuronal engulfment as explored in the previous section: nucleic acid sensing, accrued interferon production, more neuronal engulfment – rinse and repeat. While this remains to be demonstrated, an immune cell infiltration feed-forward loop exists which explains the self-sustained pathogenic interferon signatures [362, 378]. Monocytes in the meninges are found to produce CCL2 and to strongly upregulate interferon signatures [378]. That causes a CCR2-dependent recruitment process which is also remarkably required to propagate the type I interferon signatures during TBI [362], thus together suggesting a feed-forward loop of unabated interferon-response and immune cell infiltration.

Type I interferon initiates a potent neuroinflammatory response to brain injury, causing direct neuron and oligodendrocyte damage, and instigating monocyte and T-cell infiltration that potentiates and prolongs detrimental responses. The mechanisms identified validate targeting type I interferon or its downstream effectors as a therapeutic strategy in TBI. Inhibiting type I interferons leads to an ordered conclusion of the self-sustained inflammatory process, thereby promoting favourable neurocognitive outcomes.

Put together, the mechanisms by which type I interferons are known to affect the brain are numerous and act by perturbing multiple key processes required for neuronal function. From promoting neurodegeneration, to causing microangiopathy, to precipitating premature ageing, to promoting depressive states, type I interferons are an important neuroinflammatory target (Figure 3).

Figure 3.

Putative mechanisms of concerted psychocognitive disturbance by pathogenic type I interferon dysregulation. Neurodegeneration in the context of Alzheimer’s disease-related proteinopathies implicates type I interferon at multiple levels. DNA associated with amyloid plaques triggers the production of type I interferons by microglia, which become reactive in response to paracrine IFNAR signalling [228, 229]. Activation of IFNAR signalling in microglia causes upregulation of DNA sensors amplifying the response and promoting self-sustained activation [266]. CXCL10, produced in response to type I interferons, signals to CXCR3-expressing neuronal presynaptic terminals. This causes hyperexcitability and initiates chronic responses, leading to the impairment of new memory formation [286, 288, 504]. Interferon also activates complement driven neuronal synapse pruning by astrocytes contributing to loss of existing memory networks and cognitive decline [228, 229, 352]. They also induce direct effects on neurons. Neurons upregulate IFITM3, which enhances γ-secretase activity, increasing APP processing, and Aβ production [229, 275]. Aβ triggers type I interferon production by microglia [228] and neurons [247] while contributing to amyloid plaque formation when coupled with interferon-induced defective phagocytosis from microglia [262]. MEF2C, a cognitive resilience transcription factor, is downregulated in microglia and neurons. This leads to dysfunctional pro-inflammatory responses in microglia and the initiation of a premature cell senescence programme, promoting neuronal loss [233, 249, 268]. Interferons trigger mitochondrial destabilisation in neurons, contributing to neuronal cell death [263, 264, 265] and through concurrent activation of upregulated nucleic acid sensors [266] and mitochondrial DNA release, they cause neuronal type I interferon production [505, 506, 507]. Signalling to oligodendrocytes [239] leads to as yet undefined consequences, and in neurons promotes neuronal tau hyperphosphorylation and seeded aggregation [237] further contributing to the neurodegeneration process. Type I interferons also contribute to significant changes in the blood-brain barrier. Endothelial cells respond to type I interferons by producing chemokines CCL2 and CXCL10 which mediate perivascular immune cell infiltration [166, 168, 171, 294, 295, 296]. Recruited immune cells are activated by interferons to upregulate ROS via NOX2 and IDO1, contributing to neuronal and BBB damage [156, 384, 387, 396, 397, 398, 399, 401]. Vascular endothelial cells from the BBB are concurrently activated by IFNAR-induced mediators CXCL10 and CCL2 to downregulate adherens and tight junctions, resulting in a leaky barrier and promoting microangiopathy [279, 280, 281, 282, 300]. The mechanism by which type I interferons cause calcification of vessels in the systemic circulation and basal ganglia is unknown, but it is thought to be related to premature ageing. MEF2-family of transcription factors are gatekeepers of cellular senescence. Cognitive resilience-associated MEF2C is downregulated by IFNAR signalling, precipitating loss of endogenous retroviral element regulators such as TREX1, thereby inducing aberrant triggering of nucleic acid sensors and leading to self-amplified type I interferon production associated with the senescence-associated secretory phenotype (SASP) [258, 268, 348, 349, 350, 351, 352] as is the loss of MEF2A which also causes unabated IFNβ production through increased DNA:RNA hybrid accumulation [508]. Type I interferon-induced CCL2 and CCL11 are thought to be linked to induction of cell senescence [166, 171, 359, 360, 361, 362] though the exact mechanisms remain to be fully elucidated. Finally, inhibition of NRF2 by IFNAR signalling leads to increased ROS which exacerbates the inflammaging process [339, 340, 341]. Put together, these mechanisms are responsible for ageing-related cognitive decline. These outcomes are overly apparent at the choroid plexus and likely take place diffusely across the brain parenchyma. Type I interferon is also well known to be an inducer of depression through metabolite, inflammatory, and hormonal mechanisms. IDO is induced in response to interferon and perturbs the balance between the excitotoxic quinolinic acid metabolite and the neuron protective kynurenic acid and serotonin, causing excessive NMDAR agonism [104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 156]. Cyclooxygenase inhibition via NSAIDs also attenuates IFNα-induced depression through undescribed mechanisms [173]. Concurrently, the interferon-inducible CXCL10 is also known to lead to weakened synaptic long-term potentiation in the hippocampus, resulting in a depressive phenotype [166, 171]. It remains to be elucidated whether the protective effects of COX1, -2, and PLA2 inhibition are acting through inhibition of the interferon-induced synaptic CXCL10-CXCR3 signalling. Together, all these mechanisms likely act in concert to cause the detrimental neuropsychiatric effects caused by type I interferons.

Advertisement

6. Therapeutic approaches

There are numerous existing approaches for targeting type I interferons and their downstream signalling, with great therapeutic application potential.

6.1 Targeting the receptor

The entire type I interferon pathway is dependent on a single juncture point, the IFNAR. This effectively acts as a central hub controlling all its downstream signalling. The advantage of targeting IFNAR over individually targeting any of its 13 IFNα subtypes, IFNβ, IFNε, IFNκ or IFNω, or combinations thereof, is evident. The generation of a mouse anti-IFNAR1 blocking monoclonal antibody [509] was a turning point for the field and for the pharmaceutical industry, both by facilitating research in vivo and elucidating the role of the pathway outside of genetic manipulation, and by providing a surrogate for the potential of therapeutic targeting in human. A fully human monoclonal IgG1κ antibody targets IFNAR1 with high affinity and specificity, neutralising its receptor activity [510]. Anifrolumab is now approved in most countries for the treatment of SLE following breakthrough improvement in disease scores, higher rate of achievement of remission, high retention rate and low immunogenicity and side effects [92, 93, 511, 512, 513, 514]. It is important to note that the percentage of herpes zoster reactivation was increased by anifrolumab compared to placebo, surprisingly no other notable adverse events were seen up to 3-years after treatment initiation [512, 515]. Furthermore, an indirect analysis of results from the TULIP-1 and TULIP-2 phase III trials of anifrolumab, and BLISS-52 and BLISS-76 trials, two phase III trials randomised, double-blind, placebo-controlled tials of belimumab, an anti-BAFF monoclonal antibody, reveals that treatment with anifrolumab was associated with significantly greater benefits [516]. Belimumab had been a revolution in the treatment of SLE [517, 518], and is the only other and first approved biologic for SLE. Its potent B-cell survival inhibitory effect is being leveraged in combination therapies with tacrolimus [519] and rituximab [520, 521]. While no head-to-head comparisons have been made between anifrolumab and belimumab, warranting caution on conclusions drawn, anifrolumab far exceeds the efficacy outlook for the current approved treatment landscape for SLE. That includes sifalimumab and rontalizumab, two anti-IFNα monoclonal antibodies, which have shown efficacy and good tolerability profiles [90, 522, 523, 524], but which lack the broad efficacy achieved by the complete blockade of type I interferon signalling.

The approval of anifrolumab, only the second biologic for lupus, reinvigorated the field and validated a long-known central role of type I interferons in SLE. This created excitement beyond the lupus field, as trials are being conducted for other indications including for Rheumatoid Arthritis (NCT03435601), Primary Sjögren’s Syndrome (NCT05383677), Systemic Sclerosis (NCT05925803), Progressive Vitiligo (NCT05917561), and Hidradenitis Suppurativa (NCT06374212). Regarding monogenic type I interferonopathies, as of yet, only one single patient carrying a DNASE2 mutation has received anifrolumab, with remarkable amelioration [525]. While neurological involvement is frequent in DNASE2 loss-of-function interferonopathies, this patient had normal neurologic status at the time of their treatment, thus it is not known whether this approach may be useful for treating psychocognitive symptoms.

6.2 JAK inhibitors

Inhibitors targeting the JAK/STAT signalling pathway have shown efficacy and promising results in various diseases. JAK inhibitors like tofacitinib, filgotinib, and upadacitinib have been approved for use in multiple disorders, modifying treatment algorithms across a range of disease states, including myeloproliferative neoplasms, rheumatoid arthritis, and various inflammatory dermatological disorders. The rapid and broad bench-to-bedside translation is a testament to the importance of this pathway to different disease mechanisms. For neurological diseases, however, there has been slower adoption, despite promising preclinical results. Notably, inhibition of JAK/STAT signalling in mouse models of DS resulted in attenuated interferon-response signatures [307]. Dp16 mice, which harbour a duplication of murine chromosome 16 and which is orthologous to human chromosome 21, show upregulation of inflammatory and interferon responses in brain, along with other tissues [307]. Treatment with the JAK1/2 inhibitor baricitinib caused attenuation of many dysregulated signatures of Dp16, including of interferon-response genes, and including in the brain. Importantly, a patient treated with tofacitinib for an underlying alopecia areata [526] displayed marked inhibition of interferon-response signatures in the circulation to levels similar to euploid and healthy individuals, including abrogation of the major signalling chemokines CXCL9 and CXCL10 [307]. Beyond neuroinflammation due to genomic aberrations specifically associated with DS, JAK/STAT signalling inhibition using ruxolitinib was also capable of inhibiting type I interferon-driven neurodegeneration in a model of ALS-FTD [527]. Specifically, C9orf72-mutation associated FTD and ALS are found to accumulate cytoplasmic dsRNAs which trigger production of type I interferons, and culminate in JAK/STAT-mediated neuronal cell death. Proof-of-concept evidence exists that the interferonopathy in DS and nucleic acid accumulation in FTD and ALS can be modulated pharmacologically by JAK/STAT inhibitors, and taken together with the mouse model assessments suggests that neurocognitive protection may be possible.

There are still questions about the efficacy of JAK/STAT inhibition for neurological disease. In both AGS [528, 529, 530, 531, 532], SPENCD [533], and SAVI [530] JAK1/2 inhibitors caused systemic amelioration of dermatologic or pulmonary disease, including of systemic type I interferon signatures, but neurological improvement was limited or absent. Frequent infections are also seen, as essentially all cytokine signalling is inhibited, and is an important consideration especially for paediatric patients lacking fully developed adaptive immunity relying on innate immunity. Often, this has led to transient discontinuation of therapy, which may have impacted outcomes. This suggests that targeted therapy, particularly one specifically targeting type I interferons, may be more efficacious. A handful of case studies have reported better success for neurologic signs in AGS [534, 535, 536], of which one demonstrated reduced type I interferon in CSF for one patient [534], arguing perhaps in favour of early and uninterrupted therapy. Assessment of CSF exposure of the inhibitors reveals levels below 10% that of plasma, which is reflected by poor biomarker responses in CSF compared to circulation [531] indicating a need for brain penetrant molecules for consistent responses.

6.3 Experimental therapeutic approaches

Few other interferon-targeting treatment approaches exist, and most remain for now experimental. Reverse-transcriptase inhibitors were found to strongly inhibit interferon-responses in circulation and in CSF in AGS [537]. It is thought that repressing expression of otherwise uncontrolled endogenous retroelements may be sufficient to regulate type I interferons, but perhaps not all patients may benefit similarly. These changes were most efficacious in patients carrying mutations in components of the RNASEH2 complex, and signatures were reversed upon discontinuation indicating specificity of the signal to the treatment. It is unclear if this treatment approach could be used for lasting responses with favourable clinical outcome, especially in the brain. It is also difficult to extrapolate from the clinical course of HAND where, despite anti-retroviral therapy, neurocognitive deterioration continues, as pathological mechanisms are different. Interestingly, trials are being conducted in Alzheimer’s disease (NCT04552795) which may yield better understanding of how retroelements are implicated in neuroinflammation [538].

Synthetic nucleotides for gene silencing are being intensely researched for drug development [539]. Antisense oligonucleotides (ASOs) hybridise cellular RNA and modulate processing, splicing, cause competitive inhibition, block the translational machinery, or degrade the bound target mRNAs. Treatments based on ASOs have already reached the clinic for genetic disorders such as Duchenne muscular dystrophy [540] and SOD1 mutation-associated ALS [541]. In a type I interferonopathy mouse model, intrathecal delivery of ASOs targeting the Ifnar1 gene rescued neuropathological phenotypes by reducing type I interferon-responses, which led to reduced gliosis, immune cell infiltration in brain, reduced neuronal death and tissue destruction, and prevention of BBB leakage [542]. Such an approach lends itself well to monogenic type I interferonopathies and, coupled with a rationale-driven administration method, may benefit patients based on their personalised symptomatology.

Advertisement

7. Conclusion and future directions

7.1 Summary and implications

Type I interferons are cytokines that play a crucial role in the innate immune response against viral infections and in anti-tumour activities within the CNS. Overactivity of this pathway, however, results in aberrant responses that cause neurodegeneration. Concrete examples of this are persistent viral infections of the brain causing HAND, neurocognitive symptoms in archetypically type I interferon-driven diseases such as SLE and AGS, and the profound psychocognitive effects of therapies with IFNα and IFNβ. Type I interferons are also found aberrantly overexpressed in more common diseases such as major depression, AD and dementia attributed to TBI, DS, and ageing, giving rise to the hypothesis that they may be detrimental more widely across neurodegeneration. With compelling evidence from preclinical models in all aforementioned disorders, there is interest in further developing therapeutics that target this pathway.

This also has implications for current clinical practice. The historical significance, extensive clinical experience, and diverse applications of IFNα and IFNβ therapies contribute to their sustained use in treatment practices. These once pioneering biologics remain relevant due to their foundational role in immunology and their proven benefits in some clinical indications. However, their long-lasting effects on the brain are an important consideration, and perhaps even more so due to their seeming implication in natural and disease-associated cognitive decline.

7.2 Limitations, challenges, and open questions

Open questions remain on the therapeutic potential of targeting type I interferon signalling for neurological disorders. SLE, a disease caused by systemic type I interferons, leads to neuropsychiatric symptoms, including depression, in a substantial proportion of patients, mirroring exogenous therapy with IFNα or IFNβ. It will therefore be important to evaluate if blockade of type I interferon signalling affects neuropsychiatric outcomes in lupus. The most direct evidence for this may come with the now marketed IFNAR1-targeting anifrolumab. Though trials performed to date were not powered for addressing this, long-term follow-up and more extensive clinical experience should clarify this. Also, it is unclear whether targeting the IFNAR systemically will be sufficient to influence the CNS. This may depend on the CNS-penetrance of anifrolumab, which has not been evaluated. Typical CNS penetrance of antibodies is 0.1%, so for a dose of 300 mg it may be conceivable that sufficient antibody reaches the brain parenchyma, but not necessarily that sufficient target is neutralised. High, ubiquitous, target expression explains a short half-life of the antibody, meaning lower amounts of free antibody capable of reaching the brain. PK/PD relationship studies from the TULIP-1 trial reveal that only the 300 mg dosing schedule allowed for approximately IC90 inhibition of interferon-response [513]. The lower dose group receiving 150 mg had sub-optimal or no inhibition of the systemic 21-IFNGS when the C average concentration in plasma was below 11.5 μg/mL, and at least 32 μg/mL plasma exposure was necessary to consistently and durably achieve IC90 [513]. This indicates that the current 300 mg IV Q4W regimen, while sufficient to block type I interferon signalling in circulation, may not be sufficient to achieve an effect in brain, and higher doses may be needed. Importantly, this problem applies to all neurologic and neurodegenerative diseases caused by type I interferons, so it remains to be determined whether anifrolumab in its current format may be sufficient to treat type I interferonopathies, AD, TBI, or depression.

Delivery of antibodies across the BBB is a hot research topic and remains a current challenge. Brain shuttling technologies are being developed [543], and higher brain penetrance is being achieved by adding shuttle peptides [544], receptor-mediated transcytosis mechanisms [545], and exosome [546] or viral [547] vector-mediated expression. Developing brain-penetrant IFNAR-inhibiting therapeutic compounds may be required to achieve neuropsychiatric or neurodegenerative improvement in type I interferon-driven diseases. Small molecules targeting upstream or downstream elements of the type I interferon pathway exist, and may be easier to be made brain penetrant.

Achieving clinical efficacy for neurologic disorders often requires long trials because of the inherent complexity of the nervous system and length of disease progression. Trials with anifrolumab have confirmed the importance of the type I interferon pathway in viral defence. Thus, further safety considerations may need to be addressed by future type I interferon-directed therapies, especially if made brain-targeting. Herpes zoster screening and patient selection may be needed to stave off serious adverse events, particularly in long trials. From a personalised medicine perspective, it may also become important to evaluate whether wider viral screening could inform response rates and side effects, as other immunological mechanisms may take over antiviral function.

In recent years there has been enormous advance in the use of neurological biomarkers, particularly NfL, following regulatory approval by the FDA as a surrogate marker of neurodegeneration in ALS [548, 549]. Unsurprisingly, the number of clinical trials of CNS diseases utilising this biomarker has been steadily increasing [548]. Broadened clinical use may also give further clues about the relationship with type I interferons and whether interferon-targeting therapies lead to any meaningful change in plasma or CSF NfL. NfL has already been shown to be correlated with IFNα in HAND [425]. It is unknown whether type I interferonopathies – archetypical neuroinflammatory diseases – display NfL release in plasma or CSF. This assessment could allow a better understanding of the mechanisms of neurodegeneration in a purely type I interferon-driven group of diseases In SLE, a disease characterised by systemic type I interferons, elevated NfL levels, particularly in patients with neuropsychiatric symptoms [83, 84, 86], may correlate with CSF or plasma IFNα or interferon-response signatures. As neuropsychiatric amelioration has for the most part not correlated well with general disease response, another interesting metric will be the assessment of NfL during therapy, and especially with the IFNAR pathway targeting therapies.

An important current consideration is whether it will be feasible to move away entirely from IFNα and IFNβ therapies. For MS, emerging treatments are proving to be more effective than IFNβ therapy, indicating a potential shift in therapeutic approaches [550]. In the context of chronic HCV, patients report better quality of life with oral antivirals than antivirals plus IFNα [551]. Newer generation antivirals may slowly phase out the need for interferons in viral infection [182, 183]. For cancer, new interest in the tumour microenvironment has created excitement leading to reticence in phasing out interferons from clinical care [552]. Some use may still remain for exogenous interferon therapies, particularly for high-risk patients in low-survivable diseases [553] where long-term neurocognitive effects may be less relevant or managed for the survival duration. Nevertheless, checkpoint blockade inhibitors [184], and immunomodulating therapies [155, 185] have allowed for advances in our understanding of cancer biology, and the creation of new classes of therapeutics.

Rare diseases can often inform findings relevant to more common diseases. Neurologic disorders are no exception. A prominent example is the role that early onset familial Alzheimer’s disease, a very rare monogenic form of Alzheimer’s, has had on shaping the understanding of sporadic disease. Perhaps the most important open question is whether our understanding of these relatively newly discovered type I interferonopathies may help develop therapeutics for more common, or equally devastating, neurologic diseases.

Advertisement

Acknowledgments

The author wishes to acknowledge the helpful editing, discussion, and scientific literature knowledge and contributions of Dr. Morgan Le Roux-Bourdieu and Rosaria Luongo. The author apologises for not being able to include the many other important articles that have contributed to the development of the type I interferon field.

Advertisement

Conflict of interest

A.M. is a full-time employee of AC Immune SA, a for-profit clinical-stage biotech. Other than salary, A.M. holds no equity, shares or financial conflicts of interest to disclose. The views expressed in this chapter are solely those of the author and the company had no role in the writing or editing of this chapter. At the time of writing, AC Immune SA is not pursuing any commercial interests related to interferons or their clinical use.

References

  1. 1. Crow YJ. Type I interferonopathies: A novel set of inborn errors of immunity. Annals of the New York Academy of Sciences. 2011;1238:91-98
  2. 2. Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proceedings of the Royal Society of London. Series B, Biological Sciences. 1957;147:258-267
  3. 3. Isaacs A, Lindenmann J, Valentine RC. Virus interference. II. Some properties of interferon. Proceedings of the Royal Society B: Biological Sciences. 1957;147:268-273
  4. 4. Merigan TC, Reed SE, Hall TS, et al. Inhibition of respiratory virus infection by locally applied interferon. Lancet. 1973;1:563-567
  5. 5. Gresser I, Bourali C, Lévy JP, et al. Increased survival in mice inoculated with tumor cells and treated with interferon preparations. Proceedings of the National Academy of Sciences of the United States of America. 1969;63:51-57
  6. 6. Haglund S, Lundquist PG, Cantell K, et al. Interferon therapy in juvenile laryngeal papillomatosis. Archives of Otolaryngology. 1981;107:327-332
  7. 7. Jacobs L, O’Malley J, Freeman A, et al. Intrathecal interferon reduces exacerbations of multiple sclerosis. Science. 1981;214:1026-1028
  8. 8. Priestman TJ. Initial evaluation of human lymphoblastoid interferon in patients with advanced malignant disease. Lancet. 1980;2:113-118
  9. 9. Scott GM, Secher DS, Flowers D, et al. Toxicity of interferon. British Medical Journal (Clinical Research Ed.). 1981;282:1345-1348
  10. 10. Smedley H, Katrak M, Sikora K, et al. Neurological effects of recombinant human interferon. British Medical Journal (Clinical Research Ed.). 1983;286:262-264
  11. 11. Rönnblom LE, Alm GV, Oberg KE. Possible induction of systemic lupus erythematosus by interferon-alpha treatment in a patient with a malignant carcinoid tumor. Journal of Internal Medicine. 1990;227:207-210
  12. 12. Gresser J, Morel-Maroger L, Verroust P, et al. Anti-interferon globulin inhibits the development of glomerulonephritis in mice infected at birth with lymphocytic choriomeningitis virus. Proceedings of the National Academy of Sciences of the United States of America. 1978;75:3413-3416
  13. 13. Gresser I. On the varied biologic effects of interferon. Cellular Immunology. 1977;34:406-415
  14. 14. Elliott MJ, Maini RN, Feldmann M, et al. Treatment of rheumatoid arthritis with chimeric monoclonal antibodies to tumor necrosis factor alpha. Arthritis and Rheumatism. 1993;58:S92-S101
  15. 15. Loncharich MF, Anderson CW. Interferon inhibition for lupus with anifrolumab: Critical appraisal of the evidence leading to FDA approval. ACR Open Rheumatology. 2022. pp. 486-491. DOI: 10.1002/acr2.11414
  16. 16. Aicardi J, Goutières F. A progressive familial encephalopathy in infancy with calcifications of the basal ganglia and chronic cerebrospinal fluid lymphocytosis. Annals of Neurology. 1984;15:49-54
  17. 17. Lebon P, Badoual J, Ponsot G, et al. Intrathecal synthesis of interferon-alpha in infants with progressive familial encephalopathy. Journal of the Neurological Sciences. 1988;84:201-208
  18. 18. Crow YJ, Hayward BE, Parmar R, et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 cause Aicardi-Goutières syndrome at the AGS1 locus. Nature Genetics. 2006;38:917-920
  19. 19. Crow YJ, Leitch A, Hayward BE, et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutières syndrome and mimic congenital viral brain infection. Nature Genetics. 2006;38:910-916
  20. 20. Duncan CJA, Thompson BJ, Chen R, et al. Severe type I interferonopathy and unrestrained interferon signaling due to a homozygous germline mutation in STAT2. Science Immunology. 2019;4. DOI: 10.1126/sciimmunol.aav7501
  21. 21. Duncan CJA, Hambleton S. Human disease phenotypes associated with loss and gain of function mutations in STAT2: Viral susceptibility and type I interferonopathy. Journal of Clinical Immunology. 2021;41:1446-1456
  22. 22. Kozlova АL, Leonteva МЕ, Burlakov VI, et al. Clinical case of type I interferonopathy: Homozygous STAT2 gain-of-function mutation. Voprosy Gematologii/onkologii i Immunopatologii v Pediatrii. 2021;20:132-139
  23. 23. Zhu G, Badonyi M, Franklin L, et al. Type I interferonopathy due to a homozygous loss-of-inhibitory function mutation in STAT2. Journal of Clinical Immunology. 2023;43:808-818
  24. 24. Weidler S, Koss S, Wolf C, et al. A rare manifestation of STING-associated vasculopathy with onset in infancy: A case report. Pediatric Rheumatology Online Journal. 2024;22:9
  25. 25. Schlee M, Hartmann G. Discriminating self from non-self in nucleic acid sensing. Nature Reviews. Immunology. 2016;16:566-580
  26. 26. Platanias LC. Introduction: Interferon signals: What is classical and what is nonclassical? Journal of Interferon & Cytokine Research. 2005;25:732
  27. 27. Majoros A, Platanitis E, Kernbauer-Hölzl E, et al. Canonical and non-canonical aspects of JAK–STAT signaling: Lessons from interferons for cytokine responses. Frontiers in Immunology. 2017;8:29
  28. 28. Mazewski C, Perez RE, Fish EN, et al. Type I interferon (IFN)-regulated activation of canonical and non-canonical signaling pathways. Frontiers in Immunology. 2020;11:606456
  29. 29. Kadowaki N, Antonenko S, Lau JY, et al. Natural interferon alpha/beta-producing cells link innate and adaptive immunity. The Journal of Experimental Medicine. 2000;192:219-226
  30. 30. Hoebe K, Janssen E, Beutler B. The interface between innate and adaptive immunity. Nature Immunology. 2004;5:971-974
  31. 31. Hertzog PJ. Overview. Type I interferons as primers, activators and inhibitors of innate and adaptive immune responses. Immunology and Cell Biology. 2012;90:471-473
  32. 32. Nesterova IV, Kovaleva SV, Malinovskaya VV, et al. Congenital and acquired interferonopathies: Differentiated approaches to interferon therapy. In: Innate Immunity in Health and Disease. London, UK: IntechOpen; 2020. Epub ahead of print 2020. DOI: 10.5772/intechopen.91723
  33. 33. Møller RS, Zhao L, Shoaff JR, et al. Incidence of Aicardi-Goutières syndrome and KCNT1-related epilepsy in Denmark. Molecular Genetics and Metabolism Reports. 2022;33:100924
  34. 34. Liu A, Ying S. Aicardi-Goutières syndrome: A monogenic type I interferonopathy. Scandinavian Journal of Immunology. 2023;98:e13314
  35. 35. du Moulin M, Nürnberg P, Crow YJ, et al. Cerebral vasculopathy is a common feature in Aicardi-Goutieres syndrome associated with SAMHD1 mutations. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:E232; author reply E233
  36. 36. Piccoli C, Bronner N, Gavazzi F, et al. Late-onset Aicardi-Goutières syndrome: A characterization of presenting clinical features. Pediatric Neurology. 2021;115:1-6
  37. 37. Barth PG, Walter A, van Gelderen I. Aicardi-Goutières syndrome: A genetic microangiopathy? Acta Neuropathologica. 1999;98:212-216
  38. 38. Barth PG. The neuropathology of Aicardi-Goutières syndrome. European Journal of Pediatric Neurology. 2002;6(Suppl. A):A27-A31 discussion A37
  39. 39. Østergaard JR, Christensen T. Aicardi-Goutières syndrome: Neuroradiological findings after nine years of follow-up. European Journal of Pediatric Neurology. 2004;8:243-246
  40. 40. Rasmussen M, Skullerud K, Bakke SJ, et al. Cerebral thrombotic microangiopathy and antiphospholipid antibodies in Aicardi-Goutieres syndrome–report of two sisters. Neuropediatrics. 2005;36:40-44
  41. 41. Maheshwari U, Huang S-F, Sridhar S, et al. The interplay between brain vascular calcification and microglia. Frontiers in Aging Neuroscience. 2022;14:848495
  42. 42. Rodero MP, Decalf J, Bondet V, et al. Detection of interferon alpha protein reveals differential levels and cellular sources in disease. The Journal of Experimental Medicine. 2017;214:1547-1555
  43. 43. Lodi L, Melki I, Bondet V, et al. Differential expression of interferon-alpha protein provides clues to tissue specificity across type I interferonopathies. Journal of Clinical Immunology. 2021;41:603-609
  44. 44. Viengkhou B, Hayashida E, McGlasson S, et al. The brain microvasculature is a primary mediator of interferon-α neurotoxicity in human cerebral interferonopathies. Immunity. 2024;57:1696-1709.e10
  45. 45. Rice GI, Forte GMA, Szynkiewicz M, et al. Assessment of interferon-related biomarkers in Aicardi-Goutières syndrome associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR: A case–control study. Lancet Neurology. 2013;12:1159-1169
  46. 46. Wang BX, Grover SA, Kannu P, et al. Interferon-stimulated gene expression as a preferred biomarker for disease activity in Aicardi-Goutières syndrome. Journal of Interferon & Cytokine Research. 2017;37:147-152
  47. 47. Kim H, de Jesus AA, Brooks SR, et al. Development of a validated interferon score using nanostring technology. Journal of Interferon & Cytokine Research. 2018;38:171-185
  48. 48. Pescarmona R, Belot A, Villard M, et al. Comparison of RT-qPCR and nanostring in the measurement of blood interferon response for the diagnosis of type I interferonopathies. Cytokine. 2019;113:446-452
  49. 49. de Jesus AA, Hou Y, Brooks S, et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. The Journal of Clinical Investigation. 2020;130(4):1669-1682
  50. 50. Demers-Mathieu V. Optimal selection of IFN-α-inducible genes to determine type I interferon signature improves the diagnosis of systemic lupus erythematosus. Biomedicine. 2023;11. DOI: 10.3390/biomedicines11030864
  51. 51. Rice GI, Bond J, Asipu A, et al. Mutations involved in Aicardi-Goutières syndrome implicate SAMHD1 as regulator of the innate immune response. Nature Genetics. 2009;41:829-832
  52. 52. Rice GI, Kasher PR, Forte GMA, et al. Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a type I interferon signature. Nature Genetics. 2012;44:1243-1248
  53. 53. Gray EE, Treuting PM, Woodward JJ, et al. Cutting edge: cGAS is required for lethal autoimmune disease in the Trex1-deficient mouse model of Aicardi-Goutières syndrome. Journal of Immunology. 2015;195:1939-1943
  54. 54. Ablasser A, Hemmerling I, Schmid-Burgk JL, et al. TREX1 deficiency triggers cell-autonomous immunity in a cGAS-dependent manner. Journal of Immunology. 2014;192:5993-5997
  55. 55. Lee-Kirsch MA, Gong M, Chowdhury D, et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 are associated with systemic lupus erythematosus. Nature Genetics. 2007;39:1065-1067
  56. 56. Rice G, Newman WG, Dean J, et al. Heterozygous mutations in TREX1 cause familial chilblain lupus and dominant Aicardi-Goutieres syndrome. American Journal of Human Genetics. 2007;80:811-815
  57. 57. Günther C, Kind B, Reijns MAM, et al. Defective removal of ribonucleotides from DNA promotes systemic autoimmunity. The Journal of Clinical Investigation. 2015;125:413-424
  58. 58. Pokatayev V, Hasin N, Chon H, et al. RNase H2 catalytic core Aicardi-Goutières syndrome-related mutant invokes cGAS-STING innate immune-sensing pathway in mice. The Journal of Experimental Medicine. 2016;213:329-336
  59. 59. Uggenti C, Lepelley A, Depp M, et al. cGAS-mediated induction of type I interferon due to inborn errors of histone pre-mRNA processing. Nature Genetics. 2020;52:1364-1372
  60. 60. Jiao H, Wachsmuth L, Wolf S, et al. ADAR1 averts fatal type I interferon induction by ZBP1. Nature. 2022;607:776-783
  61. 61. Hubbard NW, Ames JM, Maurano M, et al. ADAR1 mutation causes ZBP1-dependent immunopathology. Nature. 2022;607:769-775
  62. 62. de Reuver R, Verdonck S, Dierick E, et al. ADAR1 prevents autoinflammation by suppressing spontaneous ZBP1 activation. Nature. 2022;607:784-789
  63. 63. Rice GI, Del Toro DY, Jenkinson EM, et al. Gain-of-function mutations in IFIH1 cause a spectrum of human disease phenotypes associated with upregulated type I interferon signaling. Nature Genetics. 2014;46:503-509
  64. 64. Oda H, Nakagawa K, Abe J, et al. Aicardi-Goutières syndrome is caused by IFIH1 mutations. American Journal of Human Genetics. 2014;95:121-125
  65. 65. Zhang X, Bogunovic D, Payelle-Brogard B, et al. Human intracellular ISG15 prevents interferon-α/β over-amplification and auto-inflammation. Nature. 2015;517:89-93
  66. 66. Martin-Fernandez M, Bravo García-Morato M, Gruber C, et al. Systemic type I IFN inflammation in human ISG15 deficiency leads to necrotizing skin lesions. Cell Reports. 2020;31:107633
  67. 67. Meuwissen MEC, Schot R, Buta S, et al. Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome. The Journal of Experimental Medicine. 2016;213:1163-1174
  68. 68. François-Newton V, de Freitas M, Almeida G, Payelle-Brogard B, et al. USP18-based negative feedback control is induced by type I and type III interferons and specifically inactivates interferon α response. PLoS One. 2011;6:e22200
  69. 69. Francois-Newton V, Livingstone M, Payelle-Brogard B, et al. USP18 establishes the transcriptional and anti-proliferative interferon α/β differential. The Biochemical Journal. 2012;446:509-516
  70. 70. Goldmann T, Zeller N, Raasch J, et al. USP18 lack in microglia causes destructive interferonopathy of the mouse brain. The EMBO Journal. 2015;34:1612-1629
  71. 71. Girschick H, Wolf C, Morbach H, et al. Severe immune dysregulation with neurological impairment and minor bone changes in a child with spondyloenchondrodysplasia due to two novel mutations in the ACP5 gene. Pediatric Rheumatology Online Journal. 2015;13:37
  72. 72. Briggs TA, Rice GI, Adib N, et al. Spondyloenchondrodysplasia due to mutations in ACP5: A comprehensive survey. Journal of Clinical Immunology. 2016;36:220-234
  73. 73. Bilginer Y, Düzova A, Topaloğlu R, et al. Three cases of spondyloenchondrodysplasia (SPENCD) with systemic lupus erythematosus: A case series and review of the literature. Lupus. 2016;25:760-765
  74. 74. Kim H, Sanchez GAM, Goldbach-Mansky R. Insights from mendelian interferonopathies: Comparison of CANDLE, SAVI with AGS, Monogenic Lupus. Journal of Molecular Medicine. 2016;94:1111-1127
  75. 75. Demirkaya E, Sahin S, Romano M, et al. New horizons in the genetic etiology of systemic lupus erythematosus and lupus-like disease: Monogenic lupus and beyond. Journal of Clinical Medicine. 2020;9. DOI: 10.3390/jcm9030712
  76. 76. Tian J, Zhang D, Yao X, et al. Global epidemiology of systemic lupus erythematosus: A comprehensive systematic analysis and modeling study. Annals of the Rheumatic Diseases. 2023;82:351-356
  77. 77. Hanly JG, Li Q , Su L, et al. Psychosis in systemic lupus erythematosus: Results from an international inception cohort study. Arthritis & Rhematology. 2019;71:281-289
  78. 78. Govoni M, Hanly JG. The management of neuropsychiatric lupus in the 21st century: Still so many unmet needs? Rheumatology (Oxford). 2020;59:v52-v62
  79. 79. Unterman A, Nolte JES, Boaz M, et al. Neuropsychiatric syndromes in systemic lupus erythematosus: A meta-analysis. Seminars in Arthritis and Rheumatism. 2011;41:1-11
  80. 80. Sarwar S, Mohamed AS, Rogers S, et al. Neuropsychiatric systemic lupus erythematosus: A 2021 update on diagnosis, management, and current challenges. Cureus. 2021;13:e17969
  81. 81. ACR Ad Hoc Committee on Neuropsychiatric Lupus Nomenclature. The American college of rheumatology nomenclature and case definitions for neuropsychiatric lupus syndromes. Arthritis and Rheumatism. 1999;42:599-608
  82. 82. Tjensvoll AB, Lauvsnes MB, Zetterberg H, et al. Neurofilament light is a biomarker of brain involvement in lupus and primary Sjögren’s syndrome. Journal of Neurology. 2021;268:1385-1394
  83. 83. Engel S, Boedecker S, Marczynski P, et al. Association of serum neurofilament light chain levels and neuropsychiatric manifestations in systemic lupus erythematosus. Therapeutic Advances in Neurological Disorders. 2021;14:17562864211051496
  84. 84. Zervides KA, Janelidze S, Nystedt J, et al. Plasma and cerebrospinal fluid neurofilament light concentrations reflect neuronal damage in systemic lupus erythematosus. BMC Neurology. 2022;22:467
  85. 85. Gaetani L, Blennow K, Calabresi P, et al. Neurofilament light chain as a biomarker in neurological disorders. Journal of Neurology, Neurosurgery, and Psychiatry. 2019;90:870-881
  86. 86. Lauvsnes MB, Zetterberg H, Blennow K, et al. Neurofilament light in plasma is a potential biomarker of central nervous system involvement in systemic lupus erythematosus. Journal of Neurology. 2022;269:3064-3074
  87. 87. de Vries B, Steup-Beekman GM, Haan J, et al. TREX1 gene variant in neuropsychiatric systemic lupus erythematosus. Annals of the Rheumatic Diseases. 2010;69:1886-1887
  88. 88. Namjou B, Kothari PH, Kelly JA, et al. Evaluation of the TREX1 gene in a large multi-ancestral lupus cohort. Genes and Immunity. 2011;12:270-279
  89. 89. Labouret M, Costi S, Bondet V, et al. Juvenile neuropsychiatric systemic lupus erythematosus: Identification of novel central neuroinflammation biomarkers. Journal of Clinical Immunology. 2023;43:615-624
  90. 90. Khamashta M, Merrill JT, Werth VP, et al. Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: A randomized, double-blind, placebo-controlled study. Annals of the Rheumatic Diseases. 2016;75:1909-1916
  91. 91. Houssiau FA, Thanou A, Mazur M, et al. IFN-α kinoid in systemic lupus erythematosus: Results from a phase IIb, randomized, placebo-controlled study. Annals of the Rheumatic Diseases. 2020;79:347-355
  92. 92. Vital EM, Merrill JT, Morand EF, et al. Anifrolumab efficacy and safety by type I interferon gene signature and clinical subgroups in patients with SLE: Post hoc analysis of pooled data from two phase III trials. Annals of the Rheumatic Diseases. 2022;81:951-961
  93. 93. Kalunian KC, Furie R, Morand EF, et al. A randomized, placebo-controlled phase III extension trial of the long-term safety and tolerability of anifrolumab in active systemic lupus erythematosus. Arthritis & Rhematology. 2023;75:253-265
  94. 94. Yerram KV, Baisya R, Kumar P, et al. Serum interferon-alpha predicts in-hospital mortality in patients hospitalized with acute severe lupus. Lupus Science & Medicine. 2023;10. DOI: 10.1136/lupus-2023-000933
  95. 95. Lebon P, Lenoir GR, Fischer A, et al. Synthesis of intrathecal interferon in systemic lupus erythematosus with neurological complications. British Medical Journal (Clinical Research Edition). 1983;287:1165
  96. 96. Shiozawa S, Kuroki Y, Kim M, et al. Interferon-alpha in lupus psychosis. Arthritis and Rheumatism. 1992;35:417-422
  97. 97. Fragoso-Loyo H, Atisha-Fregoso Y, Núñez-Alvarez CA, et al. Utility of interferon-α as a biomarker in central neuropsychiatric involvement in systemic lupus erythematosus. The Journal of Rheumatology. 2012;39:504-509
  98. 98. Deane JA, Pisitkun P, Barrett RS, et al. Control of toll-like receptor 7 expression is essential to restrict autoimmunity and dendritic cell proliferation. Immunity. 2007;27:801-810
  99. 99. Richard ML, Gilkeson G. Mouse models of lupus: What they tell us and what they do not. Lupus Science & Medicine. 2018;5:e000199
  100. 100. Aw E, Zhang Y, Yalcin E, et al. Spatial enrichment of the type 1 interferon signature in the brain of a neuropsychiatric lupus murine model. Brain, Behavior, and Immunity. 2023;114:511-522
  101. 101. Nomura A, Noto D, Murayama G, et al. Unique primed status of microglia under the systemic autoimmune condition of lupus-prone mice. Arthritis Research & Therapy. 2019;21:303
  102. 102. Ramanujam M, Kahn P, Huang W, et al. Interferon-alpha treatment of female (NZW x BXSB)F(1) mice mimics some but not all features associated with the Yaa mutation. Arthritis and Rheumatism. 2009;60:1096-1101
  103. 103. Zeng J, Meng X, Zhou P, et al. Interferon-α exacerbates neuropsychiatric phenotypes in lupus-prone mice. Arthritis Research & Therapy. 2019;21:205
  104. 104. Åkesson K, Pettersson S, Ståhl S, et al. Kynurenine pathway is altered in patients with SLE and associated with severe fatigue. Lupus Science & Medicine. 2018;5:e000254
  105. 105. Vogelgesang SA, Heyes MP, West SG, et al. Quinolinic acid in patients with systemic lupus erythematosus and neuropsychiatric manifestations. The Journal of Rheumatology. 1996;23:850-855
  106. 106. Eryavuz Onmaz D, Tezcan D, Yilmaz S, et al. Altered kynurenine pathway metabolism and association with disease activity in patients with systemic lupus. Amino Acids. 2023;55:1937-1947
  107. 107. Anderson EW, Fishbein J, Hong J, et al. Quinolinic acid, a kynurenine/tryptophan pathway metabolite, associates with impaired cognitive test performance in systemic lupus erythematosus. Lupus Science & Medicine. 2021;8. DOI: 10.1136/lupus-2021-000559
  108. 108. Yan EB, Frugier T, Lim CK, et al. Activation of the kynurenine pathway and increased production of the excitotoxin quinolinic acid following traumatic brain injury in humans. Journal of Neuroinflammation. 2015;12:110
  109. 109. Pérez-De La Cruz V, Carrillo-Mora P, Santamaría A. Quinolinic acid, an endogenous molecule combining excitotoxicity, oxidative stress and other toxic mechanisms. International Journal of Tryptophan Research. 2012;5:1-8
  110. 110. Latif-Hernandez A, Shah D, Ahmed T, et al. Quinolinic acid injection in mouse medial prefrontal cortex affects reversal learning abilities, cortical connectivity and hippocampal synaptic plasticity. Scientific Reports. 2016;6:36489
  111. 111. Anderson EW, Jin Y, Shih A, et al. Associations between circulating interferon and kynurenine/tryptophan pathway metabolites: Support for a novel potential mechanism for cognitive dysfunction in SLE. Lupus Science & Medicine. 2022;9. DOI: 10.1136/lupus-2022-000808
  112. 112. Hestad K, Alexander J, Rootwelt H, et al. The role of tryptophan dysmetabolism and quinolinic acid in depressive and neurodegenerative diseases. Biomolecules. 2022;12. DOI: 10.3390/biom12070998
  113. 113. Wichers MC, Koek GH, Robaeys G, et al. IDO and interferon-alpha-induced depressive symptoms: A shift in hypothesis from tryptophan depletion to neurotoxicity. Molecular Psychiatry. 2005;10:538-544
  114. 114. Lood C, Tydén H, Gullstrand B, et al. Type I interferon-mediated skewing of the serotonin synthesis is associated with severe disease in systemic lupus erythematosus. PLoS One. 2015;10:e0125109
  115. 115. Müller N, Schwarz MJ. The immune-mediated alteration of serotonin and glutamate: Toward an integrated view of depression. Molecular Psychiatry. 2007;12:988-1000
  116. 116. Meyerhoff J, Dorsch CA. Decreased platelet serotonin levels in systemic lupus erythematosus. Arthritis and Rheumatism. 1981;24:1495-1500
  117. 117. Nikolopoulos D, Nakos-Bimpos M, Manolakou T, et al. Impaired serotonin synthesis in hippocampus of murine lupus represents an early neuropsychiatric event. Lupus. 2024;33:166-171
  118. 118. Merimsky O, Reider-Groswasser I, Inbar M, et al. Interferon-related mental deterioration and behavioral changes in patients with renal cell carcinoma. European Journal of Cancer. 1990;26:596-600
  119. 119. Poutiainen E, Hokkanen L, Niemi ML, et al. Reversible cognitive decline during high-dose alpha-interferon treatment. Pharmacology, Biochemistry, and Behavior. 1994;47:901-905
  120. 120. Valentine AD, Meyers CA, Kling MA, et al. Mood and cognitive side effects of interferon-alpha therapy. Seminars in Oncology. 1998;25:39-47
  121. 121. Capuron L, Ravaud A. Prediction of the depressive effects of interferon alfa therapy by the patient’s initial affective state. The New England Journal of Medicine. 1999;340:1370-1370
  122. 122. Dieperink E, Willenbring M, Ho SB. Neuropsychiatric symptoms associated with hepatitis C and interferon alpha: A review. The American Journal of Psychiatry. 2000;157:867-876
  123. 123. Musselman DL, Lawson DH, Gumnick JF, et al. Paroxetine for the prevention of depression induced by high-dose interferon alfa. The New England Journal of Medicine. 2001;344:961-966
  124. 124. Hauser P, Khosla J, Aurora H, et al. A prospective study of the incidence and open-label treatment of interferon-induced major depressive disorder in patients with hepatitis C. Molecular Psychiatry. 2002;7:942-947
  125. 125. Gohier B, Goeb J-L, Rannou-Dubas K, et al. Hepatitis C, alpha interferon, anxiety and depression disorders: A prospective study of 71 patients. The World Journal of Biological Psychiatry. 2003;4:115-118
  126. 126. Reichenberg A, Gorman JM, Dieterich DT. Interferon-induced depression and cognitive impairment in hepatitis C virus patients: A 72 week prospective study. AIDS. 2005;19(Suppl. 3):S174-S178
  127. 127. Heinze S, Egberts F, Rötzer S, et al. Depressive mood changes and psychiatric symptoms during 12-month low-dose interferon-alpha treatment in patients with malignant melanoma: Results from the multicenter DeCOG trial. Journal of Immunotherapy. 2010;33:106-114
  128. 128. Udina M, Castellví P, Moreno-España J, et al. Interferon-induced depression in chronic hepatitis C: A systematic review and meta-analysis. The Journal of Clinical Psychiatry. 2012;73:1128-1138
  129. 129. Baranyi A, Meinitzer A, Stepan A, et al. A biopsychosocial model of interferon-alpha-induced depression in patients with chronic hepatitis C infection. Psychotherapy and Psychosomatics. 2013;82:332-340
  130. 130. Cattie JE, Letendre SL, Woods SP, et al. Persistent neurocognitive decline in a clinic sample of hepatitis C virus-infected persons receiving interferon and ribavirin treatment. Journal of Neurovirology. 2014;20:561-570
  131. 131. Huckans M, Fuller B, Wheaton V, et al. A longitudinal study evaluating the effects of interferon-alpha therapy on cognitive and psychiatric function in adults with chronic hepatitis C. Journal of Psychosomatic Research. 2015;78:184-192
  132. 132. Sarkar S, Sarkar R, Berg T, et al. Sadness and mild cognitive impairment as predictors for interferon-alpha-induced depression in patients with hepatitis C. The British Journal of Psychiatry. 2015;206:45-51
  133. 133. Chiu WC, Su YP, Su KP, et al. Recurrence of depressive disorders after interferon-induced depression. Translational Psychiatry. 2017;7:e1026
  134. 134. Jain KK. Drug-induced Neurological Disorders. Cham: Springer International Publishing; 2021. Epub ahead of print 2021. DOI: 10.1007/978-3-030-73503-6
  135. 135. Meyers CA, Scheibel RS, Forman AD. Persistent neurotoxicity of systemically administered interferon-alpha. Neurology. 1991;41:672-676
  136. 136. Pavol MA, Meyers CA, Rexer JL, et al. Pattern of neurobehavioral deficits associated with interferon alfa therapy for leukemia. Neurology. 1995;45:947-950
  137. 137. Capuron L, Ravaud A, Dantzer R. Timing and specificity of the cognitive changes induced by interleukin-2 and interferon-alpha treatments in cancer patients. Psychosomatic Medicine. 2001;63:376-386
  138. 138. Capuron L, Gumnick JF, Musselman DL, et al. Neurobehavioral effects of interferon-alpha in cancer patients: Phenomenology and paroxetine responsiveness of symptom dimensions. Neuropsychopharmacology. 2002;26:643-652
  139. 139. Wichers MC, Koek GH, Robaeys G, et al. Early increase in vegetative symptoms predicts IFN-alpha-induced cognitive-depressive changes. Psychological Medicine. 2005;35:433-441
  140. 140. Raison CL, Demetrashvili M, Capuron L, et al. Neuropsychiatric adverse effects of interferon-alpha: Recognition and management. CNS Drugs. 2005;19:105-123
  141. 141. Kluin-Nelemans HC, Buck G, le Cessie S, et al. Randomized comparison of low-dose versus high-dose interferon-Alfa in chronic myeloid leukemia: Prospective collaboration of 3 joint trials by the MRC and HOVON groups. Blood. 2004;103:4408-4415
  142. 142. Morasco BJ, Loftis JM, Indest DW, et al. Prophylactic antidepressant treatment in patients with hepatitis C on antiviral therapy: A double-blind, placebo-controlled trial. Psychosomatics. 2010;51:401-408
  143. 143. Diez-Quevedo C, Masnou H, Planas R, et al. Prophylactic treatment with escitalopram of pegylated interferon alfa-2a-induced depression in hepatitis C: A 12-week, randomized, double-blind, placebo-controlled trial. The Journal of Clinical Psychiatry. 2011;72:522-528
  144. 144. Dipasquale O, Cooper EA, Tibble J, et al. Interferon-α acutely impairs whole-brain functional connectivity network architecture - A preliminary study. Brain, Behavior, and Immunity. 2016;58:31-39
  145. 145. Højsgaard Chow H, Schreiber K, Magyari M, et al. Progressive multiple sclerosis, cognitive function, and quality of life. Brain and Behavior: A Cognitive Neuroscience Perspective. 2018;8:e00875
  146. 146. Heesen C, Schulz KH, Fiehler J, et al. Correlates of cognitive dysfunction in multiple sclerosis. Brain, Behavior, and Immunity. 2010;24:1148-1155
  147. 147. Rocca MA, Valsasina P, Hulst HE, et al. Functional correlates of cognitive dysfunction in multiple sclerosis: A multicenter fMRI Study. Human Brain Mapping. 2014;35:5799-5814
  148. 148. Chiba K, Kataoka H, Seki N, et al. Fingolimod (FTY720), sphingosine 1-phosphate receptor modulator, shows superior efficacy as compared with interferon-β in mouse experimental autoimmune encephalomyelitis. International Immunopharmacology. 2011;11:366-372
  149. 149. Pozzilli C, Prosperini L, Borriello G. Treating multiple sclerosis with fingolimod or intramuscular interferon. Expert Opinion on Pharmacotherapy. 2010;11:1957-1960
  150. 150. Cohen JA, Barkhof F, Comi G, et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. The New England Journal of Medicine. 2010;362:402-415
  151. 151. Gasperini C, Ruggieri S. Development of oral agent in the treatment of multiple sclerosis: How the first available oral therapy, fingolimod will change therapeutic paradigm approach. Drug Design, Development and Therapy. 2012;6:175-186
  152. 152. El Ayoubi NK, Bou Reslan SW, Baalbaki MM, et al. Effect of fingolimod vs. interferon treatment on OCT measurements and cognitive function in RRMS. Multiple Sclerosis and Related Disorders. 2021;53:103041
  153. 153. Krupp L, Banwell B, Chitnis T, et al. Effect of fingolimod on health-related quality of life in pediatric patients with multiple sclerosis: Results from the phase 3 PARADIGMS study. BMJ Neurology Open. 2022;4:e000215
  154. 154. Bascuñana P, Möhle L, Brackhan M, et al. Fingolimod as a treatment in neurologic disorders beyond multiple sclerosis. Drugs in Research & Development. 2020;20:197-207
  155. 155. Yang JH, Rempe T, Whitmire N, et al. Therapeutic advances in multiple sclerosis. Frontiers in Neurology. 2022;13:824926
  156. 156. Wichers MC, Maes M. The role of indoleamine 2,3-dioxygenase (IDO) in the pathophysiology of interferon-alpha-induced depression. Journal of Psychiatry & Neuroscience. 2004;29:11-17
  157. 157. Kirkegaard C, Faber J. The role of thyroid hormones in depression. European Journal of Endocrinology. 1998;138:1-9
  158. 158. Hage M, Azar S. The link between thyroid function and depression. Journal of Thyroid Research. 2020;2012
  159. 159. Thvilum M, Brandt F, Almind D, et al. Increased psychiatric morbidity before and after the diagnosis of hypothyroidism: A nationwide register study. Thyroid. 2014;24:802-808
  160. 160. Bauer M, Whybrow PC. Role of thyroid hormone therapy in depressive disorders. Journal of Endocrinological Investigation. 2021;44:2341-2347
  161. 161. Loftis JM, Wall JM, Linardatos E, et al. A quantitative assessment of depression and thyroid dysfunction secondary to interferon-alpha therapy in patients with hepatitis C. Journal of Endocrinological Investigation. 2004;27:RC16-20
  162. 162. Carella C, Mazziotti G, Amato G, et al. Interferon-α-related thyroid disease: Pathophysiological, epidemiological, and clinical aspects. The Journal of Clinical Endocrinology & Metabolism. 2004;89:3656-3661
  163. 163. Tran HA, Reeves GEM, Jones TL. The natural history of interferon-alpha2b-induced thyroiditis and its exclusivity in a cohort of patients with chronic hepatitis C infection. QJM. 2009;102:117-122
  164. 164. Yan Z, Fan K, Fan Y, et al. Thyroid dysfunction in Chinese patients with chronic hepatitis C treated with interferon alpha: Incidence, long-term outcome and predictive factors. Hepatitis Monthly. 2012;12:e6390
  165. 165. Nair Kesavachandran C, Haamann F, Nienhaus A. Frequency of thyroid dysfunctions during interferon alpha treatment of single and combination therapy in hepatitis C virus-infected patients: A systematic review based analysis. PLoS One. 2013;8:e55364
  166. 166. Blank T, Detje CN, Spieß A, et al. Brain endothelial- and epithelial-specific interferon receptor chain 1 drives virus-induced sickness behavior and cognitive impairment. Immunity. 2016;44:901-912
  167. 167. Hunt D, Kavanagh D, Drummond I, et al. Thrombotic microangiopathy associated with interferon beta. The New England Journal of Medicine. 2014;370:1270-1271
  168. 168. Kavanagh D, McGlasson S, Jury A, et al. Type I interferon causes thrombotic microangiopathy by a dose-dependent toxic effect on the microvasculature. Blood. 2016;128:2824-2833
  169. 169. Wang C, Fang W, Sun W, et al. Clinical characteristics, treatments, and outcomes of interferon-beta-induced thrombotic microangiopathy: A literature-based retrospective analysis. Therapeutic Advances in Neurological Disorders. 2023;16:17562864231216634
  170. 170. Sarbu N, Alobeidi F, Toledano P, et al. Brain abnormalities in newly diagnosed neuropsychiatric lupus: Systematic MRI approach and correlation with clinical and laboratory data in a large multicenter cohort. Autoimmunity Reviews. 2015;14:153-159
  171. 171. Vlkolinský R, Siggins GR, Campbell IL, et al. Acute exposure to CXC chemokine ligand 10, but not its chronic astroglial production, alters synaptic plasticity in mouse hippocampal slices. Journal of Neuroimmunology. 2004;150:37-47
  172. 172. Su K-P, Huang S-Y, Peng C-Y, et al. Phospholipase A2 and cyclooxygenase 2 genes influence the risk of interferon-alpha-induced depression by regulating polyunsaturated fatty acids levels. Biological Psychiatry. 2010;67:550-557
  173. 173. Mesripour A, Shahnooshi S, Hajhashemi V. Celecoxib, ibuprofen, and indomethacin alleviate depression-like behavior induced by interferon-Alfa in mice. Journal of Complementary and Integrative Medicine. 2019;17. DOI: 10.1515/jcim-2019-0016
  174. 174. Köhler O, Benros ME, Nordentoft M, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: A systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry. 2014;71:1381-1391
  175. 175. Bai S, Guo W, Feng Y, et al. Efficacy and safety of anti-inflammatory agents for the treatment of major depressive disorder: A systematic review and meta-analysis of randomized controlled trials. Journal of Neurology, Neurosurgery, and Psychiatry. 2020;91:21-32
  176. 176. Husain MI, Chaudhry IB, Khoso AB, et al. Minocycline and celecoxib as adjunctive treatments for bipolar depression: A multicentre, factorial design randomized controlled trial. Lancet Psychiatry. 2020;7:515-527
  177. 177. Hoyo-Becerra C, Huebener A, Trippler M, et al. Concomitant interferon alpha stimulation and TLR3 activation induces neuronal expression of depression-related genes that are elevated in the brain of suicidal persons. PLoS One. 2013;8:e83149
  178. 178. Mostafavi S, Battle A, Zhu X, et al. Type I interferon signaling genes in recurrent major depression: Increased expression detected by whole-blood RNA sequencing. Molecular Psychiatry. 2014;19:1267-1274
  179. 179. Wong ML, Dong C, Maestre-Mesa J, et al. Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Molecular Psychiatry. 2008;13:800-812
  180. 180. Wallensten J, Ljunggren G, Nager A, et al. Stress, depression, and risk of dementia - A cohort study in the total population between 18 and 65 years old in region Stockholm. Alzheimer’s Research & Therapy. 2023;15:161
  181. 181. Gochee PA, Powell EE, Purdie DM, et al. Association between apolipoprotein E epsilon4 and neuropsychiatric symptoms during interferon alpha treatment for chronic hepatitis C. Psychosomatics. 2004;45:49-57
  182. 182. Sperl J, Horvath G, Halota W, et al. Efficacy and safety of elbasvir/grazoprevir and sofosbuvir/pegylated interferon/ribavirin: A phase III randomized controlled trial. Journal of Hepatology. 2016;65:1112-1119
  183. 183. Murira A, Lamarre A. Type-I interferon responses: From friend to FOE in the battle against chronic viral infection. Frontiers in Immunology. 2016;7:609
  184. 184. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350-1355
  185. 185. Goldschmidt CH, Hua LH. Re-evaluating the use of IFN-β and relapsing multiple sclerosis: Safety, efficacy and place in therapy. Degenerative Neurological and Neuromuscular Disease. 2020;10:29-38
  186. 186. Hou Y, Dan X, Babbar M, et al. Aging as a risk factor for neurodegenerative disease. Nature Reviews. Neurology. 2019;15:565-581
  187. 187. Goate A, Chartier-Harlin MC, Mullan M, et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature. 1991;349:704-706
  188. 188. Chartier-Harlin MC, Crawford F, Houlden H, et al. Early-onset alzheimer’s disease caused by mutations at codon 717 of the beta-amyloid precursor protein gene. Nature. 1991;353:844-846
  189. 189. Sherrington R, Rogaev EI, Liang Y, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754-760
  190. 190. Levy-Lahad E, Wasco W, Poorkaj P, et al. Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science. 1995;269:973-977
  191. 191. Esquerda-Canals G, Montoliu-Gaya L, Güell-Bosch J, et al. Mouse models of Alzheimer’s disease. Journal of Alzheimer’s Disease. 2017;57:1171-1183
  192. 192. Budd Haeberlein S, Aisen PS, Barkhof F, et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. The Journal of Prevention of Alzheimer’s Disease. 2022;9:197-210
  193. 193. van Dyck CH, Swanson CJ, Aisen P, et al. Lecanemab in early Alzheimer’s disease. The New England Journal of Medicine. 2023;388:9-21
  194. 194. Goedert M. Tau filaments in neurodegenerative diseases. FEBS Letters. 2018;592:2383-2391
  195. 195. Augustinack JC, Schneider A, Mandelkow E-M, et al. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathologica. 2002;103:26-35
  196. 196. Braak H, Alafuzoff I, Arzberger T, et al. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathologica. 2006;112:389-404
  197. 197. Arriagada PV, Growdon JH, Hedley-Whyte ET, et al. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology. 1992;42:631-639
  198. 198. Dronse J, Fliessbach K, Bischof GN, et al. In vivo patterns of tau pathology, amyloid-β burden, and neuronal dysfunction in clinical variants of Alzheimer’s disease. Journal of Alzheimer’s Disease. 2017;55:465-471
  199. 199. Congdon EE, Ji C, Tetlow AM, et al. Tau-targeting therapies for Alzheimer disease: Current status and future directions. Nature Reviews. Neurology. 2023;19:715-736
  200. 200. Corder EH, Saunders AM, Strittmatter WJ, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261:921-923
  201. 201. Farrer LA, Cupples LA, Haines JL, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. JAMA. 1997;278:1349
  202. 202. Blumenfeld J, Yip O, Kim MJ, et al. Cell type-specific roles of APOE4 in Alzheimer disease. Nature Reviews. Neuroscience. 2024;25:91-110
  203. 203. Arboleda-Velasquez JF, Lopera F, O’Hare M, et al. Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: A case report. Nature Medicine. 2019;25:1680-1683
  204. 204. Chen Y, Song S, Parhizkar S, et al. APOE3ch alters microglial response and suppresses Aβ-induced tau seeding and spread. Cell. 2024;187:428-445.e20
  205. 205. Lambert JC, Ibrahim-Verbaas CA, Harold D, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nature Genetics. 2013;45:1452-1458
  206. 206. Marioni RE, Harris SE, Zhang Q , et al. GWAS on family history of Alzheimer’s disease. Translational Psychiatry. 2018;8:99
  207. 207. Jansen IE, Savage JE, Watanabe K, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nature Genetics. 2019;51:404-413
  208. 208. Kunkle BW, Grenier-Boley B, Sims R, et al. Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nature Genetics. 2019;51:414-430
  209. 209. Wightman DP, Jansen IE, Savage JE, et al. Largest GWAS (N = 1,126,563) of Alzheimer’s disease implicates microglia and immune cells. medRxiv. 2020. DOI: 10.1101/2020.11.20.20235275
  210. 210. Guerreiro R, Wojtas A, Bras J, et al. TREM2 variants in Alzheimer’s disease. The New England Journal of Medicine. 2013;368:117-127
  211. 211. Jonsson T, Stefansson H, Steinberg S, et al. Variant of TREM2 associated with the risk of Alzheimer’s disease. The New England Journal of Medicine. 2013;368:107-116
  212. 212. Jin SC, Benitez BA, Karch CM, et al. Coding variants in TREM2 increase risk for Alzheimer’s disease. Human Molecular Genetics. 2014;23:5838-5846
  213. 213. Cheng-Hathaway PJ, Reed-Geaghan EG, Jay TR, et al. The Trem2 R47H variant confers loss-of-function-like phenotypes in Alzheimer’s disease. Molecular Neurodegeneration. 2018;13:29
  214. 214. Song WM, Joshita S, Zhou Y, et al. Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. The Journal of Experimental Medicine. 2018;215:745-760
  215. 215. Tang F, Barbacioru C, Wang Y, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nature Methods. 2009;6:377-382
  216. 216. Mathys H, Adaikkan C, Gao F, et al. Temporal tracking of microglia activation in neurodegeneration at single-cell resolution. Cell Reports. 2017;21:366-380
  217. 217. Friedman BA, Srinivasan K, Ayalon G, et al. Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Reports. 2018;22:832-847
  218. 218. Sala Frigerio C, Wolfs L, Fattorelli N, et al. The major risk factors for Alzheimer’s disease: Age, sex, and genes modulate the microglia response to aβ plaques. Cell Reports. 2019;27:1293-1306.e6
  219. 219. Rexach JE, Polioudakis D, Yin A, et al. Tau pathology drives dementia risk-associated gene networks toward chronic inflammatory states and immunosuppression. Cell Reports. 2020;33:108398
  220. 220. Lee S-H, Meilandt WJ, Xie L, et al. Trem2 restrains the enhancement of tau accumulation and neurodegeneration by β-amyloid pathology. Neuron. 2021;109:1283-1301.e6
  221. 221. Mancuso R, Fattorelli N, Martinez-Muriana A, et al. Xenografted human microglia display diverse transcriptomic states in response to Alzheimer’s disease-related amyloid-β pathology. Nature Neuroscience. 2024;27:886-900
  222. 222. Keren-Shaul H, Spinrad A, Weiner A, et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169:1276-1290.e17
  223. 223. Deczkowska A, Keren-Shaul H, Weiner A, et al. Disease-associated microglia: A universal immune sensor of neurodegeneration. Cell. 2018;173:1073-1081
  224. 224. Prater KE, Green KJ, Mamde S, et al. Human microglia show unique transcriptional changes in Alzheimer’s disease. Nature Aging. 2023;3:894-907
  225. 225. Yamada T, Horisberger MA, Kawaguchi N, et al. Immunohistochemistry using antibodies to alpha-interferon and its induced protein, MxA, in Alzheimer’s and Parkinson’s disease brain tissues. Neuroscience Letters. 1994;181:61-64
  226. 226. Stopa EG, Tanis KQ , Miller MC, et al. Comparative transcriptomics of choroid plexus in Alzheimer’s disease, frontotemporal dementia and Huntington’s disease: Implications for CSF homeostasis. Fluids and Barriers of the CNS. 2018;15:18
  227. 227. Li QS, De Muynck L. Differentially expressed genes in Alzheimer’s disease highlighting the roles of microglia genes including OLR1 and astrocyte gene CDK2AP1. Brain, Behavior, and Immunity Health. 2021;13:100227
  228. 228. Roy ER, Wang B, Wan Y-W, et al. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease. The Journal of Clinical Investigation. 2020;130:1912-1930
  229. 229. Roy ER, Chiu G, Li S, et al. Concerted type I interferon signaling in microglia and neural cells promotes memory impairment associated with amyloid β plaques. Immunity. 2022;55:879-894.e6
  230. 230. Taylor JM, Minter MR, Newman AG, et al. Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer’s disease. Neurobiology of Aging. 2014;35:1012-1023
  231. 231. Mesquita SD, Ferreira AC, Gao F, et al. The choroid plexus transcriptome reveals changes in type I and II interferon responses in a mouse model of Alzheimer’s disease. Brain, Behavior, and Immunity. 2015;49:280-292
  232. 232. Minter MR, Moore Z, Zhang M, et al. Deletion of the type-1 interferon receptor in APPSWE/PS1ΔE9 mice preserves cognitive function and alters glial phenotype. Acta Neuropathologica Communications. 2016;4:72
  233. 233. Xue F, Tian J, Yu C, et al. Type I interferon response-related microglial Mef2c deregulation at the onset of Alzheimer’s pathology in 5 × FAD mice. Neurobiology of Disease. 2021;152:105272
  234. 234. Sayed FA, Kodama L, Fan L, et al. AD-linked R47H-TREM2 mutation induces disease-enhancing microglial states via AKT hyperactivation. Science Translational Medicine. 2021;13:eabe3947
  235. 235. Sanna PP, Cabrelle C, Kawamura T, et al. A history of repeated alcohol intoxication promotes cognitive impairment and gene expression signatures of disease progression in the 3xtg mouse model of Alzheimer’s disease. eNeuro. 2023;10. DOI: 10.1523/ENEURO.0456-22.2023
  236. 236. Shippy DC, Ulland TK. Genome-wide identification of murine interferon genes in microglial-mediated neuroinflammation in Alzheimer’s disease. Journal of Neuroimmunology. 2023;375:578031
  237. 237. Sanford SAI, Miller LVC, Vaysburd M, et al. The type-I interferon response potentiates seeded tau aggregation and exacerbates tau pathology. Alzheimer's and Dementia. 2023;20(2):1013-1025. DOI: 10.1002/alz.13493
  238. 238. Carling GK, Fan L, Foxe NR, et al. Alzheimer’s disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. BioRxiv. 2024. DOI: 10.1101/2024.01.24.577107
  239. 239. Pandey S, Shen K, Lee S-H, et al. Disease-associated oligodendrocyte responses across neurodegenerative diseases. Cell Reports. 2022;40:111189
  240. 240. Naguib S, Torres ER, Lopez-Lee C, et al. APOE3-R136S mutation confers resilience against tau pathology via cGAS-STING-IFN inhibition. BioRxiv. 2024. DOI: 10.1101/2024.04.25.591140
  241. 241. Korvatska O, Kiianitsa K, Ratushny A, et al. Triggering receptor expressed on myeloid cell 2 R47H exacerbates immune response in Alzheimer’s disease brain. Frontiers in Immunology. 2020;11:559342
  242. 242. Fancy N, Willumsen N, Tsartsalis S, et al. Mechanisms contributing to differential genetic risks for TREM2 R47H and R62H variants in Alzheimer’s disease. medRxiv. 2022. DOI: 10.1101/2022.07.12.22277509
  243. 243. Ginsberg SD, Crino PB, Lee VM-Y, et al. Sequestration of RNA in Alzheimer’s disease neurofibrillary tangles and senile plaques. Annals of Neurology. 1997;41:200-209
  244. 244. Ginsberg SD, Crino PB, Hemby SE, et al. Predominance of neuronal mRNAs in individual Alzheimer’s disease senile plaques. Annals of Neurology. 1999;45:174-181
  245. 245. Uchida Y, Takahashi H. Rapid detection of Abeta deposits in APP transgenic mice by Hoechst 33342. Neuroscience Letters. 2008;448:279-281
  246. 246. Pensalfini A, Albay R, Rasool S, et al. Intracellular amyloid and the neuronal origin of Alzheimer neuritic plaques. Neurobiology of Disease. 2014;71:53-61
  247. 247. Minter MR, Main BS, Brody KM, et al. Soluble amyloid triggers a myeloid differentiation factor 88 and interferon regulatory factor 7 dependent neuronal type-1 interferon response in vitro. Journal of Neuroinflammation. 2015;12:71
  248. 248. Xie X, Ma G, Li X, et al. Activation of innate immune cGAS-STING pathway contributes to Alzheimer’s pathogenesis in 5 × FAD mice. Nature Aging. 2023;3:202-212
  249. 249. Udeochu JC, Amin S, Huang Y, et al. Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nature Neuroscience. 2023;26:737-750
  250. 250. Gavin AL, Huang D, Huber C, et al. PLD3 and PLD4 are single-stranded acid exonucleases that regulate endosomal nucleic-acid sensing. Nature Immunology. 2018;19:942-953
  251. 251. Gavin AL, Huang D, Blane TR, et al. Cleavage of DNA and RNA by PLD3 and PLD4 limits autoinflammatory triggering by multiple sensors. Nature Communications. 2021;12:5874
  252. 252. Van Acker ZP, Perdok A, Hellemans R, et al. Phospholipase D3 degrades mitochondrial DNA to regulate nucleotide signaling and APP metabolism. Nature Communications. 2023;14:2847
  253. 253. Cruchaga C, Karch CM, Jin SC, et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer’s disease. Nature. 2014;505:550-554
  254. 254. Zhang D-F, Fan Y, Wang D, et al. PLD3 in Alzheimer’s disease: A modest effect as revealed by updated association and expression analyses. Molecular Neurobiology. 2016;53:4034-4045
  255. 255. Zhang W, Jiao B, Xiao T, et al. Targeted sequencing on neurodegenerative genes identified novel causal and risk variants of familial Alzheimer’s disease. 2020. DOI: 10.21203/rs.3.rs-24070/v1
  256. 256. Wang J, Yu J-T, Tan L. PLD3 in Alzheimer’s disease. Molecular Neurobiology. 2015;51:480-486
  257. 257. Satoh J-I, Kino Y, Yamamoto Y, et al. PLD3 is accumulated on neuritic plaques in Alzheimer’s disease brains. Alzheimer’s Research & Therapy. 2014;6:70
  258. 258. Sadler AJ, BRG W. Interferon-inducible antiviral effectors. Nature Reviews. Immunology. 2008;8:559-568
  259. 259. Lee W-B, Choi WY, Lee D-H, et al. OAS1 and OAS3 negatively regulate the expression of chemokines and interferon-responsive genes in human macrophages. BMB Reports. 2019;52:133-138
  260. 260. Salih DA, Bayram S, Guelfi S, et al. Genetic variability in response to amyloid beta deposition influences Alzheimer’s disease risk. Brain Communications. 2019;1:fcz022
  261. 261. Magusali N, Graham AC, Piers TM, et al. A genetic link between risk for Alzheimer’s disease and severe COVID-19 outcomes via the OAS1 gene. Brain. 2021;144:3727-3741
  262. 262. Moore Z, Mobilio F, Walker FR, et al. Abrogation of type-I interferon signaling alters the microglial response to Aβ1-42. Scientific Reports. 2020;10:3153
  263. 263. Dedoni S, Olianas MC, Onali P. Interferon-β induces apoptosis in human SH-SY5Y neuroblastoma cells through activation of JAK–STAT signaling and down-regulation of PI3K/Akt pathway. Journal of Neurochemistry. 2010;115:1421-1433
  264. 264. Dedoni S, Olianas MC, Ingianni A, et al. Type I interferons impair BDNF-induced cell signaling and neurotrophic activity in differentiated human SH-SY5Y neuroblastoma cells and mouse primary cortical neurons. Journal of Neurochemistry. 2012;122:58-71
  265. 265. Olianas MC, Dedoni S, Onali P. Protection from interferon-β-induced neuronal apoptosis through stimulation of muscarinic acetylcholine receptors coupled to ERK1/2 activation. British Journal of Pharmacology. 2016;173:2910-2928
  266. 266. Cox DJ, Field RH, Williams DG, et al. DNA sensors are expressed in astrocytes and microglia in vitro and are upregulated during gliosis in neurodegenerative disease. Glia. 2015;63:812-825
  267. 267. Baruch K, Deczkowska A, David E, et al. Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function. Science. 2014;346:89-93
  268. 268. Deczkowska A, Matcovitch-Natan O, Tsitsou-Kampeli A, et al. Mef2C restrains microglial inflammatory response and is lost in brain aging in an IFN-I-dependent manner. Nature Communications. 2017;8:717
  269. 269. Leifer D, Krainc D, Yu YT, et al. MEF2C, a MADS/MEF2-family transcription factor expressed in a laminar distribution in cerebral cortex. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:1546-1550
  270. 270. Li H, Radford JC, Ragusa MJ, et al. Transcription factor MEF2C influences neural stem/progenitor cell differentiation and maturation in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:9397-9402
  271. 271. Rashid AJ, Cole CJ, Josselyn SA. Emerging roles for MEF2 transcription factors in memory. Genes, Brain, and Behavior. 2014;13:118-125
  272. 272. Ruiz A, Heilmann S, Becker T, et al. Follow-up of loci from the international genomics of Alzheimer’s disease project identifies TRIP4 as a novel susceptibility gene. Translational Psychiatry. 2014;4:e358
  273. 273. Tang S-S, Wang H-F, Zhang W, et al. MEF2C rs190982 polymorphism with late-onset alzheimer’s disease in Han Chinese: A replication study and meta-analyses. Oncotarget. 2016;7:39136-39142
  274. 274. Sunderaraman P, Cosentino S, Schupf N, et al. MEF2C common genetic variation is associated with different aspects of cognition in non-hispanic white and Caribbean hispanic non-demented older adults. Frontiers in Genetics. 2021;12:642327
  275. 275. Hur J-Y, Frost GR, Wu X, et al. The innate immunity protein IFITM3 modulates γ-secretase in Alzheimer’s disease. Nature. 2020;586:735-740
  276. 276. Pyun J-M, Park YH, Hodges A, et al. Immunity gene IFITM3 variant: Relation to cognition and Alzheimer’s disease pathology. Alzheimer's & Dementia (Amsterdam, Netherlands). 2022;14:e12317
  277. 277. Jana A, Wang X, Leasure JW, et al. Increased type I interferon signaling and brain endothelial barrier dysfunction in an experimental model of Alzheimer’s disease. Scientific Reports. 2022;12:16488
  278. 278. Stone LA, Frank JA, Albert PS, et al. The effect of interferon-beta on blood–brain barrier disruptions demonstrated by contrast-enhanced magnetic resonance imaging in relapsing–remitting multiple sclerosis. Annals of Neurology. 1995;37:611-619
  279. 279. Rossi JL, Todd T, Daniels Z, et al. Interferon-stimulated gene 15 upregulation precedes the development of blood–brain barrier disruption and cerebral edema after traumatic brain injury in young mice. Journal of Neurotrauma. 2015;32:1101-1108
  280. 280. Mastorakos P, Russo MV, Zhou T, et al. Antimicrobial immunity impedes CNS vascular repair following brain injury. Nature Immunology. 2021;22:1280-1293
  281. 281. Pais TF, Ali H, Moreira da Silva J, et al. Brain endothelial STING1 activation by plasmodium-sequestered heme promotes cerebral malaria via type I IFN response. Proceedings of the National Academy of Sciences of the United States of America. 2022;119:e2206327119
  282. 282. Shafi AM, Végvári Á, Zubarev RA, et al. Brain endothelial cells exposure to malaria parasites links type I interferon signaling to antigen presentation, immunoproteasome activation, endothelium disruption, and cellular metabolism. Frontiers in Immunology. 2023;14:1149107
  283. 283. Tziortzouda P, Van Den Bosch L, Hirth F. Triad of TDP43 control in neurodegeneration: Autoregulation, localization and aggregation. Nature Reviews. Neuroscience. 2021;22:197-208
  284. 284. Bright F, Werry EL, Dobson- Stone C, et al. Neuroinflammation in frontotemporal dementia. Nature Reviews. Neurology. 2019;15:540-555
  285. 285. Grossman M, Seeley WW, Boxer AL, et al. Frontotemporal lobar degeneration. Nature Reviews. Disease Primers. 2023;9:40
  286. 286. Dunker W, Ye X, Zhao Y, et al. TDP-43 prevents endogenous RNAs from triggering a lethal RIG-I-dependent interferon response. Cell Reports. 2021;35:108976
  287. 287. Yu C-H, Davidson S, Harapas CR, et al. TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell. 2020;183:636-649.e18
  288. 288. Licht-Murava A, Meadows SM, Palaguachi F, et al. Astrocytic TDP-43 dysregulation impairs memory by modulating antiviral pathways and interferon-inducible chemokines. Science Advances. 2023;9:eade1282
  289. 289. Galimberti D, Schoonenboom N, Scheltens P, et al. Intrathecal chemokine synthesis in mild cognitive impairment and Alzheimer disease. Archives of Neurology. 2006;63:538-543
  290. 290. Xia MQ , Bacskai BJ, Knowles RB, et al. Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: In vitro ERK1/2 activation and role in Alzheimer’s disease. Journal of Neuroimmunology. 2000;108:227-235
  291. 291. Galimberti D, Bonsi R, Fenoglio C, et al. Inflammatory molecules in frontotemporal dementia: Cerebrospinal fluid signature of progranulin mutation carriers. Brain, Behavior, and Immunity. 2015;49:182-187
  292. 292. Davidson YS, Raby S, Foulds PG, et al. TDP-43 pathological changes in early onset familial and sporadic Alzheimer’s disease, late onset alzheimer’s disease and Down’s syndrome: Association with age, hippocampal sclerosis and clinical phenotype. Acta Neuropathologica. 2011;122:703-713
  293. 293. Buciuc M, Tosakulwong N, Machulda MM, et al. TAR DNA-binding protein 43 is associated with rate of memory, functional and global cognitive decline in the decade prior to death. Journal of Alzheimer’s Disease. 2021;80:683-693
  294. 294. Kaya T, Mattugini N, Liu L, et al. CD8+ T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nature Neuroscience. 2022;25:1446-1457
  295. 295. Jorfi M, Park J, Hall CK, et al. Infiltrating CD8+ T cells exacerbate Alzheimer’s disease pathology in a 3D human neuroimmune axis model. Nature Neuroscience. 2023;26:1489-1504
  296. 296. Fernando N, Gopalakrishnan J, Behensky A, et al. Single-cell multiomic analysis reveals the involvement of type I interferon-responsive CD8+ T cells in amyloid beta-associated memory loss. BioRxiv. 2023. DOI: 10.1101/2023.03.18.533293
  297. 297. Ferretti MT, Merlini M, Späni C, et al. T-cell brain infiltration and immature antigen-presenting cells in transgenic models of Alzheimer’s disease-like cerebral amyloidosis. Brain, Behavior, and Immunity. 2016;54:211-225
  298. 298. Gate D, Saligrama N, Leventhal O, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature. 2020;577:399-404
  299. 299. Unger MS, Li E, Scharnagl L, et al. CD8+ T-cells infiltrate Alzheimer’s disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain, Behavior, and Immunity. 2020;89:67-86
  300. 300. Chen X, Firulyova M, Manis M, et al. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature. 2023;615:668-677
  301. 301. Ballard C, Mobley W, Hardy J, et al. Dementia in down’s syndrome. Lancet Neurology. 2016;15:622-636
  302. 302. Bayen E, Possin KL, Chen Y, et al. Prevalence of aging, dementia, and multimorbidity in older adults with down syndrome. JAMA Neurology. 2018;75:1399-1406
  303. 303. Araya P, Waugh KA, Sullivan KD, et al. Trisomy 21 dysregulates T cell lineages toward an autoimmunity-prone state associated with interferon hyperactivity. Proceedings of the National Academy of Sciences of the United States of America. 2019;116:24231-24241
  304. 304. Waugh KA, Araya P, Pandey A, et al. Mass cytometry reveals global immune remodeling with multi-lineage hypersensitivity to type I interferon in down syndrome. Cell Reports. 2019;29:1893-1908.e4
  305. 305. Ahmed MM, Johnson NR, Boyd TD, et al. Innate immune system activation and neuroinflammation in down syndrome and neurodegeneration: Therapeutic targets or partners? Frontiers in Aging Neuroscience. 2021;13:718426
  306. 306. Kong X-F, Worley L, Rinchai D, et al. Three copies of four interferon receptor genes underlie a mild type I interferonopathy in down syndrome. Journal of Clinical Immunology. 2020;40:807-819
  307. 307. Galbraith MD, Rachubinski AL, Smith KP, et al. Multidimensional definition of the interferonopathy of down syndrome and its response to JAK inhibition. Science Advances. 2023;9:eadg6218
  308. 308. Wisniewski KE, French JH, Rosen JF, et al. Basal ganglia calcification (BGC) in Down’s syndrome (DS)–Another manifestation of premature aging. Annals of the New York Academy of Sciences. 1982;396:179-189
  309. 309. Takashima S, Becker LE. Basal ganglia calcification in Down’s syndrome. Journal of Neurology, Neurosurgery, and Psychiatry. 1985;48:61-64
  310. 310. Ieshima A, Kisa T, Yoshino K, et al. A morphometric CT study of Down’s syndrome showing small posterior fossa and calcification of basal ganglia. Neuroradiology. 1984;26:493-498
  311. 311. Sadana KS, Goraya JS. Intracranial calcification in down syndrome. Journal of Pediatric Neurosciences. 2018;13:120-121
  312. 312. Thoms L, Idowu A, Nehra A, et al. Significance of basal ganglia calcification in Down’s syndrome. Advances in Mental Health and Intellectual Disabilities. 2020;14:103-110
  313. 313. Panda PK, Elwadhi A, Sharawat IK. Intracranial calcification and seizures in Down syndrome. BML Case Reports. 2021;14. DOI: 10.1136/bcr-2021-243180
  314. 314. Almudhry M, Prasad C, Tay K, et al. Progressive neurological decline associated with intracranial calcification in down syndrome; Fahr disease Mimic? The Canadian Journal of Neurological Sciences. 2023:1-2
  315. 315. Horvath S, Garagnani P, Bacalini MG, et al. Accelerated epigenetic aging in Down syndrome. Aging Cell. 2015;14:491-495
  316. 316. Horvath S. DNA methylation age of human tissues and cell types. Genome Biology. 2013;14:R115
  317. 317. Chen BH, Marioni RE, Colicino E, et al. DNA methylation-based measures of biological age: Meta-analysis predicting time to death. Aging (Albany NY). 2016;8:1844-1865
  318. 318. Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY). 2019;11:303-327
  319. 319. Gensous N, Bacalini MG, Franceschi C, et al. Down syndrome, accelerated aging and immunosenescence. Seminars in Immunopathology. 2020;42:635-645
  320. 320. Snyder HM, Bain LJ, Brickman AM, et al. Further understanding the connection between Alzheimer’s disease and Down syndrome. Alzheimers Dement. 2020;16:1065-1077
  321. 321. Sullivan KD, Lewis HC, Hill AA, et al. Trisomy 21 consistently activates the interferon response. eLife. 2016;5. DOI: 10.7554/eLife.16220
  322. 322. Malle L, Martin-Fernandez M, Buta S, et al. Excessive negative regulation of type I interferon disrupts viral control in individuals with Down syndrome. Immunity. 2022;55:2074-2084.e5
  323. 323. Waugh KA, Minter R, Baxter J, et al. Triplication of the interferon receptor locus contributes to hallmarks of Down syndrome in a mouse model. Nature Genetics. 2023;55:1034-1047
  324. 324. Krivega M, Stiefel CM, Karbassi S, et al. Genotoxic stress in constitutive trisomies induces autophagy and the innate immune response via the cGAS-STING pathway. Communications Biology. 2021;4:831
  325. 325. Maroun LE. Anti-interferon immunoglobulins can improve the trisomy 16 mouse phenotype. Teratology. 1995;51:329-335
  326. 326. Gupta M, Dhanasekaran AR, Gardiner KJ. Mouse models of Down syndrome: Gene content and consequences. Mammalian Genome. 2016;27:538-555
  327. 327. Franceschi C, Bonafè M, Valensin S, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Annals of the New York Academy of Sciences. 2000;908:244-254
  328. 328. López-Otín C, Blasco MA, Partridge L, et al. Hallmarks of aging: An expanding universe. Cell. 2023;186:243-278
  329. 329. MacAulay N, Keep RF, Zeuthen T. Cerebrospinal fluid production by the choroid plexus: A century of barrier research revisited. Fluids and Barriers of the CNS. 2022;19:26
  330. 330. Boldrini M, Canoll PD, Klein RS. How COVID-19 affects the brain. JAMA Psychiatry. 2021;78:682-683
  331. 331. Asadi-Pooya AA, Akbari A, Emami A, et al. Long COVID syndrome-associated brain fog. Journal of Medical Virology. 2022;94:979-984
  332. 332. Xu Z, Wang H, Jiang S, et al. Brain pathology in COVID-19: Clinical manifestations and potential mechanisms. Neuroscience Bulletin. 2024;40:383-400
  333. 333. Wulf Hanson S, Abbafati C, et al. Estimated global proportions of individuals with persistent fatigue, cognitive, and respiratory symptom clusters following symptomatic COVID-19 in 2020 and 2021. JAMA. 2022;328:1604-1615
  334. 334. Suzzi S, Tsitsou-Kampeli A, Schwartz M. The type I interferon antiviral response in the choroid plexus and the cognitive risk in COVID-19. Nature Immunology. 2023;24:220-224
  335. 335. Yang AC, Kern F, Losada PM, et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature. 2021;595:565-571
  336. 336. Čarna M, Onyango IG, Katina S, et al. Pathogenesis of Alzheimer’s disease: Involvement of the choroid plexus. Alzheimers Dement. 2023;19:3537-3554
  337. 337. West AP, Shadel GS. Mitochondrial DNA in innate immune responses and inflammatory pathology. Nature Reviews. Immunology. 2017;17:363-375
  338. 338. Jauhari A, Baranov SV, Suofu Y, et al. Melatonin inhibits cytosolic mitochondrial DNA–induced neuroinflammatory signaling in accelerated aging and neurodegeneration. The Journal of Clinical Investigation. 2020;130(6):3124-3136
  339. 339. Lei Y, Guerra Martinez C, Torres-Odio S, et al. Elevated type I interferon responses potentiate metabolic dysfunction, inflammation, and accelerated aging in mtDNA mutator mice. Science Advances. 2021;7. DOI: 10.1126/sciadv.abe7548
  340. 340. Jiménez-Loygorri JI, Villarejo- Zori B, Viedma-Poyatos Á, et al. Mitophagy curtails cytosolic mtDNA-dependent activation of cGAS/STING inflammation during aging. Nature Communications. 2024;15:830
  341. 341. Ansar M, Qu Y, Ivanciuc T, et al. Lack of type I interferon signaling ameliorates respiratory syncytial virus-induced lung inflammation and restores antioxidant defenses. Antioxidants (Basel). 2021;11. DOI: 10.3390/antiox11010067
  342. 342. Wang X, Cui L, Ji X. Cognitive impairment caused by hypoxia: From clinical evidences to molecular mechanisms. Metabolic Brain Disease. 2022;37:51-66
  343. 343. Huang W, Hickson LJ, Eirin A, et al. Cellular senescence: The good, the bad and the unknown. Nature Reviews. Nephrology. 2022;18:611-627
  344. 344. Buckley MT, Sun ED, George BM, et al. Cell-type-specific aging clocks to quantify aging and rejuvenation in neurogenic regions of the brain. Nature Aging. 2023;3:121-137
  345. 345. De Cecco M, Criscione SW, Peckham EJ, et al. Genomes of replicatively senescent cells undergo global epigenetic changes leading to gene silencing and activation of transposable elements. Aging Cell. 2013;12:247-256
  346. 346. Muotri AR, Chu VT, Marchetto MCN, et al. Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature. 2005;435:903-910
  347. 347. Erwin JA, Marchetto MC, Gage FH. Mobile DNA elements in the generation of diversity and complexity in the brain. Nature Reviews. Neuroscience. 2014;15:497-506
  348. 348. Takahashi A, Loo TM, Okada R, et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nature Communications. 2018;9:1249
  349. 349. De Cecco M, Ito T, Petrashen AP, et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature. 2019;566:73-78
  350. 350. Gulen MF, Samson N, Keller A, et al. cGAS-STING drives aging-related inflammation and neurodegeneration. Nature. 2023;620:374-380
  351. 351. DeMaeyer EM, De Maeyer-Guignard J. Interferons and Other Regulatory Cytokines. illustrated ed. New York City, United States: Wiley; 1988
  352. 352. Jin M, Xu R, Wang L, et al. Type-I-interferon signaling drives microglial dysfunction and senescence in human iPSC models of Down syndrome and Alzheimer’s disease. Cell Stem Cell. 2022;29:1135-1153.e8
  353. 353. Yu Q , Katlinskaya YV, Carbone CJ, et al. DNA-damage-induced type I interferon promotes senescence and inhibits stem cell function. Cell Reports. 2015;11:785-797
  354. 354. Lau V, Ramer L, Tremblay M-È. An aging, pathology burden, and glial senescence build-up hypothesis for late onset alzheimer’s disease. Nature Communications. 2023;14:1670
  355. 355. Ng PY, Zhang C, Li H, et al. Senescent microglia represent a subset of disease-associated microglia in P301S mice. Journal of Alzheimer’s Disease. 2023;95:493-507
  356. 356. Rim C, You M-J, Nahm M, et al. Emerging role of senescent microglia in brain aging-related neurodegenerative diseases. Translational Neurodegeneration. 2024;13:10
  357. 357. Harrington A, Raissi AJ, Rajkovich K, et al. MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders. eLife. 2016;25(5)
  358. 358. Zhang L, Chen D, Song D, et al. Clinical and translational values of spatial transcriptomics. Signal Transduction and Targeted Therapy. 2022;7:111
  359. 359. Villeda SA, Luo J, Mosher KI, et al. The aging systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477:90-94
  360. 360. Bieri G, Schroer AB, Villeda SA. Blood-to-brain communication in aging and rejuvenation. Nature Neuroscience. 2023;26:379-393
  361. 361. D’Mello C, Le T, Swain MG. Cerebral microglia recruit monocytes into the brain in response to tumor necrosis factoralpha signaling during peripheral organ inflammation. The Journal of Neuroscience. 2009;29:2089-2102
  362. 362. Somebang K, Rudolph J, Imhof I, et al. CCR2 deficiency alters activation of microglia subsets in traumatic brain injury. Cell Reports. 2021;36:109727
  363. 363. Atkins CM. Decoding hippocampal signaling deficits after traumatic brain injury. Translational Stroke Research. 2011;2:546-555
  364. 364. Smith DH, Johnson VE, Stewart W. Chronic neuropathologies of single and repetitive TBI: Substrates of dementia? Nature Reviews. Neurology. 2013;9:211-221
  365. 365. Li Y, Li Y, Li X, et al. Head injury as a risk factor for dementia and Alzheimer’s disease: A systematic review and meta-analysis of 32 observational studies. PLoS One. 2017;12:e0169650
  366. 366. Fann JR, Ribe AR, Pedersen HS, et al. Long-term risk of dementia among people with traumatic brain injury in Denmark: A population-based observational cohort study. Lancet Psychiatry. 2018;5:424-431
  367. 367. Gardner RC, Burke JF, Nettiksimmons J, et al. Dementia risk after traumatic brain injury vs. nonbrain trauma: The role of age and severity. JAMA Neurology. 2014;71:1490-1497
  368. 368. Gardner RC, Burke JF, Nettiksimmons J, et al. Traumatic brain injury in later life increases risk for Parkinson disease. Annals of Neurology. 2015;77:987-995
  369. 369. Herklots MW, Kroon M, Roks G, et al. Poor outcome in frail elderly patient after severe TBI. Brain Injury. 2022;36:1118-1122
  370. 370. Albrecht JS. Impact of dementia on days at home after traumatic brain injury among older medicare beneficiaries. Alzheimer’s Dement. 2023;19. DOI: 10.1002/alz.075534
  371. 371. Shively S, Scher AI, Perl DP, et al. Dementia resulting from traumatic brain injury: What is the pathology? Archives of Neurology. 2012;69:1245-1251
  372. 372. Kuo YG, Tarzi FP, Louie S, et al. Neuroinflammation in traumatic brain injury. In: Lv X, Guo Y, Mao G, editors. Frontiers in traumatic Brain Injury. IntechOpen; 2022. Epub ahead of print November 30, 2022. DOI: 10.5772/intechopen.105178
  373. 373. Shao F, Wang X, Wu H, et al. Microglia and neuroinflammation: Crucial pathological mechanisms in traumatic brain injury-induced neurodegeneration. Frontiers in Aging Neuroscience. 2022;14:825086
  374. 374. Todd BP, Chimenti MS, Luo Z, et al. Traumatic brain injury results in unique microglial and astrocyte transcriptomes enriched for type I interferon response. Journal of Neuroinflammation. 2021;18:151
  375. 375. Hammond TR, Dufort C, Dissing-Olesen L, et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity. 2019;50:253-271.e6
  376. 376. Witcher KG, Bray CE, Chunchai T, et al. Traumatic brain injury causes chronic cortical inflammation and neuronal dysfunction mediated by microglia. The Journal of Neuroscience. 2021;41:1597-1616
  377. 377. Garza R, Sharma Y, Atacho DAM, et al. Single-cell transcriptomics of human traumatic brain injury reveals activation of endogenous retroviruses in oligodendroglia. Cell Reports. 2023;42:113395
  378. 378. Bolte AC, Shapiro DA, Dutta AB, et al. The meningeal transcriptional response to traumatic brain injury and aging. eLife. 2023;12. DOI: 10.7554/eLife.81154
  379. 379. Packer JM, Bray CE, Beckman NB, et al. Impaired cortical neuronal homeostasis and cognition after diffuse traumatic brain injury are dependent on microglia and type I interferon responses. Glia. 2024;72:300-321
  380. 380. Wangler LM, Bray CE, Packer JM, et al. Amplified gliosis and interferon-associated inflammation in the aging brain following diffuse traumatic brain injury. The Journal of Neuroscience. 2022;42:9082-9096
  381. 381. Barrett JP, Knoblach SM, Bhattacharya S, et al. Traumatic brain injury induces cGAS activation and type I interferon signaling in aged mice. Frontiers in Immunology. 2021;12:710608
  382. 382. Todd BP, Luo Z, Gilkes N, et al. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury. Acta Neuropathologica Communications. 2023;11:134
  383. 383. Karve IP, Zhang M, Habgood M, et al. Ablation of type-1 IFN signaling in hematopoietic cells confers protection following traumatic brain injury. eNeuro. 2016;3. DOI: 10.1523/ENEURO.0128-15.2016
  384. 384. Barrett JP, Henry RJ, Shirey KA, et al. Interferon-β plays a detrimental role in experimental traumatic brain injury by enhancing neuroinflammation that drives chronic neurodegeneration. The Journal of Neuroscience. 2020;40:2357-2370
  385. 385. Abdullah A, Zhang M, Frugier T, et al. STING-mediated type-I interferons contribute to the neuroinflammatory process and detrimental effects following traumatic brain injury. Journal of Neuroinflammation. 2018;15:323
  386. 386. Fritsch LE, Ju J, Gudenschwager Basso EK, et al. Type I interferon response is mediated by NLRX1-cGAS-STING signaling in brain injury. Frontiers in Molecular Neuroscience. 2022;15:852243
  387. 387. Fritsch LE, Kelly C, Leonard J, et al. STING-dependent signaling in microglia or peripheral immune cells orchestrates the early inflammatory response and influences brain injury outcome. The Journal of Neuroscience. 2024;44. DOI: 10.1523/JNEUROSCI.0191-23.2024
  388. 388. Fryer AL, Abdullah A, Mobilio F, et al. Pharmacological inhibition of STING reduces neuroinflammation-mediated damage post-traumatic brain injury. British Journal of Pharmacology. 2024. pp. 1-18. DOI: 10.1111/bph.16347
  389. 389. Saleh A, Macia A, Muotri AR. Transposable elements, inflammation, and neurological disease. Frontiers in Neurology. 2019;10:894
  390. 390. Vaibhav K, Braun M, Alverson K, et al. Neutrophil extracellular traps exacerbate neurological deficits after traumatic brain injury. Science Advances. 2020;6:eaax8847
  391. 391. Mi L, Min X, Shi M, et al. Neutrophil extracellular traps aggravate neuronal endoplasmic reticulum stress and apoptosis via TLR9 after traumatic brain injury. Cell Death & Disease. 2023;14:374
  392. 392. Walko TD, Bola RA, Hong JD, et al. Cerebrospinal fluid mitochondrial DNA: A novel DAMP in pediatric traumatic brain injury. Shock. 2014;41:499-503
  393. 393. Rodrigues Filho EM, Simon D, Ikuta N, et al. Elevated cell-free plasma DNA level as an independent predictor of mortality in patients with severe traumatic brain injury. Journal of Neurotrauma. 2014;31:1639-1646
  394. 394. Ritzel RM, Li Y, Lei Z, et al. Functional and transcriptional profiling of microglial activation during the chronic phase of TBI identifies an age-related driver of poor outcome in old mice. Geroscience. 2022;44:1407-1440
  395. 395. Sen T, Saha P, Gupta R, et al. Aberrant ER stress induced neuronal-IFNβ elicits white matter injury due to microglial activation and T-cell infiltration after TBI. The Journal of Neuroscience. 2020;40:424-446
  396. 396. Dohi K, Ohtaki H, Nakamachi T, et al. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. Journal of Neuroinflammation. 2010;7:41
  397. 397. Zhang Q-G, Laird MD, Han D, et al. Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One. 2012;7:e34504
  398. 398. Kumar A, Barrett JP, Alvarez-Croda D-M, et al. NOX2 drives M1-like microglial/macrophage activation and neurodegeneration following experimental traumatic brain injury. Brain, Behavior, and Immunity. 2016;58:291-309
  399. 399. Glennon-Alty L, Moots RJ, Edwards SW, et al. Type I interferon regulates cytokine-delayed neutrophil apoptosis, reactive oxygen species production and chemokine expression. Clinical and Experimental Immunology. 2021;203:151-159
  400. 400. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nature Immunology. 2018;19:327-341
  401. 401. Kwidzinski E, Bechmann I. IDO expression in the brain: A double-edged sword. Journal of Molecular Medicine. 2007;85:1351-1359
  402. 402. Berger EA, Murphy PM, Farber JM. Chemokine receptors as HIV-1 coreceptors: Roles in viral entry, tropism, and disease. Annual Review of Immunology. 1999;17:657-700
  403. 403. Bekker L-G, Beyrer C, Mgodi N, et al. HIV infection. Nature Reviews. Disease Primers. 2023;9:42
  404. 404. Day C, Manning K, Abdullah F, et al. Delirium in HIV-infected patients admitted to acute medical wards post universal access to antiretrovirals in South Africa. South African Medical Journal. 2021;111:974-980
  405. 405. Tang Y, Chaillon A, Gianella S, et al. Brain microglia serve as a persistent HIV reservoir despite durable antiretroviral therapy. The Journal of Clinical Investigation. 2023;133. DOI: 10.1172/JCI167417
  406. 406. Luo X, He JJ. Cell–cell contact viral transfer contributes to HIV infection and persistence in astrocytes. Journal of Neurovirology. 2015;21:66-80
  407. 407. Wahl A, Al-Harthi L. HIV infection of non-classical cells in the brain. Retrovirology. 2023;20:1
  408. 408. Bandera A, Taramasso L, Bozzi G, et al. HIV-associated neurocognitive impairment in the modern ART era: Are we close to discovering reliable biomarkers in the setting of virological suppression? Frontiers in Aging Neuroscience. 2019;11:187
  409. 409. Mastrorosa I, Pinnetti C, Brita AC, et al. Declining prevalence of human immunodeficiency virus (HIV)-associated neurocognitive disorders in recent years and associated factors in a large cohort of antiretroviral therapy-treated individuals with HIV. Clinical Infectious Diseases. 2023;76:e629-e637
  410. 410. Borjabad A, Morgello S, Chao W, et al. Significant effects of antiretroviral therapy on global gene expression in brain tissues of patients with HIV-1-associated neurocognitive disorders. PLoS Pathogens. 2011;7:e1002213
  411. 411. Shityakov S, Dandekar T, Förster C. Gene expression profiles and protein–protein interaction network analysis in AIDS patients with HIV-associated encephalitis and dementia. HIV/AIDS (Auckland, N.Z.). 2015;7:265-276
  412. 412. Solomon IH, De Girolami U, Chettimada S, et al. Brain and liver pathology, amyloid deposition, and interferon responses among older HIV-positive patients in the late HAART era. BMC Infectious Diseases. 2017;17:151
  413. 413. Sanfilippo C, Pinzone MR, Cambria D, et al. OAS gene family expression is associated with HIV-related neurocognitive disorders. Molecular Neurobiology. 2018;55:1905-1914
  414. 414. Garces A, Martinez B, De La Garza R, et al. Differential expression of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) in Alzheimer’s disease and HIV-1 associated neurocognitive disorders. Scientific Reports. 2023;13:3276
  415. 415. Tavazzi E, Morrison D, Sullivan P, et al. Brain inflammation is a common feature of HIV-infected patients without HIV encephalitis or productive brain infection. Current HIV Research. 2014;12:97-110
  416. 416. Akwa Y, Hassett DE, Eloranta ML, et al. Transgenic expression of IFN-alpha in the central nervous system of mice protects against lethal neurotropic viral infection but induces inflammation and neurodegeneration. Journal of Immunology. 1998;161:5016-5026
  417. 417. Sas AR, Bimonte-Nelson H, Smothers CT, et al. Interferon-alpha causes neuronal dysfunction in encephalitis. The Journal of Neuroscience. 2009;29:3948-3955
  418. 418. Singh H, Ojeda-Juárez D, Maung R, et al. A pivotal role for Interferon-α receptor-1 in neuronal injury induced by HIV-1. Journal of Neuroinflammation. 2020;17:226
  419. 419. Mandl JN, Barry AP, Vanderford TH, et al. Divergent TLR7 and TLR9 signaling and type I interferon production distinguish pathogenic and nonpathogenic AIDS virus infections. Nature Medicine. 2008;14:1077-1087
  420. 420. Jakobsen MR, Bak RO, Andersen A, et al. IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:E4571-E4580
  421. 421. Wang MQ , Huang YL, Huang J, et al. RIG-I detects HIV-1 infection and mediates type I interferon response in human macrophages from patients with HIV-1-associated neurocognitive disorders. Genetics and Molecular Research. 2015;14:13799-13811
  422. 422. Rho MB, Wesselingh S, Glass JD, et al. A potential role for interferon-alpha in the pathogenesis of HIV-associated dementia. Brain, Behavior, and Immunity. 1995;9:366-377
  423. 423. Krivine A, Force G, Servan J, et al. Measuring HIV-1 RNA and interferon-alpha in the cerebrospinal fluid of AIDS patients: Insights into the pathogenesis of AIDS Dementia Complex. Journal of Neurovirology. 1999;5:500-506
  424. 424. Perrella O, Carreiri PB, Perrella A, et al. Transforming growth factor beta-1 and interferon-alpha in the AIDS dementia complex (ADC): Possible relationship with cerebral viral load? European Cytokine Network. 2001;12:51-55
  425. 425. Anderson AM, Lennox JL, Mulligan MM, et al. Cerebrospinal fluid interferon alpha levels correlate with neurocognitive impairment in ambulatory HIV-Infected individuals. Journal of Neurovirology. 2017;23:106-112
  426. 426. Kong W, Frouard J, Xie G, et al. Neuroinflammation generated by HIV-infected microglia promotes dysfunction and death of neurons in human brain organoids. PNAS Nexus. 2024;3:pgae179
  427. 427. Lamers SL, Rose R, Maidji E, et al. HIV DNA is frequently present within pathologic tissues evaluated at autopsy from combined antiretroviral therapy-treated patients with undetectable viral loads. Journal of Virology. 2016;90:8968-8983
  428. 428. Osborne O, Peyravian N, Nair M, et al. The paradox of HIV blood–brain barrier penetrance and antiretroviral drug delivery deficiencies. Trends in Neurosciences. 2020;43:695-708
  429. 429. Ene L, Duiculescu D, Ruta SM. How much do antiretroviral drugs penetrate into the central nervous system? Journal of Medicine and Life. 2011;4:432-439
  430. 430. Helms J, Kremer S, Merdji H, et al. Neurologic features in severe SARS-CoV-2 Infection. The New England Journal of Medicine. 2020;382:2268-2270
  431. 431. Mao L, Jin H, Wang M, et al. Neurologic manifestations of hospitalized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurology. 2020. pp. 683-690. DOI: 10.1001/jamaneurol.2020.1127
  432. 432. Hampshire A, Trender W, Chamberlain SR, et al. Cognitive deficits in people who have recovered from COVID-19. EClinicalMedicine. 2021;39:101044
  433. 433. Liu Y-H, Chen Y, Wang Q-H, et al. One-year trajectory of cognitive changes in older survivors of COVID-19 in Wuhan, China: A longitudinal cohort study. JAMA Neurology. 2022;79:509-517
  434. 434. Pellegrini L, Albecka A, Mallery DL, et al. SARS-CoV-2 infects the brain choroid plexus and disrupts the blood-CSF barrier in human brain organoids. Cell Stem Cell. 2020;27:951-961.e5
  435. 435. van Heteren JT, Rozenberg F, Aronica E, et al. Astrocytes produce interferon-alpha and CXCL10, but not IL-6 or CXCL8, in Aicardi-Goutières syndrome. Glia. 2008;56:568-578
  436. 436. Cuadrado E, Jansen MH, Anink J, et al. Chronic exposure of astrocytes to interferon-α reveals molecular changes related to Aicardi-Goutieres syndrome. Brain. 2013;136:245-258
  437. 437. Sase S, Takanohashi A, Vanderver A, et al. Astrocytes, an active player in Aicardi-Goutières syndrome. Brain Pathology. 2018;28:399-407
  438. 438. Cuadrado E, Michailidou I, van Bodegraven EJ, et al. Phenotypic variation in Aicardi-Goutières syndrome explained by cell-specific IFN-stimulated gene response and cytokine release. Journal of Immunology. 2015;194:3623-3633
  439. 439. Giordano AMS, Luciani M, Gatto F, et al. DNA damage contributes to neurotoxic inflammation in Aicardi-Goutières syndrome astrocytes. The Journal of Experimental Medicine. 2022;219. DOI: 10.1084/jem.20211121
  440. 440. Jin M, Shiwaku H, Tanaka H, et al. Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nature Communications. 2021;12:6565
  441. 441. Préhaud C, Mégret F, Lafage M, et al. Virus infection switches TLR-3-positive human neurons to become strong producers of beta interferon. Journal of Virology. 2005;79:12893-12904
  442. 442. Lafaille FG, Pessach IM, Zhang S-Y, et al. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature. 2012;491:769-773
  443. 443. Tang S-C, Arumugam TV, Xu X, et al. Pivotal role for neuronal toll-like receptors in ischemic brain injury and functional deficits. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:13798-13803
  444. 444. Jovasevic V, Wood EM, Cicvaric A, et al. Formation of memory assemblies through the DNA-sensing TLR9 pathway. Nature. 2024;628:145-153
  445. 445. Ma Y, Haynes RL, Sidman RL, et al. TLR8: An innate immune receptor in brain, neurons and axons. Cell Cycle. 2007;6:2859-2868
  446. 446. Liu T, Xu Z-Z, Park C-K, et al. Toll-like receptor 7 mediates pruritus. Nature Neuroscience. 2010;13:1460-1462
  447. 447. Lehmann SM, Krüger C, Park B, et al. An unconventional role for miRNA: Let-7 activates toll-like receptor 7 and causes neurodegeneration. Nature Neuroscience. 2012;15:827-835
  448. 448. Nazmi A, Dutta K, Basu A. RIG-I mediates innate immune response in mouse neurons following Japanese encephalitis virus infection. PLoS One. 2011;6:e21761
  449. 449. Liu Y, Jesus AA, Marrero B, et al. Activated STING in a vascular and pulmonary syndrome. The New England Journal of Medicine. 2014;371:507-518
  450. 450. Chaudhuri A, Duan F, Morsey B, et al. HIV-1 activates proinflammatory and interferon-inducible genes in human brain microvascular endothelial cells: Putative mechanisms of blood–brain barrier dysfunction. Journal of Cerebral Blood Flow and Metabolism. 2008;28:697-711
  451. 451. Kang L, Yu H, Yang X, et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nature Communications. 2020;11:2488
  452. 452. Ma B, Dela Cruz CS, Hartl D, et al. RIG-like helicase innate immunity inhibits vascular endothelial growth factor tissue responses via a type I IFN-dependent mechanism. American Journal of Respiratory and Critical Care Medicine. 2011;183:1322-1335
  453. 453. Sprokholt JK, Kaptein TM, van Hamme JL, et al. RIG-I-like receptor triggering by dengue virus drives dendritic cell immune activation and TH1 differentiation. Journal of Immunology. 2017;198:4764-4771
  454. 454. Venkatesh D, Ernandez T, Rosetti F, et al. Endothelial TNF receptor 2 induces IRF1 transcription factor-dependent interferon-β autocrine signaling to promote monocyte recruitment. Immunity. 2013;38:1025-1037
  455. 455. Kim H-J, Kim H, Lee J-H, et al. Toll-like receptor 4 (TLR4): New insight immune and aging. Immunity & Ageing. 2023;20:67
  456. 456. Colleselli K, Stierschneider A, Wiesner C. An update on Toll-like receptor 2, its function and dimerization in pro- and anti-inflammatory processes. International Journal of Molecular Sciences. 2023;24. DOI: 10.3390/ijms241512464
  457. 457. Pan W, Banks WA, Kastin AJ. Permeability of the blood–brain and blood-spinal cord barriers to interferons. Journal of Neuroimmunology. 1997;76:105-111
  458. 458. Wang J, Campbell IL, Zhang H. Systemic interferon-alpha regulates interferon-stimulated genes in the central nervous system. Molecular Psychiatry. 2008;13:293-301
  459. 459. Aw E, Zhang Y, Carroll M. Microglial responses to peripheral type 1 interferon. Journal of Neuroinflammation. 2020;17:340
  460. 460. Felger JC. Role of inflammation in depression and treatment implications. Handbook of Experimental Pharmacology. 2019;250:255-286
  461. 461. Torres-Platas SG, Cruceanu C, Chen GG, et al. Evidence for increased microglial priming and macrophage recruitment in the dorsal anterior cingulate white matter of depressed suicides. Brain, Behavior, and Immunity. 2014;42:50-59
  462. 462. Curzytek K, Leśkiewicz M. Targeting the CCL2-CCR2 axis in depressive disorders. Pharmacological Reports. 2021;73:1052-1062
  463. 463. Blomberg S, Eloranta M-L, Magnusson M, et al. Expression of the markers BDCA-2 and BDCA-4 and production of interferon-alpha by plasmacytoid dendritic cells in systemic lupus erythematosus. Arthritis and Rheumatism. 2003;48:2524-2532
  464. 464. Rönnblom L, Pascual V. The innate immune system in SLE: Type I interferons and dendritic cells. Lupus. 2008;17:394-399
  465. 465. Lindau D, Mussard J, Rabsteyn A, et al. TLR9 independent interferon α production by neutrophils on NETosis in response to circulating chromatin, a key lupus autoantigen. Annals of the Rheumatic Diseases. 2014;73:2199-2207
  466. 466. Nehar-Belaid D, Hong S, Marches R, et al. Mapping systemic lupus erythematosus heterogeneity at the single-cell level. Nature Immunology. 2020;21:1094-1106
  467. 467. Karnell JL, Wu Y, Mittereder N, et al. Depleting plasmacytoid dendritic cells reduces local type I interferon responses and disease activity in patients with cutaneous lupus. Science Translational Medicine. 2021;13. DOI: 10.1126/scitranslmed.abf8442
  468. 468. West PK, McCorkindale AN, Guennewig B, et al. The cytokines interleukin-6 and interferon-α induce distinct microglia phenotypes. Journal of Neuroinflammation. 2022;19:96
  469. 469. Li W, Viengkhou B, Denyer G, et al. Microglia have a more extensive and divergent response to interferon-α compared with astrocytes. Glia. 2018;66:2058-2078
  470. 470. Qin H, Wilson CA, Lee SJ, et al. IFN-beta-induced SOCS-1 negatively regulates CD40 gene expression in macrophages and microglia. The FASEB Journal. 2006;20:985-987
  471. 471. Albini A, Marchisone C, Del Grosso F, et al. Inhibition of angiogenesis and vascular tumor growth by interferon-producing cells: A gene therapy approach. The American Journal of Pathology. 2000;156:1381-1393
  472. 472. Indraccolo S. Interferon-alpha as angiogenesis inhibitor: Learning from tumor models. Autoimmunity. 2010;43:244-247
  473. 473. Romagnani P, Annunziato F, Lasagni L, et al. Cell cycle-dependent expression of CXC chemokine receptor 3 by endothelial cells mediates angiostatic activity. The Journal of Clinical Investigation. 2001;107:53-63
  474. 474. Campanella GSV, Colvin RA, Luster AD. CXCL10 can inhibit endothelial cell proliferation independently of CXCR3. PLoS One. 2010;5:e12700
  475. 475. Jian D, Wang W, Zhou X, et al. Interferon-induced protein 35 inhibits endothelial cell proliferation, migration and re-endothelialization of injured arteries by inhibiting the nuclear factor-kappa B pathway. Acta Physiologica (Oxford, England). 2018;223:e13037
  476. 476. Kalucka J, de Rooij LPMH, Goveia J, et al. Single-cell transcriptome atlas of murine endothelial cells. Cell. 2020;180:764-779.e20
  477. 477. Floris S, Ruuls SR, Wierinckx A, et al. Interferon-beta directly influences monocyte infiltration into the central nervous system. Journal of Neuroimmunology. 2002;127:69-79
  478. 478. Buttmann M, Goebeler M, Toksoy A, et al. Subcutaneous interferon-beta injections in patients with multiple sclerosis initiate inflammatory skin reactions by local chemokine induction. Journal of Neuroimmunology. 2005;168:175-182
  479. 479. Campbell IL, Krucker T, Steffensen S, et al. Structural and functional neuropathology in transgenic mice with CNS expression of IFN-alpha. Brain Research. 1999;835:46-61
  480. 480. Wiley CA, Steinman RA, Wang Q. Innate immune activation without immune cell infiltration in brains of murine models of Aicardi-Goutières syndrome. Brain Pathology. 2023;33:e13118
  481. 481. Kettwig M, Ternka K, Wendland K, et al. Interferon-driven brain phenotype in a mouse model of RNaseT2 deficient leukoencephalopathy. Nature Communications. 2021;12:6530
  482. 482. Larochelle C, Grand’maison F, Bernier GP, et al. Thrombotic thrombocytopenic purpura-hemolytic uremic syndrome in relapsing–remitting multiple sclerosis patients on high-dose interferon β. Multiple Sclerosis. 2014;20:1783-1787
  483. 483. Popson SA, Ziegler ME, Chen X, et al. Interferon-induced transmembrane protein 1 regulates endothelial lumen formation during angiogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014;34:1011-1019
  484. 484. Herrmann JR, Simon DW. Interfer(on)-ing with vascular repair after acute brain injury. Nature Immunology. 2021;22:1205-1206
  485. 485. Weller RO, Boche D, Nicoll JAR. Microvasculature changes and cerebral amyloid angiopathy in Alzheimer’s disease and their potential impact on therapy. Acta Neuropathologica. 2009;118:87-102
  486. 486. Charidimou A, Boulouis G, Gurol ME, et al. Emerging concepts in sporadic cerebral amyloid angiopathy. Brain. 2017;140:1829-1850
  487. 487. Greenberg SM, Bacskai BJ, Hernandez-Guillamon M, et al. Cerebral amyloid angiopathy and Alzheimer disease - One peptide, two pathways. Nature Reviews. Neurology. 2020;16:30-42
  488. 488. Petersen MA, Ryu JK, Akassoglou K. Fibrinogen in neurological diseases: Mechanisms, imaging and therapeutics. Nature Reviews. Neuroscience. 2018;19:283-301
  489. 489. Merlini M, Rafalski VA, Rios Coronado PE, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an Alzheimer’s disease model. Neuron. 2019;101:1099-1108.e6
  490. 490. Mendiola AS, Yan Z, Dixit K, et al. Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nature Immunology. 2023;24:1173-1187
  491. 491. Klok MD, Bakels HS, Postma NL, et al. Interferon-α and the calcifying microangiopathy in Aicardi-Goutières syndrome. Annals of Clinical Translational Neurology. 2015;2:774-779
  492. 492. Nazmi A, Field RH, Griffin EW, et al. Chronic neurodegeneration induces type I interferon synthesis via STING, shaping microglial phenotype and accelerating disease progression. Glia. 2019;67:1254-1276
  493. 493. Lall D, Lorenzini I, Mota TA, et al. C9orf72 deficiency promotes microglial-mediated synaptic loss in aging and amyloid accumulation. Neuron. 2021;109:2275-2291.e8
  494. 494. Stevens B, Allen NJ, Vazquez LE, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164-1178
  495. 495. Stevens B, Johnson MB. The complement cascade repurposed in the brain. Nature Reviews. Immunology. 2021;21:624-625
  496. 496. Escoubas CC, Dorman LC, Nguyen PT, et al. Type-I-interferon-responsive microglia shape cortical development and behavior. Cell. 2024;187:1936-1954.e24
  497. 497. Baker CA, Iwasaki A. Beyond antiviral: Role of IFN-I in brain development. Trends in Immunology. 2024. pp. 322-324. DOI: 10.1016/j.it.2024.04.004
  498. 498. Ramlackhansingh AF, Brooks DJ, Greenwood RJ, et al. Inflammation after trauma: Microglial activation and traumatic brain injury. Annals of Neurology. 2011;70:374-383
  499. 499. Smith C, Gentleman SM, Leclercq PD, et al. The neuroinflammatory response in humans after traumatic brain injury. Neuropathology and Applied Neurobiology. 2013;39:654-666
  500. 500. Kou Z, VandeVord PJ. Traumatic white matter injury and glial activation: From basic science to clinics. Glia. 2014;62:1831-1855
  501. 501. Graham NSN, Jolly A, Zimmerman K, et al. Diffuse axonal injury predicts neurodegeneration after moderate–severe traumatic brain injury. Brain. 2020;143:3685-3698
  502. 502. Raj R, Kaprio J, Jousilahti P, et al. Risk of dementia after hospitalization due to traumatic brain injury: A longitudinal population-based study. Neurology. 2022;98:e2377-e2386
  503. 503. Johnson VE, Stewart JE, Begbie FD, et al. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain. 2013;136:28-42
  504. 504. Krauthausen M, Kummer MP, Zimmermann J, et al. CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer’s disease model. The Journal of Clinical Investigation. 2015;125:365-378
  505. 505. Yu Z-X, Song H-M. Toward a better understanding of type I interferonopathies: A brief summary, update and beyond. World Journal of Pediatrics. 2020;16:44-51
  506. 506. Matsui H, Ito J, Matsui N, et al. Cytosolic dsDNA of mitochondrial origin induces cytotoxicity and neurodegeneration in cellular and zebrafish models of Parkinson’s disease. Nature Communications. 2021;12:3101
  507. 507. Yoshimoto N, Nakamura Y, Hisaoka-Nakashima K, et al. Mitochondrial dysfunction and type I interferon signaling induce anxiodepressive-like behaviors in mice with neuropathic pain. Experimental Neurology. 2023;367:114470
  508. 508. Smith JR, Dowling JW, McFadden MI, et al. MEF2A suppresses stress responses that trigger DDX41-dependent IFN production. Cell Reports. 2023;42:112805
  509. 509. Sheehan KCF, Lai KS, Dunn GP, et al. Blocking monoclonal antibodies specific for mouse IFN-alpha/beta receptor subunit 1 (IFNAR-1) from mice immunized by in vivo hydrodynamic transfection. Journal of Interferon & Cytokine Research. 2006;26:804-819
  510. 510. Peng L, Oganesyan V, Wu H, et al. Molecular basis for antagonistic activity of anifrolumab, an anti-interferon-α receptor 1 antibody. MAbs. 2015;7:428-439
  511. 511. Furie R, Khamashta M, Merrill JT, et al. Anifrolumab, an anti-interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis & Rhematology. 2017;69:376-386
  512. 512. Morand EF, Furie R, Tanaka Y, et al. Trial of anifrolumab in active systemic lupus erythematosus. The New England Journal of Medicine. 2020;382:211-221
  513. 513. Tang W, Tummala R, Almquist J, et al. Clinical pharmacokinetics, pharmacodynamics, and immunogenicity of anifrolumab. Clinical Pharmacokinetics. 2023;62:655-671
  514. 514. Tanaka Y. Viewpoint on anifrolumab in patients with systemic lupus erythematosus and a high unmet need in clinical practice. RMD Open. 2023;9. DOI: 10.1136/rmdopen-2023-003270
  515. 515. Chatham WW, Furie R, Saxena A, et al. Long-term safety and efficacy of anifrolumab in adults with systemic lupus erythematosus: Results of a phase II open-label extension study. Arthritis & Rhematology. 2021;73:816-825
  516. 516. Bruce IN, Golam S, Steenkamp J, et al. Indirect treatment comparison of anifrolumab efficacy versus belimumab in adults with systemic lupus erythematosus. Journal of Comparative Effectiveness Research. 2022;11:765-777
  517. 517. Furie R, Rovin BH, Houssiau F, et al. Two-year, randomized, controlled trial of belimumab in lupus nephritis. The New England Journal of Medicine. 2020;383:1117-1128
  518. 518. Brunner HI, Abud-Mendoza C, Viola DO, et al. Safety and efficacy of intravenous belimumab in children with systemic lupus erythematosus: Results from a randomized, placebo-controlled trial. Annals of the Rheumatic Diseases. 2020;79:1340-1348
  519. 519. Nakai T, Fukui S, Kidoguchi G, et al. Effect and safety profile of belimumab and tacrolimus combination therapy in thirty-three patients with systemic lupus erythematosus. Clinical Rheumatology. 2022;41:3735-3745
  520. 520. Teng YKO, Bruce IN, Diamond B, et al. Phase III, multicentre, randomized, double-blind, placebo-controlled, 104-week study of subcutaneous belimumab administered in combination with rituximab in adults with systemic lupus erythematosus (SLE): Bliss-believe study protocol. BMJ Open. 2019;9:e025687
  521. 521. Shipa M, Embleton-Thirsk A, Parvaz M, et al. Effectiveness of belimumab after rituximab in systemic lupus erythematosus: A randomized controlled trial. Annals of Internal Medicine. 2021;174:1647-1657
  522. 522. Merrill JT, Wallace DJ, Petri M, et al. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon α monoclonal antibody, in systemic lupus erythematosus: A phase I, multicentre, double-blind randomized study. Annals of the Rheumatic Diseases. 2011;70:1905-1913
  523. 523. McBride JM, Jiang J, Abbas AR, et al. Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: Results of a phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis and Rheumatism. 2012;64:3666-3676
  524. 524. Kalunian KC, Merrill JT, Maciuca R, et al. A Phase II study of the efficacy and safety of rontalizumab (rhuMAb interferon-α) in patients with systemic lupus erythematosus (ROSE). Annals of the Rheumatic Diseases. 2016;75:196-202
  525. 525. Doroudchi M-A, Thauland TJ, Patel BA, et al. Anifrolumab to treat a monogenic interferonopathy. The Journal of Allergy and Clinical Immunology in Practice. 2024;12:1374-1376.e1
  526. 526. Rachubinski AL, Estrada BE, Norris D, et al. Janus kinase inhibition in Down syndrome: 2 cases of therapeutic benefit for alopecia areata. JAAD Case Reports. 2019;5:365-367
  527. 527. Rodriguez S, Sahin A, Schrank BR, et al. Genome-encoded cytoplasmic double-stranded RNAs, found in C9ORF72 ALS-FTD brain, propagate neuronal loss. Science Translational Medicine. 2021;13. DOI: 10.1126/scitranslmed.aaz4699
  528. 528. Vanderver A, Adang L, Gavazzi F, et al. Janus kinase inhibition in the Aicardi-Goutières syndrome. The New England Journal of Medicine. 2020;383:986-989
  529. 529. Neven B, Al Adba B, Hully M, et al. JAK inhibition in the Aicardi-Goutières syndrome. The New England Journal of Medicine. 2020;383:2190-2191
  530. 530. Li W, Wang W, Wang W, et al. Janus kinase inhibitors in the treatment of type I interferonopathies: A case series from a single center in china. Frontiers in Immunology. 2022;13:825367
  531. 531. Frémond M-L, Hully M, Fournier B, et al. JAK inhibition in Aicardi-Goutières syndrome: A monocentric multidisciplinary real-world approach study. Journal of Clinical Immunology. 2023;43:1436-1447
  532. 532. Jafarpour S, Suddock J, Hawes D, et al. Neuropathologic impacts of JAK inhibitor treatment in Aicardi-Goutières syndrome. Journal of Clinical Immunology. 2024;44:68
  533. 533. Chougule A, Taur P, Gowri V, et al. SPENCD presenting with Evans phenotype and clinical response to JAK1/2 inhibitors-A report of 2 cases. Journal of Clinical Immunology. 2023;43:331-334
  534. 534. Kothur K, Bandodkar S, Chu S, et al. An open-label trial of JAK 1/2 blockade in progressive IFIH1-associated neuroinflammation. Neurology. 2018;90:289-291
  535. 535. Cattalini M, Galli J, Zunica F, et al. Case report: The JAK-inhibitor ruxolitinib use in Aicardi-Goutieres syndrome due to ADAR1 mutation. Frontiers in Pediatrics. 2021;9:725868
  536. 536. Galli J, Cattalini M, Loi E, et al. Treatment response to Janus kinase inhibitor in a child affected by Aicardi-Goutières syndrome. Clinical Case Reports. 2023;11:e7724
  537. 537. Rice GI, Meyzer C, Bouazza N, et al. Reverse-transcriptase inhibitors in the Aicardi–Goutières syndrome. The New England Journal of Medicine. 2018;379:2275-2277
  538. 538. Sullivan AC, Zuniga G, Ramirez P, et al. A pilot study to investigate the safety and feasibility of antiretroviral therapy for Alzheimer’s disease (ART-AD). medRxiv. 2024. DOI: 10.1101/2024.02.26.24303316
  539. 539. Kulkarni JA, Witzigmann D, Thomson SB, et al. The current landscape of nucleic acid therapeutics. Nature Nanotechnology. 2021;16:630-643
  540. 540. Finkel RS, Mercuri E, Darras BT, et al. Nusinersen versus Sham control in infantile-onset spinal muscular atrophy. The New England Journal of Medicine. 2017;377:1723-1732
  541. 541. Miller TM, Cudkowicz ME, Genge A, et al. Trial of antisense oligonucleotide tofersen for SOD1 ALS. The New England Journal of Medicine. 2022;387:1099-1110
  542. 542. Viengkhou B, Hong C, Mazur C, et al. Interferon-α receptor antisense oligonucleotides reduce neuroinflammation and neuropathology in a mouse model of cerebral interferonopathy. The Journal of Clinical Investigation. 2024;134. DOI: 10.1172/JCI169562
  543. 543. Terstappen GC, Meyer AH, Bell RD, et al. Strategies for delivering therapeutics across the blood–brain barrier. Nature Reviews. Drug Discovery. 2021;20:362-383
  544. 544. Oller-Salvia B, Sánchez-Navarro M, Giralt E, et al. Blood–brain barrier shuttle peptides: An emerging paradigm for brain delivery. Chemical Society Reviews. 2016;45:4690-4707
  545. 545. Niewoehner J, Bohrmann B, Collin L, et al. Increased brain penetration and potency of a therapeutic antibody using a monovalent molecular shuttle. Neuron. 2014;81:49-60
  546. 546. Das T, Chen Z, Hendriks RW, et al. A20/tumor necrosis factor α-induced protein 3 in immune cells controls development of autoinflammation and autoimmunity: Lessons from mouse models. Frontiers in Immunology. 2018;9:104
  547. 547. Deverman BE, Pravdo PL, Simpson BP, et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nature Biotechnology. 2016;34:204-209
  548. 548. Mullard A. NfL makes regulatory debut as neurodegenerative disease biomarker. Nature Reviews. Drug Discovery. 2023;22:431-434
  549. 549. Benatar M, Ostrow LW, Lewcock JW, et al. Biomarker qualification for neurofilament light chain in amyotrophic lateral sclerosis: Theory and practice. Annals of Neurology. 2024;95:211-216
  550. 550. Hauser SL, Kappos L, Arnold DL, et al. Five years of ocrelizumab in relapsing multiple sclerosis: OPERA studies open-label extension. Neurology. 2020;95:e1854-e1867
  551. 551. Younossi ZM, Stepanova M, Esteban R, et al. Superiority of interferon-free regimens for chronic Hepatitis C: The effect on health-related quality of life and work productivity. Medicine (Baltimore). 2017;96:e5914
  552. 552. Borden EC. Interferons α and β in cancer: Therapeutic opportunities from new insights. Nature Reviews. Drug Discovery. 2019;18:219-234
  553. 553. Kirkwood JM, Strawderman MH, Ernstoff MS, et al. Interferon Alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The eastern cooperative oncology group trial EST 1684. Journal of Clinical Oncology. 2023;41:425-435

Written By

Alessio Mylonas

Submitted: 13 June 2024 Reviewed: 24 June 2024 Published: 31 July 2024