Open access peer-reviewed chapter

Omics and Network-Based Approaches in Understanding HD Pathogenesis

Written By

Christiana C. Christodoulou and Eleni Zamba Papanicolaou

Submitted: 01 June 2023 Reviewed: 05 June 2023 Published: 29 February 2024

DOI: 10.5772/intechopen.1001983

From the Edited Volume

Rare Neurodegenerative Disorders - New Insights

Liam Chen

Chapter metrics overview

55 Chapter Downloads

View Full Metrics

Abstract

Huntington’s Disease (HD) is a rare, progressive neurodegenerative disease caused by CAG repeat expansion in the Huntingtin gene. HD is an incurable disease; therefore, there is a growing need for effective therapeutic treatments and candidate biomarkers for prognosis and diagnosis of HD. Technological advancements over the past couple of years, have led to high-throughput experiments and omics data. The use of System Bioinformatics (SB) approaches, allows for the integration of information across different -omics, this can clarify synergistic relationships across biological molecules, resulting in complex biological networks. SB and network-based approaches, are able to shed light on the potential interactions of genes, proteins, metabolites and pathways participating in HD pathogenesis and how dysregulation of these biological entities, can affect age on onset, disease severity and progression. Moreover, −omics data analysis and network-based approaches can provide better understanding how these biological molecules interact with each other and provides potential drug targets and biomarkers that can be used to treat HD or delay symptom onset; therefore, opening the door towards precision medicine. The aim of the following chapter, is to discuss the most popular -omics related to HD research, and the growing popularity of single cell analysis, repositories and software available for bulk and single cell analysis. In addition, network-based approaches regarding HD will also be mentioned.

Keywords

  • Huntington’s disease
  • omics
  • bioinformatics
  • systems bioinformatics
  • single cell analysis
  • network-based approaches

1. Introduction

Huntington’s Disease (HD) was first described in 1842 by Charles Oscar Waters and by 1872 a description of the disease was given by George Huntington, who assessed the medical history of several generations of a family exhibiting similar symptoms [1]. In 1983, a linkage on chromosome 4 was performed and by 1993 the Huntingtin (HTT) gene was discovered [1], leading to an increase in the interest of HD. Moreover, the identification of the gene resulted in the development of new animal models, research and therapeutic treatments and drugs to treat HD [1, 2]. HD is the most common monogenic neurological diseases in the developed world [3]. It is a rare, progressive neurodegenerative disease (ND) with autosomal dominant inheritance [1, 2, 3] affecting the medium spiny neurons (MSN) of the basal ganglia. HD is characterized by motor, cognitive and behavioral impairment caused by CAG trinucleotide repeat expansion at the N-terminus of the HTT gene responsible for encoding the HTT protein [4].

HD as other NDs, including Alzheimer’s disease (AD), Parkinson’s disease (PD) and Amyotrophic lateral Sclerosis (ALS) remain incurable [5]. Due to the overwhelming nature of these diseases, high economic and social costs and lack of effective therapeutic treatments [5]. Therefore, there is an increased need for novel biomarkers, pathways, drug targets and new novel pharmacotherapies that will enable the predication, prevention, understanding of disease pathways and most importantly effective treatments for NDs such as HD [5].

In the recent decade, bioinformatics approaches have become of increasing interest especially in NDs as they enable the identification of candidate biomarkers, pathways and mechanisms implicated in disease and potential drugs and their targets [5]. Bioinformatics is a multidisciplinary field utilizing methods from statistics, data-analysis, computer science, mathematics and biology to solve complex biological questions [5]. Bioinformatics is essential for analyzing and interpreting data from high-throughput technologies (DNA- and RNA-sequencing, proteomics, metabolomics, lipidomics etc.) [5, 6]. Due to the large amount of data produced, bioinformatics-based data management, and analysis tools are needed for the extraction, analysis, interpretation, visualization and storage of the data [5]. A relatively new approach is System Bioinformatics (SB), an intersection between systems biology and bioinformatics, which focuses on the integration of information across different -omics levels [6]. The goal of SB is to elucidate synergistic relationships between numerous factors in contrast to representing them as single biological entities, resulting in complex molecular networks of interactions [5]. This allows to better understand how different biological entities interact with one another, thus providing a clear understanding of disease pathogenesis, pathways, genes, proteins, metabolite, lipids and several additional types of biological information interacting together to lead to disease development [5]. Therefore, it is not a surprise that bioinformatics methods are used increasingly in ND research [5, 6].

Cells are complex, heterogenous and show a radical variation at the individual level and recent technological advancements, have enabled cell profiling at the individual level known as single cell analysis (SCA); the analysis of this data using bioinformatics pipelines is a practical solution for researchers; shedding light in understanding diseases, pathogenic mechanisms and the identification of potential biomarkers of diseases. The following chapter will discuss the most popular -omics approaches, SC technologies, data repositories and network-based analysis in HD research.

Advertisement

2. Omics

The past 15 years have seen the affluent use and integration of -omics approaches that include genomics and epigenomics for DNA, transcriptomics for RNA, proteomics for proteins and post-translational modification, metabolomics for metabolites, lipidomics for lipids and several additional -omics approaches (pharmacogenomics, radiomics etc.) [7, 8]. Technological advancement has allowed for the comprehensive measurement and analysis and the global assessment of biological molecules [7, 8] of genes, proteins, metabolites, lipids and several additional omics types within biological fluids, tissues or cells [8, 9], to assist in the understanding and investigation of human health and diseases [7]. These high-throughput technologies can be applied to the biological system and disease of interest to obtain a snapshot of the underlying biology at a resolution which was not previously possible [8], leading to unprecedented ways to better understanding NDs in terms of their pathogenesis, pathways, mechanisms and the interaction between different biological entities. Omics can contribute to biomarker, drug discovery and drug repurposing resulting in novel therapeutic treatments for NDs. Omics data is analyzed using bioinformatics pipelines and tools to obtain a list of genes, proteins, metabolites or lipids from the disease of interest [10], biological interpretation using different bioinformatics approachesTable 1 (pathway enrichment analysis, protein–protein interaction networks (PPIs), protein-metabolite networks etc.) is vital to gain insight into the mechanisms, pathways and molecules affected in disease. The most common omics and the research conducted in regards to HD will be discussed below (Figure 1).

NameTypeOmics contentRefWebsite (If Available)
GalaxyOpen accessGenomics
Transcriptomics
Proteomics
Metabolomics
Metagenomics
[11]https://usegalaxy.org/
LimmaR-packageTranscriptomics
Proteomics
Metabolomics
[12]https://www.bioconductor.org/packages/release/BiocViews.html#___Metabolomics
Ingenuity pathway analysis (IPA)Commercial SoftwareTranscriptomics
Proteomics
Metabolomics
NextFlowSoftwareGenomics
Transcriptomics
Proteomics
Metabolomics
https://www.nextflow.io/
EdgeRR packageGenomics
Transcriptomics
[13, 14, 15]
MaxQuant & PerseusOpen accessProteomics[16]https://www.maxquant.org/
OpenMSC++ libraryProteomics
Metabolomics
[17]https://openms.de/
MsCoreUtilsR packageProteomics
Metabolomics
[18]https://www.bioconductor.org/packages/release/bioc/html/MsCoreUtils.html
MetaboAnaylstOpen access & R packageMetabolomics
Lipidomics
Multi-omics
[19, 20, 21]https://www.metaboanalyst.ca/
MetaCycOpen accessMetabolomics[22]https://metacyc.org/
XCMSOpen access & R packageMetabolomics[23, 24, 25]https://xcmsonline.scripps.edu/landing_page.php?pgcontent=mainPage
MzmineR packageMetabolomics[26]http://mzmine.github.io/

Table 1.

Online tools, software and R-packages for omics data analysis.

Figure 1.

Omics data types and bioinformatics based approaches.

2.1 Genomics

Since the discovery of the DNA structure by Rosalind Franklin, Frances Crick and James Watson in 1953, there has been a revolution in the field of biological sciences regarding technological advancements such as Sanger sequencing [27, 28, 29] and the discovery of the polymerase chain reaction [30] at the beginning of the 21st century. NGS became available, enabling the production of huge amounts of data along with the ability to produce highly efficient, rapid, low-cost approach along with accurate DNA sequencing [31]. Genomics is defined as the interdisciplinary field of biology focusing on genes, their structure, function, evolution, expression, mapping and genome editing [32]. The genome refers to all the genetic material present in any organism, including chromosomal and extrachromosomal DNA, coding and non-coding genes, microRNAs (miRNAs) and single nucleotide polymorphisms (SNPs) [33]. Sequencing of the human genome is important as it can provide insight into: i) gene expression profile of a specific tissue, organ or tumor, ii) human variation and how genomic alterations lead to disease development, iii) identification of genetic modifiers of disease, and iv) determine rate sensitivity to drugs in patients, based on their DNA sequences, this is known as pharmacogenomics [34]. Genomics has the ability to discover and identify genes associated with a disease; making it a vital omics component towards precision medicine. The majority of studies that have taken place regarding HD research have involved either the identification of genetic modifiers that may play a role in the early or delayed onset of HD.

2.1.1 Genomics in HD patients

Marchina et al. [35] investigated the gene expression profiles of fibroblasts of HD patients and healthy controls using microarray technology, and RT-PCR to validate the consistency of the microarray data. Analysis was carried out on nine genes, namely APC, CDC42EP2, CTNNB1, DNM1L, PLCB4, ROCK1, ROCK2, SSH1, and UBE2D3. These lists of genes, were chosen as some of these genes showed increased differential up-regulation (PLCB4, APC) or down-regulation (SSH1, CDC42EP2); while other genes were selected due to their possible involvement in HD pathology (UBE2D3, ROCK1, ROCK2, DNM1L, CTNNB1) as indicated by previous studies. The APC, PLCB4, ROCK1, and UBE2D3 genes were found to be up-regulated in HD patients in comparison to controls [35]. Some gene ontology (GO) terms identified are, i) transcription, ii) regulation of transcription, DNA-dependent, iii) cell cycle, iv) response to DNA damage stimulus, v) DNA repair, vi) ubiquitin dependent protein catabolic process and vii) DNA recombination. Based on this evidence the gene expression profiles of fibroblasts seem to be altered in HD patients compared to healthy controls [35]. Validation of the differential expressions at the protein level is needed to confirm if fibroblasts can be considered as a suitable model for the identification of HD biomarkers [35].

A study by Wright et al. [36] investigated the gene expression profiles complementing the analysis of genomic modifiers in HD. CAG repeat length is not the only contributing factor to disease onset but genetic modifiers also contribute to disease onset [36]. A genome-wide association study assessing heritable differences in genetically determined expression in diverse tissues, with genome-wide data taken from 4000 HD patients was conducted. In addition, functional validation of prioritized genes was performed in isogenic HD stem cells and post-mortem brain tissue [36]. The genes of FAN1, GPR161, PMS2 and SUMF2 are associated with age of onset in HD and showed co-localization with gene expression signals in brain tissue [36].

Other genomic studies conducted in HD patients or post-mortem brain tissue include Moss et al. [27], Li et al. [28].

2.1.2 Genomics in HD animal models

One study by Tang et al. [29] performed gene expression profiling on the R6/2 HD transgenic mice model, with varying CAG repeat lengths to reveal genes associated with HD onset and progression [29]. The R6/2 transgenic mice with >300 CAG repeats had prolonged HD progression and a longer lifespan than the parent R6/2 mice with 150 CAG repeats. However, the mechanisms regarding this phenotypic amelioration remains unknown [29].

The expression profiles of the striatum of R6/2 transgenic mice with >300 CAG repeats (R6/2Q300 transgenic mice) and R6/2 transgenic mice with 150 CAG repeats (R6/2Q150 transgenic mice) and littermate wild-type (WT) controls were performed to identify, genes playing an important role, in HD onset and progression. Both R6/2 mouse models demonstrated decreased expression while up-regulated gene expression was seen in R6/2Q300 mice [29]. The up-regulated genes play a role in ubiquitin ligase complex, cell adhesion, protein folding and establishment of protein localization. Increased gene expression for Lrsam1, Erp29, Nasp, Tap1, Rab9b and Pfdn5 was validated using qPCR. Moreover, Lrsam1 and Erp29 were significantly up-regulated in R6/2Q300 mice and may have potential neuroprotective effects in primary striatal cultures over-expressing mHTT [29]. Over-expression of Lrsam1 prevented the loss of NeuN-positive cell bodies in htt171-82Q cultures, associated with a decrease of nuclear HTT aggregates. However, over-expression of Erp29 demonstrated no significant effect in cells [29]. Therefore, prolonged HD onset and progression seen in R6/2 mice with increased CAG repeat expansions are a result of differential up-regulation in genes involved in protein localization and clearance. These genes may be possible novel therapeutic avenues in decreasing HTT aggregation toxicity and neuronal cell death, with Lrsam1 being a promising, novel candidate disease modifier [29].

Langfelder et al. [37] integrated genomics and proteomics data to define HTT CAG length networks in HD mice. To gain insight into how mHTT CAG repeat length modifies HD pathogenesis, profiling of mRNA in 600 brain and peripheral tissue samples obtained from HD knock-in mice with increased CAG repeat length was performed [37]. The study found, the repeat length dependent transcriptional signatures were notable in the striatum while the cortex and liver showed less transcriptional signatures [37]. Co-expression networks revealed 13 and 5 striatal and cortical modules, respectively, that were highly correlated with CAG repeat length and age, and are preserved in HD models. The top striatal modules suggest mutant HTT (mHTT) CAG length and age impair the MSN, while a dysregulation of genes and pathways involved in cAMP signaling, cell death, and protocadherin were seen [37]. Proteomics analysis was used to confirm, 790 genes and 5 striatal modules with CAG length-dependent dysregulation was observed at both the RNA and protein level, 22 striatal module genes were validated as modifiers of mHTT toxicities in vivo [37].

Other genomics studies in HD animal models or cells include Choudhury et al. [38] and Alcalá-Vida et al. [39].

2.2 Transcriptomics

The transcriptome is the complete set of transcripts within a cell, and their quantity, for a physiological condition or specific developmental stage [40]. Understanding the transcriptome is crucial for interpreting the functional components of the genome and revealing the molecular entities of cells and tissues and also for understanding development and disease pathogenesis [40]. The aim of transcriptomics is to: i) catalog the transcripts, including mRNAs, non-coding RNAs and small RNAs; of all species to determine the transcriptional structure of genes, regarding their start sites, 5′ and 3′ ends, alternative splicing patterns and any additional post-transcriptional modifications; and ii) quantify the fluctuating expression levels of each transcript under different conditions and during development [40]. The development of innovative high-throughput DNA sequencing methods known as RNA Sequencing (RNA-Seq) has allowed the mapping and quantification of transcriptomes. Genomics and transcriptomics are the two most popular omics used for HD research; therefore, it is vital to examine whether peripheral tissues may serve as a possible source of readily accessible biological signatures at not only the DNA and RNA level but also at the protein level and not only. There are numerous studies that have used transcriptomics to perform studies regarding either peripheral tissues, cerebrospinal fluid (CSF) or post-mortem brain tissue samples obtained from HD patients and animal models.

2.2.1 Transcriptomics in HD patients

A study by Neueder et al. [41] investigated the abnormal molecular signatures that characterize inflammation, energy metabolism and vesicle biology in human HD peripheral tissues. Specifically, skeletal muscle, adipose tissue and skin from the quadriceps femoris muscle and blood was collected from 21 pre-HD patients, 20 early motor manifest HD patients and 20 healthy controls. Furthermore, primary fibroblast and myoblast cell lines were established [41]. RNA-Seq and proteomics analysis were used to investigate the involvement of inflammation and energy metabolism in HD pathogenesis.

Initially, RNA-Seq analysis was performed on adipose and muscle tissue from the pre-HD patients (pre-HD), early motor manifest HD patients (early HD) and healthy control groups. In total, 78 and 53 genes were identified to be significantly dysregulated in adipose and skeletal muscle tissue, respectively [41]. Distinctly, only RPH3A, PAX6 and AC016582.1 were regulated similarly in the pre-HD and early HD groups in comparison to controls. Furthermore, some genes were differentially regulated in both groups [41]. TBC1D3D, was differentially regulated in muscle and adipose tissue, the gene was found to be up-regulated in the early HD group in comparison to the pre-HD group [41].

GO enrichment analysis in adipose tissue identified the terms of protein sumoylation (GO: 0016), interleukin-1 mediated signaling pathway (GO:0006470) and cellular response to organic cyclic compound (GO: 0071407) [41]. For the different comparisons, the RE1 silencing TF (REST) target gene was dysregulated only in the pre-HD group, while sumoylation was dysregulated only in the early HD group. Alteration in protein sumoylation is a well-known mechanism and has been implicated in HD pathogenesis [41].

GO analysis in muscle tissue, identified a disruption in homeostatic pathways, including anterograde trans-synaptic signaling (GO: 0098916), regulation of fatty acid oxidation (GO:0046320), regulation of glucose metabolic process (GO:0010906), negative regulation of cell communication (GO:00106458), protein dephosphorylation (GO:0006470), regulation of protein kinase B signaling (GO:0051896) and regulation of MAPK cascade (GO:0043408) particularly in the pre-HD group [41]. PAX6 is a vital regulator of assorted peripheral and central nervous system processes, it was identified to be highly and gradually up-regulated in both HD groups; therefore, suggesting a compensatory mechanism of muscle regeneration in response to mutant HTT expression [41]. Enrichment analysis of the dysregulated genes in the muscle tissue of the pre-HD group suggest that peroxisome proliferator activated receptor alpha (PPARA) acts as a regulatory protein [41].

In addition, RNA-Seq analysis was also performed on the fibroblast cell lines. Only a few genes were significantly dysregulated between pre-HD and early-HD patient groups compared to healthy controls [41]. Subsequently, GO enrichment analysis resulted in the identification of a few enriched terms, such as phosphorylation (GO:0016310) and regulation of apoptotic processes (GO:0042981) [41]. Interestingly, TBC1D3D, was found to be dysregulated not only in adipose and muscle but also in fibroblasts [41]. Furthermore, proteomics analysis was performed on the tissues obtained; however, this will be further discussed in the proteomics section of the chapter.

Miller et al. [42], performed RNA-Seq analysis on myeloid cells of HD patients to identify transcriptional dysregulation associated with proinflammatory pathway activation. Specifically, whole transcriptome analysis of primary monocytes from 30 manifesting HD patients and 31 healthy control subjects, with or without proinflammatory stimulus [42].

HD monocytes exhibit resting proinflammatory transcriptional changes, 12,599 genes were identified to be differentially expressed (DE) and analysis of said data was conducted, to determine the genes significantly altered between HD and control monocytes, whereas the stimulated and unstimulated monocytes were analyzed separately [43]. Analysis of unstimulated monocytes revealed 130 DE genes, of which 101 were up-regulated and 29 down-regulated genes in resting HD monocytes compared to healthy controls. The DE genes were associated with proinflammatory cytokines and chemokines. HD monocytes had significantly increased expression of IL6, IL12B, IL19, IL23A, CCL8, CCL19, CCL20, CXCL6 and CSF2 gene transcripts. All transcripts have a > 2-fold increase in mRNA expression [42]. In contrast, the stimulated monocytes showed little indication of differential expression between HD and controls, the genes of DNAJB13, STAC and RASEF were DE. Furthermore, each of these genes were observed to be DE in unstimulated monocytes [42]. Generally, gene expression differences between HD and controls were lower in the stimulated 116 DE genes in comparison to the unstimulated 130 DE genes in monocytes [43]. Moreover, gene set enrichment analysis (GSEA) was performed for the up- and down regulated gene expression for both the stimulated and unstimulated monocyte datasets [43]. Analysis of unstimulated monocytes revealed a total of 85 enriched gene sets, a majority of pathways relating to innate immunity, inflammatory response, cytokine production and secretion were identified. Furthermore, pathways such as NFkB and JAK/STAT signaling pathways were also identified to be significantly enriched [43]. In comparison to the down-regulated genes, 6 gene sets were identified and included significantly enriched pathways related to vacuole, lysosome and catabolic functions [42]. Interestingly, the simulated dataset did not reveal any enriched gene sets among the up-regulated genes. However, 83 gene sets were significantly enriched among the down-regulated genes, pathways relating to cholesterol homeostasis, cellular components such as cellular membrane, mitochondria and lysosomes [42].

To understand the mechanisms underlying differential gene expression in unstimulated HD monocytes, upstream regulator analysis was performed using the Ingenuity Pathway Analysis (IPA) software. A total of 155 upstream regulators were identified for the unstimulated dataset, of these 125 were predicted to be significantly activated, while 30 were predicted to be significantly inhibited. A large number of molecules associated with intracellular signaling pathways downstream of the TLR4 receptor were represented in the unstimulated group. The following data were consistent with previous studies, showing NFkB dysregulation in HD myeloid cells, the RELA and the NFkB complex were among the top most significant results ranked by z-score activation [43]. Other prominent potential regulators include NFkB1, ERK and p38 MAPKs, in addition to the transcription factor STAT3 [42]. The study suggests that transcriptional changes observed in the RNA-Seq analysis of stimulated and unstimulated HD monocytes is related to the abnormal activation of specific upstream signaling molecules responsible for driving gene expression in unstimulated HD myeloid cells [42]. Furthermore, HD myeloid cells have a proinflammatory phenotype in the absence of stimulation; consistent with the priming effect of mutant huntingtin (mHTT), whereas basal dysfunction results in an exaggerated inflammatory response once a stimulus is encountered [42]. The following data provides an understanding of mHTT pathogenesis, establishing unstimulated myeloid cells as a vital source of HD immune dysfunction, and demonstrating the importance of immunity as a potential HD treatment.

Other transcriptomic studies conducted in HD patients include Seefelder and Kochanek [44], Sinha et al. [45], Mastrokolias et al. [46, 47], Borovecki et al. [48], Lin et al. [40], Cha [49], Moily et al. [43], Mehta et al. [50], Stopa et al. [51] and Runne et al. [52].

2.2.2 Transcriptomics in HD mouse models

Jin et al. [53] investigating the miRNA and mRNA expression profiles of the cerebral cortex of N171-82Q HD mice [53]. There is growing evidence indicating that miRNAs may play a role in HD pathogenesis [53]. During disease progression significant changes of miRNAs in the cerebral cortex were also detected in the striatum of HD mice [53]. The study revealed that a significant alteration in the miR-200 miRNA family, more specifically, miR-200a and miR-200c in the cerebral cortex and striatum were seen during the early HD stages in N171-82Q mice.

A computational bioinformatics approach was used to integrate miRNA and mRNA profiles. The gene targets were identified using TargetScan, for miR-200a and miR-200C, there are 680 and 462 gene targets respectively. Some of the predicted gene targets for the miRNAs include KIF3a, NRXN1, PTPRD, TRIM2, ATXN1, KCNA2 and numerous additional targets [53]. The predicted targets play a role in regulating synaptic function, neuronal survival and neurodevelopment. The results of the study suggest that altered miR-200a and miR-200c expression may interrupt protein production involved in neuronal plasticity and survival [53]. Therefore, further investigation of the involvement of dysfunctional miRNA expression in HD is required and this may result in novel approaches for HD therapy.

Hervás-Corpión [54] investigated the early alternations of epigenetic-related transcription in HD mouse models. The gene expression profiles of the cortex, striatum, hippocampus and cerebellum of juvenile R6/1 and N171-82Q mice, were studied, the profiles consisted of tissular and neuronal-specific genes and showed significant correspondence with transcriptional alteration in HD mouse models deficient of epigenetic regulatory genes [54]. A noteworthy case was the conditional knockout of the lysine acetyltransferase CBP in post-mitotic forebrain neurons, the double knockout of the histone methyltransferases Ezh1 and Ezh2, components of the polycomb repressor complex 2 (PRC2), and the conditional mutants of the histone methyltransferases G9a (Ehmt2) and GLP (Ehmt1) [54]. It is likely that the neuronal epigenetic status is compromised prior to HD onset resulting in transcriptional dysregulation [54].

Other transcriptomic studies conducted in HD animal models include Bensale et al. [55], Dickson et al. [56], Reyes-Ortiz [57], Chaves et al. [58] and Huang et al. [59].

2.3 Proteomics

Proteomics is a new omics type that has rapidly developed especially in the fields of therapeutics and biomarkers discovery [60]. Proteomics is defined as the study of interactions, functions, composition and structure of proteins and their cellular activities. The proteome is defined as the complete set of proteins within a cell, tissue, organism or biological fluid. Proteomics allows for a better understanding of the structure and function of an organism than genomics. However, proteomics is much more cumbersome than genomics as the protein expression is altered according to environmental conditions and time. It is approximated that there are almost one million human proteins, many of which contain some form of post-translational modifications [60]. The majority of proteomics studies have been conducted in HD mouse models, there seems to be a lack of human HD proteomics studies. Proteomics can assist in the discovery of new therapies, biomarkers and better understanding of proteins and protein-complexes in disease.

2.3.1 Proteomics in HD patients

The study by Neueder et al. [49] which previously performed transcriptomic analysis as mentioned in Section 2.2.1 also performed proteomics analysis and revealed, 1347, 2671 and 4640 proteins for adipose, muscle and skin tissue respectively [49]. A progressive increase in the number of dysregulated proteins from the pre-HD to early HD stage was observed for all three tissues [49]. In adipose tissue, there was no overlap of commonly dysregulated proteins between the three comparisons. In muscle tissue, syntaxin-binding protein 3 (STXBP3) was up-regulated in both pre- and early-HD samples. Interestingly, the major change was observed in the skin samples, MOB4 was up-regulated in both pre- and early HD samples [49]. A total of 162 and 23 proteins were significantly dysregulated in the early and pre-HD stages respectively.

GO enrichment on adipose dataset (early HD vs. controls) and skin dataset (early HD vs. controls and the comparison of pre-HD vs. early HD), revealed dysregulated lipid metabolism and proteasome function in the adipose tissue dataset. While proteins related to gene expression such RNA processing and translation and amino acid metabolism are mainly affecting in the early HD samples vs. controls [49]. The predicted regulators are all genes involved in gene expression, specifically, lysine acetyltransferase 2A (KAT2A) and the androgen receptor (AR), in a complex with ataxin 7 (ATXN7) which are associated with polyglutamine diseases [49]. The GO terms for pre- and early HD in the skin datasets, included the regulation of cell survival and proliferation. The results obtained from the above study, indicate that alterations in biological signatures at the RNA and protein level point towards the direction of inflammation, energy metabolism and vesicle biology in peripheral tissues in HD. The following biological signatures may act as suitable biomarkers in clinical trials upon further validation.

A proteomics study by Fang et al. [61] integrated five sets of proteomics data profiling the CSF derived from HD affected and unaffected individuals with genomics data profiling, various human and mouse tissue, including the human HD brain [61]. According to the integrated analysis, brain specific proteins were 1.8 times more likely to be observed in CSF compared to plasma. Furthermore, brain specific proteins decrease in HD CSF compared to unaffected CSF [61].

Approximately, 81% of brain specific proteins have quantitative changes that agree with transcriptional changes seen in different HD brain regions, while the proteins identified to increase in HD CSF tend to be liver associated. The protein changes are consistent with microgliosis, astrocytosis and neurodegeneration which are known to occur in HD [61]. The most significantly over and under abundant dysregulated proteins in CSF between HD affected and unaffected individuals, include, chromogranin B (CHGB), isoform I of Sialate O-actelyesterase precursor (SIAE), isoform long of iduronate 2-sulfatase precursor (IDS), Neurexin 3-alpha (NRXN3) Endonuclease domain-containing 1 protein precursor (ENDOD1), major prion protein precursor (PRNP) and several additional proteins have a trend of decreasing with disease progression (controls > HD early > mid-HD). Some of the over abundant proteins identified are, complement component 1, q subunit, c chain precursor (C1QC), hemopexin (HPX), Triosephosphate isomerase (TPI1), Isoform M1 of Pyruvate kinase isozymes M1/M2; Isoform R-type of Pyruvate kinase isozymes R/L (PKM2/PKLR), Lysozyme C precursor (LYZ) and several other proteins, which have shown to have a trend of increasing as the disease progresses (control > HD early > mid HD) [61].

Other proteomics studies conducted in HD patients include, Chen et al. [62], Schönberger et al. [63], Sorolla et al. [64] and Chae et al. [65], McQuade et al. [66] and Dalrymple et al. [67].

2.3.2 Proteomics in mouse models of HD patients

A previous study by Agrawal and Fox [68] performed targeted proteomics analysis to investigate novel proteomic alterations in brain mitochondria to reveal insights into mitochondrial dysfunction in HD mouse models. Mitochondrial dysfunction is one of contributing pathophysiological mechanisms in HD. The following study used R6/2 and YAC128 HD mouse models to represent different HD progression rates to pinpoint HD brain mitochondrial proteomic signatures. Cerebral cortical mitochondrial of HD and WT littermates were compared by 2D SDS–PAGE electrophoresis and MALDI-TOF/TOF mass spectrometry (MS) [68].

Proteomic analyses concluded 17 and 12 DE proteins in 12-week R6/2- and 15-month YAC128 HD mice, respectively compared to controls. The proteins of peroxiredoxin 3, stress–70, DJ–1, isocitrate dehydrogenase [NAD] α subunit and ATP synthase subunit D were DE in both HD mouse models. Pathway analysis was performed using PANTHER [68], the GO molecular function terms obtained for the DE mitochondrial proteins are i) catalytic activity (GO:0003824), binding (GO:0005488) and antioxidant activity (GO:0016209); most GO biological process proteins belonged to metabolic (GO:0008152) and cellular process (GO:0009987) [68]. While for YAC128 mice, the molecular functions of the DE mitochondrial proteins were catalytic activity (GO:0003824) and binding (GO: 0005488); for GO biological processes most of the proteins also belonged to the metabolic (GO:0008152) and cellular process (GO:0009987) and biogenesis (GO:0071840). The results identify a proteomic signature of HD mitochondria in mouse models that includes previously unrecognized proteins [68].

One study by Choudhary et al. [38] performed differential proteomics and genomic profiling of mouse striatal cell model of HD vs. healthy controls. Transcriptional dysregulation is one of the pathogenic mechanisms contributing to HD. Various studies have identified altered gene expression in HD patient brains and animal models. 2D SDS-PAGE/MALDI-MS coupled with 2D-DIGE and real time PCR on an array of genes concentrated on HD pathways to identified altered proteins and gene expression in STHdhQ111/HdhQ111 compared to STHdhQ7/HdhQ7 (wild-type). Seventy-six proteins were annotated in HD cells while 31 proteins were DE by 2D-DIGE. The bioinformatics tool GeneCodis3 [38] was used to perform pathway analysis using KEGG and GO biological terms. The pathways included, unfolded protein binding (GO:0051082), negative regulation of neuron apoptosis (GO:0006916), response to superoxide’s (GO:0006950) and several other pathways. The PCR experiments showed altered gene expression of 47 genes. Altogether, 77 genes/proteins were identified in HD cell lines with potential relevance to HD biology [38].

Other proteomic studies conducted in HD animal models and cells include Mees et al. [69], Liu et al. [70], Perluigi et al. [71], Deschepper et al. [72], Culver et al. [73], Cozzolino et al. [74], Vodicka et al. [75], Sap et al. [76], Ratovitski et al. [77] and Zabel et al. [78].

2.4 Metabolomics

Traditionally, a small number of metabolites have been used for the diagnosis of complex metabolic diseases and monogenic complex diseases such as inborn errors of metabolism [79]. Metabolomics is defined as the comprehensive measurement of metabolites within a biological tissue, cell, organ or fluid (CSF, urine, plasma or serum) [79]. The metabolome is the number of metabolites in an organism. Metabolomics is an emerging technology that holds promise for the development and practice of precision medicine. In addition, metabolomics is able to provide detailed characterization of metabolic phenotypes and can enable precision medicine at numerous levels such as characterization of metabolites underlying disease, discovery of new therapeutic targets, biomarker discovery which may be used for prognosis, diagnosis or monitor activity of therapeutics [79]. Similarly with proteomics, the majority of studies in metabolomics and HD are conducted in HD mouse models.

2.4.1 Metabolomics in HD patients

One study by Rosas et al. [80] investigated the plasma metabolome of HD. The study consisted of targeted metabolomics analysis using the plasma obtained from 52 pre-HD, 102 early symptomatic HD and 140 controls [80].

The pathways altered include i) tryptophan, ii) tyrosine, iii) purine, iv) methionine, v) antioxidant pathways and numerous pathways relating to energetic and oxidative stress derived from the gut microbiome. The tryptophan, tyrosine and purine pathways were altered in prodromal and early HD stages. The selective dysregulation of a good few pathways and the increased regulation of other pathways suggest complex alterations in the feedback controls of underlying genes, proteins, enzymes and metabolites. In addition, multivariate statistical modeling demonstrated mutually distinct metabolomics profiles, suggesting that the process determining onset was likely distinct from processes determining progression [80]. Surprisingly, controls, pre-HD and early HD plasma metabolomes were mutually distinct rather than differing, suggesting varying influences during the prodromal and symptomatic disease stages [80].

Numerous metabolites differentiating the control from the pre-HD and early HD metabolomes, are linked to the gut microbiome, suggesting mHTT favors a distinct microbiome. The systemic effects of HD on the gut microbiome may possibly influence energy homeostasis, vitamin and mineral supply, metabolites and neuroimmune functions while impacting HD expression [80]. The gut microbiome derived metabolites were differentiated in the pre-HD metabolome, while the symptomatic HD metabolome was mostly influenced by metabolites likely reflecting mHTT toxicity and neurodegeneration [80]. The study suggests that the pre-HD metabolome is influenced more by the gut microbiome than the HD metabolome, possibly due to the increasing effects of mHTT toxicity and neurodegeneration. The understanding of these complex alterations is a delicate balance between the metabolome and gut microbiome in HD, and they relate to disease onset, severity, progression and phenotypic variability in HD are vital questions for future research and clinical studies in HD [80].

Herman et al. [81] investigated the metabolic alterations in tyrosine and phenylalanine pathways in the CSF of HD patients. The study consisted of 13 pre-HD mHTT carriers and 13 symptomatic HD patients and 42 controls. The comparison of symptomatic HD patient’s vs. controls, identified 24 metabolites to be significantly dysregulated, this included the metabolites of Lumichrome, Xanthine, O-succinyl-homoserine, N-acetylproline, Phenylacetate, Isoleucine, L-DOPA, Leucine, Corticosterone, Ophthalmate, Tyrosine, Valine, Salicylate, Phenylalanine and others. Pathway analysis disclosed 5 biochemical pathways affected in symptomatic HD vs. controls namely i) aminoacyl-tRNA biosynthesis; ii) phenylalanine metabolism; iii) valine, leucine and isoleucine biosynthesis; iv) valine, leucine, isoleucine degradation and v) purine metabolism. Phenylalanine metabolism was highly affected in symptomatic HD patients.

Comparing symptomatic HD patient’s vs. pre-HD patients, 28 metabolites were revealed to be significantly altered. Some of the metabolites were, L-DOPA, Xanthine, Ophthalmate, Creatinine, Tyrosine, 5-hydroxytryptophan, Adenosine, Phenylalanine, Phenylacetate, Thyroxine, Glutarylcarnitine, O-succinyl-homoserine, Adenine, Isoleucine, Aldosterone/Cortisone and numerous other metabolites. Fourteen of these were able to distinguish symptomatic HD patient’s from controls. Univariate analysis illustrated 11 metabolites were dysregulated (L-DOPA, xanthine, ophthalmate, creatinine, tyrosine, 5-hydroxytryptophan, adenosine and phenylalanine) remained significant after correcting for multiple comparison testing. Furthermore, 4-acetamidobutanoate and S-adenosylhomocysteine had been corrected for age dependence. There was a notable longitudinal decrease in O-acetylcarnitine in the symptomatic HD patients. Significantly altered abundances of Ophthalmate, Phenylalanine, 4-quinolinecarboxylic and N,N,N-trimethyl lysine were observed in six pre-HD patients. Pathway analysis, illustrated 8 biological pathways of: i) aminoacyl-tRNA biosynthesis; ii) phenylalanine, tyrosine and tryptophan biosynthesis; iii) valine, leucine and isoleucine biosynthesis; iv) tyrosine metabolism; v) nitrogen metabolism; vi) valine, leucine and isoleucine degradation; vii) phenylalanine metabolism and viii) purine metabolism. Tyrosine and phenylalanine metabolism pathways were significantly altered between symptomatic and pre-HD patients.

To investigate the effects of disease progression, the association between CSF concentration of altered metabolites and measure of disease severity were researched in all mHTT carriers and to a five-year risk onset of developing HD in pre-HD patients. Hierarchical clustering revealed that tyrosine metabolism, including tyrosine, thyroxine, L-DOPA and dopamine, was significantly dysregulated in symptomatic vs. pre-HD patients. These metabolites displayed moderate to strong associations of measures to disease severity and symptoms. Furthermore, Thyroxine and Dopamine were also correlated with the five-year risk of onset in pre-HD patients. Phenylalanine and purine metabolism were also significantly altered, but associated with decreased disease severity. Decreased levels of Lumichrome were frequent in mHTT carriers and concentrations correlated with five-year risk of HD onset in pre-HD carriers. Biochemical profiling illustrates that the CSF metabolome may be used to characterize molecular pathogenesis in HD, and may be vital for future development of HD therapies.

Other metabolomics studies conducted in HD patients include, McGarry et al. [82], Mastrokolias et al. [47], Cheng et al. [83], Graham [84, 85] and Patassini et al. [86].

2.4.2 Metabolomics in HD animal models

Targeted metabolic profiling on plasma of a pre-HD transgenic sheep model in order to identify potential candidate biomarkers was investigated by Skene et al. [87].

One hundred and thirty metabolites were obtained, Alanine, Arginine, Citrulline, Glutamine, Glutamate, Glycine, Histidine, Isoleucine, Phenylalanine, Proline, Tryptophan, Valine, Creatine, Kynurenine, Serotonin and several additional metabolites [87]. Citrulline and Arginine showed significantly increased levels in HD compared to control sheep, both are involved in the urea cycle, although Ornithine was also identified and is part of the urea cycle, no significant difference between HD vs. healthy controls was seen. Citrulline demonstrated the most significant change in transgenic HD sheep. Regarding the 20 amino acids measured, 10 had shown significantly decreased concentration in HD sheep, the branched amino acids (BCAA) of Valine, Leucine and Isoleucine showed the most notable effect, which have been previously identified as potential biomarkers. This was followed by Threonine, Tyrosine, Methionine, Alanine, Asparagine, Phenylalanine and Glutamine. Significant alterations in respect to genotype were distinguished in 89/130 identified metabolites, including Sphingolipids, Biogenic amines, amino acids and Urea. A significant increase in Urea, Arginine, Citrulline, asymmetric and symmetric dimethylarginine and a decrease in Sphingolipids [87].

Quantitative enrichment analysis using MetaboAnalyst [19, 20, 21], identified the top five metabolite pathway-associated metabolite sets of: i) aspartate metabolism; ii) arginine and proline metabolism; iii) valine, leucine and isoleucine degradation; iv) fatty acid metabolism and v) urea cycle. The urea cycle and nitric oxide pathways become dysregulated during the early HD stages. Additionally, logistic prediction modeling identified 8 potential biomarkers (Citrulline, Valine, PC aa C40:4, PC aa C36:5, lysoPC a C17:0, SM (OH) C24:1, Threonine, Tetradecenoylcarnitine (C14:1)). In HD sheep, the metabolites of Citrulline, PC aa C36:5 and lysoPC a C17:0 were increased significantly while Valine, PC aa C40:4, SM (OH) C24:1, Threonine and Tetradecenoylcarnitine (C14:1) were significantly decreased compared to control sheep [87]. The degree of sensitivity, using minimally invasive methods, puts forward a novel approach for monitoring disease progression in HD patients.

Andersen et al. [88] investigated the branched amino acids (BCAA) and their metabolism in the cerebral cortex of a R6/2 HD mouse model using metabolomics analysis [88]. Deficiencies in cerebral energy and neurotransmitter are suggested to play a role in neuronal dysfunction in HD. The BCAAs of Valine, Leucine and Isoleucine are vital in cerebral nitrogen homeostasis, neurotransmitter recycling and are utilized as energy substrates in the tricarboxylic acid (TCA) cycle [88]. Decreased levels of BCAAs in HD haven been previously validated in several studies. However, it remains unclear how cerebral BCAA metabolism is regulated in HD.

Isolated cerebral cortical and striatal slices of controls and R6/2 mice were incubated with labeled Leucine and Isoleucine; however, no differences in Leucine or Isoleucine concentration were shown between R6/2 and control striatal or cerebral cortical slices [88]. Suggesting that the cellular uptake of these BCAAs likely remains unaffected in the R6/2 slices compared to controls. Metabolism of Leucine and Isoleucine, entering oxidative metabolism as acetyl CoA, was preserved in R6/2 mice. However, metabolism of Isoleucine, entering the TCA cycle as Succinyl CoA, was increased in the cerebral cortical and striatal slices of R6/2 mice; therefore, suggesting a rise in metabolic influx via the replenishing of Oxaloacetate in the citric acid cycle. Enzyme expression in the BCAA metabolism was assessed, enzymes related to BCAA metabolism displayed an increased expression in the R6/3 brain, particularly enzymes related to Isoleucine metabolism [88]. This indicates that the capacity for cerebral BCAA metabolism, primarily Isoleucine, is heightened in R6/2 brain tissue indicative of alterations in cerebral BCAA homeostasis.

Other metabolomics studies conducted in HD animal models include Chang et al. [89], Chaves et al. [58], Bertrand et al. [90], Verwaest et al. [91], Hashimoto et al. [92], Kumar et al. [93] and Tsang et al. [94].

Advertisement

3. Single cell omics

Organisms and complex tissues are formed by a heterogeneity of cells undergoing cell division, proliferation, differentiation during various physiological states such as development and adulthood [95]. The fate of each cell is intrinsically determined and is influenced both by external factors and by cell–cell interactions [95]. Various processes taking place within individual cells are a result of complex interactions between chromatin, transcripts, proteins, metabolites, lipids and other biological entities [95, 96]. To identify these activity dependent regulatory processes the majority of approaches used include transcriptomics, proteomics, metabolomics or lipidomics methods on tissues, cells and biological fluids [95]. Although the bulk approaches are useful, they have a drawback, as they average the information derived from thousands to millions of cells, leading to masking of cell specific features or features involved in developmental processes [95]. There are some studies of single cell RNA-Seq analysis in HD [97, 98, 99].

Over the past few years, the development of methodological approaches for single cell analysis has greatly increased and addressed the drawback of bulk analysis [95]. Single cell analysis (SCA) has recently been included into multi-omics strategies, which brings together concurrent biological information from different molecular modalities and their relationship with individual cells [95]. Furthermore, analyzing cells individually at a higher resolution results in a more accurate representation of cell–cell variations in comparison to bulk analysis measurements [95]. SCA approaches include: i) single cell genomics, ii) single cell epigenomics iii) single cell transcriptomics, iv) single cell proteomics and v) single cell metabolomics. To fully grasp and understand cellular complexity and specificity of cells, tissues or biological fluid microenvironments in physiological or disease conditions, it is important to measure molecular signatures at the single cell resolution [100]. Benefits of SCA include: i) improvements in experimental design and data analysis for single cells for a disease of interest, ii) targeting of specific cell populations therefore elucidating signaling pathways and networks, iii) cell-to cell communication variation for understanding disease onset, progression and therapeutic response, iv) differentiate normal cells and comprised cells at various developmental stages, v) identify cells and their distinctive susceptibilities, vi) clarify neural communication in unprecedented detail, resulting in new strategies to understand and treat NDs [100]. SCA can provide insight for potential biomarkers, therapeutic targets, pathways and mechanisms in disease involvement [100]. Tables 2 and 3 indicate the databases and tools used for analysis of SCA, while Table 4 illustrates software and databases for cell-to-cell communication.

NameTypeOmics contentRefWebsite (If Available)
Gene expression omnibusRepositorySingle cell RNA-Seq[101]https://www.ncbi.nlm.nih.gov/geo/
Single cell epression atlasRepositorySingle cell RNA-Seq[102]https://www.ebi.ac.uk/gxa/home
Slavov laboratory/Quantitative biologyRepository/DatabaseSingle cell Proteomics[103]https://slavovlab.net/data_webs.htm
Proteome exchangeRepository/DatabaseSingle cell Proteomics[104, 105]https://www.proteomexchange.org/
Metabolomics workbenchRepository/DatabaseSingle cell metabolomics[106]https://www.metabolomicsworkbench.org/data/index.php

Table 2.

Single cell repositories and databases.

NameTypeOmics contentRefWebsite (If Available)
SeuratR packageSingle cell RNA-Seq[107, 108, 109]https://satijalab.org/seurat/
GalaxyOpen accessSingle cell RNA-Seq[11]https://usegalaxy.org/
scpdataR packageSingle cell Proteomicshttps://github.com/UCLouvain-CBIO/scpdata

Table 3.

Online tools, software and R-packages for single cell omics analysis.

NameTypeOmics contentRefWebsite (If Available)
CellChatDBOpen access & R packageSingle cell RNA-Seq[110]http://www.cellchat.org/
NICHESR packageSingle cell RNA-Seq[111]https://github.com/msraredon/NICHES
CITEdbOpen access & R packageSingle cell RNA-Seq[112]https://citedb.cn/#/index
CellPhoneDBOpen accessSingle cell RNA-Seq[113]https://www.cellphonedb.org/

Table 4.

Software and databases for cell-to-cell communication.

Advertisement

4. Bioinformatics

In recent years, technological advancements have made great progress in understanding the genetics, transcripts, proteins and metabolites seen in disease, resulting in an explosion of big data, opening endless scientific possibilities [114]. However, this vast amount of data generated has its own challenges related to: i) data storage, ii) software to process such large amounts of data and ii) analysis and biological interpretation of data. In this circumstance, bioinformatics and computational biology have sought to overcome these challenges in big data generation and analysis [114]. Bioinformatics is a multi-interdisciplinary approach combining mathematics, physics, biology and computer science, it is defined as the application of computational methods and tools for the organization, analysis, understanding, visualization and storage of information associated with biological entities [114]. The development of high-throughput technologies such as NGS, RNA-Seq and liquid chromatography-mass spectrometry (LC–MS) and the analysis of data using bioinformatic approaches has opened a host of new possibilities including but not limited to, gene expression studies, methylation patterns, epigenetic markers, proteins, metabolites, lipids and others [114]. In the recent decade, bioinformatics approaches have become of increasing interest especially in NDs for novel biomarkers and drug discovery and pathways [6]. Specifically, a multi-interdisciplinary approach such as SB can enhance the contribution of computational therapeutics and diagnostics for NDs, hence providing a stepping stone towards precision medicine.

In the following section, we look into the tools and databases available for.

-omics in regards to obtaining data and analysis of said data. Moreover, we will discuss network-based approaches used in HD research.

4.1 Omics databases and tools

The databases for obtaining publicly available data of different omics data at the single and bulk -omics level is constantly changing with new data becoming available every day. In addition, the analysis of such data is increasingly important, and various tools, software and R-packages are also developed to assist researchers in the analysis of their data. Table 5 includes repositories and databases where omics data can be found, not only specific for HD but also for other NDs, as well various diseases. Table 1 illustrates the tools, software and packages mainly in R, used for analysis of -omics data. The tables contain some of the most popular databases, repositories, software, tools and R-packages used, however there are numerous others resources available which have not been listed.

NameTypeOmics contentRefWebsite (If Available)
Gene expression omnibusRepositoryGenomics, Epigenomics, Transcriptomics,[101]https://www.ncbi.nlm.nih.gov/geo/
Expression atlasRepositoryGenomics[102, 115]https://www.ebi.ac.uk/gxa/home
Array expressRepository/DatabaseGenomics[116]https://www.ebi.ac.uk/biostudies/arrayexpress
European variation archiveRepository/DatabaseGenomics[117]https://www.ebi.ac.uk/eva/?Home
EnsemblDatabaseGenomics[118]http://www.ensembl.org/index.html
Database of genomic variants archiveDatabaseGenomicshttps://www.ebi.ac.uk/dgva/
European genome-phenome archiveRepository/DatabaseGenomicshttps://ega-archive.org/
Answer ALSRepositoryGenomics Transcriptomics, Proteomics, Metabolomics[119]https://dataportal.answerals.org/home
Huntington’s disease in high definition (HDinHD)DatabaseGenomicshttps://www.hdinhd.org/
Enroll HDRepository/DatabaseGenomics Transcriptomics, Proteomics[120]https://www.enroll-hd.org/
Parkinson’s progression markers initiativeRepository/DatabaseGenomics Transcriptomics, Proteomics, Metabolomics[121]https://www.ppmi-info.org/
Alzheimer’s disease neuroimaging initiativeRepository/DatabaseGenetics
Radiomics
[122]https://adni.loni.usc.edu/
PRIDEDatabaseProteomics[123]https://www.ebi.ac.uk/pride/
Proteome exchangeRepository/DatabaseProteomics[104, 105]https://www.proteomexchange.org/
Japan proteome standardRepository/DatabaseProteomics[124]https://jpostdb.org/
Mass spectrometry interactive virtual environmentRepository/DatabaseProteomicshttps://massive.ucsd.edu/ProteoSAFe/static/massive.jsp
ProteomicsDBRepository/DatabaseProteomics[125]https://www.proteomicsdb.org/
Human peptide atlasRepository/DatabaseProteomics[126]https://peptideatlas.org/
Metabolomics workbenchRepository/DatabaseMetabolomics[106]https://www.metabolomicsworkbench.org/data/index.php
MetaboLightsRepository/DatabaseMetabolomics[127]https://www.ebi.ac.uk/metabolights/index
Metabolome exchangeRepository/DatabaseMetabolomicshttp://www.metabolomexchange.org/site/
Human metabolome databaseDatabaseMetabolomics[128]https://hmdb.ca/

Table 5.

Omics repositories and databases.

4.2 Network-based approaches

In all living organisms, most cellular components exert their functions via interactions with other components, the entirety of these interactions represents the human interactome [129]. The cells and their response to changes in their environment involve coordinated activity of mRNAs, proteins, metabolites and lipids [130]. The fundamentals of proper cellular function are molecular networks connecting these components to process extra-cellular environmental signals driving dynamic cellular responses [130]. Network-based approaches aim to systematically integrate measurements obtained from high-throughput experiments to gain insight of the cellular functions undergoing changes resulting in disease [130]. Systematic integration of varying biological entities is essential to identify molecular networks controlling normal and disease states, and in time, predict complex phenotypes from molecular markers [130]. Network-based approaches in human disease can lead to multiple biological and clinical applications, over the past decade, there has been an exceptional increase in human-specific molecular interaction data, resulting in a greater understanding of how different biological entities interact with one another, in biological networks and how they play a role in human diseases [129]. Examples of molecular networks are i) PPI networks, ii) metabolic networks, iii) regulatory networks, iv) RNA networks, v) cell–cell interaction networks and vi) multi-omics networks [129]. Few studies have been performed in regards to network and bioinformatic based approaches on HD. Some of these studies will be discussed below.

Chandrasekaran and Bonchev [131], performed network analysis on post-mortem tissue of the cerebellum, frontal cortex and caudate nucleus of HD patients. The microarray dataset, GSE3790, has 44 and 36 HD patients and controls respectively [131]. The dataset consists of both Affymetrix GeneChip Human Genome HG-U133A and B [131]. The seed genes are referred to as the significantly differentially expressed genes (SDEGs) [131]. In the GSE3790 HG-U133A, 617 overlapping seed genes were found between the four sets of SDEGs, while in GSE3790 HG-U133B, 351 seed genes were found [131]. Altogether, 925 seed genes were identified; however, network evaluation was performed only for the SDEGs with a p-value <0.01, with this new cut-off threshold, 531 seed genes were identified and underwent network analysis [131].

Pathway enrichment was performed using Pathway Studio 9.0 software along with the molecular interaction database ResNet 9.0 [131], to construct direct interaction, shortest-path and miRNA regulation networks. Gene prioritizing approaches based on network topological measures, high node connectivity, centrality and guilt by association were applied, based on this approach 19 novel genes were found CEBPA, CDK1, CX3CL1, EGR1, E2F1, ERBB2, LRP1, HSP90AA1 and ZNF148; these genes may be of particular interest to undergo experimental validation [131]. The seed genes underwent GO enrichment analysis using DAVID [131], while the IPA software and Pathway Enrichment Analysis in Pathway studio was used to explore the canonical pathways affected in HD [131]. The pathways identified with DAVID are i) neuron development (GO:0048666), ii) neuron differentiation (GO:0030182), iii) regulation of glucose import (GO:0046324), iv) neuron projection (GO:0043005), v) regulation of lipid catabolic process (GO: 0050994) and various additional GO terms. The KEGG pathways from DAVID are i) HD (hsa: 05016), ii) MAPK signaling (hsa:04010), iii) ErbB signaling (hsa:04012), iv) Alzheimer’s Disease (hsa: 05010), v) Amyotrophic Lateral Sclerosis (hsa: 05014) and numerous other pathways [131]. The miRNA regulatory network analysis, found miR-124, mir-135a, miR-141, miR-182 and miR-19a to be the top five scoring miRNA within the network [131]. The SDEGs, miRNA and pathways obtained, in combination with experimental validation can shed light onto possible genes, miRNAs and mechanisms affected in HD, which can lead to targeted therapeutic strategies.

Other network and bioinformatics-based approaches conducted in HD include Sneha et al. [132], Pirhaji et al. [133], Pradhan et al. [134], Xiang et al. [135], Zaho et al. [136], Fu et al. [137], Shirasaki et al. [138], Christodoulou [139, 140, 141], and Onisiforou [142].

Advertisement

5. Discussion and conclusion

HD remains one of the most debilitating and incurable ND, as currently there is a lack of effective treatments, although clinical trials have been conducted using different strategies such as gene silencing to decrease mHTT protein production [1, 2, 3, 5]. However, these attempts were unsuccessful as in some cases, no difference between the drug and placebo groups or no symptom improvement was observed. Therefore, there is a need for effective drugs to be identified and tested and used as pharmacotherapies for HD. Furthermore, a better understanding of how different biological molecules (genes, proteins, metabolites, lipids etc.) interact with not only each other but also with disease pathways, and possibly drugs is vital to understand how these interactions are affected in HD as it will allow to shed light on why some therapeutic treatments are not effective [5]. Dysregulation, in any specific cell type, biological molecule or pathway can influence the entire biological system leading to disease progression [7].

In the past decade technological advancements has led to high-throughput experiments resulting in -omics data and their analysis. In addition, there is a growing interest in SC-omics, allowing analysis of individual cells at a higher resolution, allowing for the accurate representation of cell–cell variation in tissues [95]. Bioinformatics, specifically network-based approaches, can be applied to investigate and understand in depth the relationship between the different biological molecules and their interaction with the HTT protein, each other and within pathways [5]. The advancement of bioinformatics has led to the progress and identification of novel candidate biomarkers and drugs, affected pathway and mechanisms and genes, proteins and metabolites affected in a disease state compared to controls [5, 6]. In addition, there has been an exponential increase in the number of repositories and database available for both bulk and SC analysis for genomics, transcriptomics, proteomics and metabolomics [95, 100]. In addition, this has resulted in numerous bioinformatics tools, software and R-packages, which have been developed to assist researchers in omics data analysis, in order to have meaningful biological interpretation of the data to make proper conclusions that will possibly lead to the discovery of potential biomarkers and drugs resulting in better prognosis, diagnosis and drug sensitivity of patients [100, 114]. The contribution of SB can provide a stepping stone towards precision medicine and potentially address the absence of HD treatments.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Additional information

The Cyprus Institute of Neurology and Genetics is a full member of the european reference network for rare neurological diseases (ERN-RND).

References

  1. 1. Roos RA. Huntington’s disease: A clinical review. Orphanet Journal of Rare Diseases. 2010;5:40. DOI: 10.1186/1750-1172-5-40
  2. 2. Novak MJU, Tabrizi SJ. CliniCal review Huntington’s disease. BMJ. 2010:340. DOI: 10.1136/bmj.c3109
  3. 3. Bates GP, Dorsey R, Gusella JF, Hayden MR, Kay C, Leavitt BR, et al. Huntington Disease; 2015
  4. 4. Lanciego JL, Luquin N, Obeso JA. Functional neuroanatomy of the basal ganglia. Cold Springer Harbor Perspective Medicine. 2012;2:1-20. DOI: 10.1101/cshperspect.a009621
  5. 5. Paananen J. Bioinformatics in the identification of novel targets and pathways in neurodegenerative diseases. Current Genetics in Medical Report. 2017;5:15-21. DOI: 10.1007/s40142-017-0115-8
  6. 6. Oulas A, Minadakis G, Zachariou M, Sokratous K, Bourdakou MM, Spyrou GM. Systems bioinformatics: Increasing precision of Computational Diagnostics and therapeutics through network-based approaches. Briefings in Bioinformatics. 2019;20:806-824. DOI: 10.1093/bib/bbx151
  7. 7. Diaz-Ortiz ME, Chen-Plotkin AS. Omics in neurodegenerative disease: Hope or hype? Trends in Genetics. 2020;36:152-159
  8. 8. Micheel C, Nass SJ, Omenn GS. Institute of Medicine (U.S.). Committee on the Review of Omics-Based Tests for Predicting Patient Outcomes in Clinical Trials Evolution of Translational Omics : Lessons Learned and the Path Forward; ISBN 9780309224185
  9. 9. Chen C, McGarvey PB, Huang H, Wu CH. Protein bioinformatics infrastructure for the integration and analysis of multiple high-throughput omics data. Advances in Bioinformatics. 2010;2010:1-19. DOI: 10.1155/2010/423589
  10. 10. Paszkiewicz KH, Giezen M. Bioinformatics, and infectious disease research. In: Genetics and Evolution of Infectious Diseases. Amsterdam, The Netherlands: Elsevier Inc.; 2011. pp. 523-539
  11. 11. Afgan E, Nekrutenko A, Grüning BA, Blankenberg D, Goecks J, Schatz MC, et al. The galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2022 update. Nucleic Acids Research. 2022;50:W345-W351. DOI: 10.1093/nar/gkac247
  12. 12. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research. 2015;43:e47. DOI: 10.1093/nar/gkv007
  13. 13. Robinson MD, McCarthy DJ, Smyth GK. EdgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2009;26:139-140. DOI: 10.1093/bioinformatics/btp616
  14. 14. Chen Y, Lun ATL, Smyth GK. From reads to genes to pathways: Differential expression analysis of RNA-Seq experiments using Rsubread and the EdgeR quasi-likelihood pipeline. F1000Res. 2016;5:1438. DOI: 10.12688/f1000research.8987.1
  15. 15. McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Research. 2012;40:4288-4297. DOI: 10.1093/nar/gks042
  16. 16. Tyanova S, Temu T, Cox J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nature Protocols. 2016;11:2301-2319. DOI: 10.1038/nprot.2016.136
  17. 17. Sturm M, Bertsch A, Gröpl C, Hildebrandt A, Hussong R, Lange E, et al. OpenMS - An open-source software framework for mass spectrometry. BMC Bioinformatics. 2008;9:1-11. DOI: 10.1186/1471-2105-9-163
  18. 18. Rainer J, Vicini A, Salzer L, Stanstrup J, Badia JM, Neumann S, et al. A modular and expandable ecosystem for metabolomics data annotation in R. Metabolites. 2022;12:1-13. DOI: 10.3390/metabo12020173
  19. 19. Xia J, Wishart DS. Metabolomic data processing, analysis, and interpretation using metaboanalyst. Current Protocols in Bioinformatics. 2011;34(1);1-48. DOI: 10.1002/0471250953.bi1410s34
  20. 20. Xia J, Sinelnikov IV, Han B, Wishart DS. MetaboAnalyst 3.0-making metabolomics more meaningful. Nucleic Acids Research. 2015;2015:1-7. DOI: 10.1093/nar/gkv380
  21. 21. Chong J, Soufan O, Li C, Caraus I, Li S, Bourque G, et al. MetaboAnalyst 4.0: Towards more transparent and integrative metabolomics analysis. Nucleic Acids Research. 2018;46:W486-W494. DOI: 10.1093/nar/gky310
  22. 22. Caspi R, Foerster H, Fulcher CA, Kaipa P, Krummenacker M, Latendresse M, et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Research. 2008;36:623-631. DOI: 10.1093/nar/gkm900
  23. 23. Huan T, Forsberg EM, Rinehart D, Johnson CH, Ivanisevic J, Benton HP, et al. Systems biology guided by XCMS online metabolomics. Nature Methods. 2017;2017:1-5
  24. 24. Tautenhahn R, Patti GJ, Rinehart D, Siuzdak G. XCMS online: A web-based platform to process untargeted metabolomic data. Analytical Chemistry. 2012;84:5035-5039. DOI: 10.1021/ac300698c
  25. 25. Smith CA, Want EJ, O’Maille G, Abagyan R, Siuzdak G. XCMS: Processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Analytical Chemistry. 2006;78:779-787. DOI: 10.1021/ac051437y
  26. 26. Myers OD, Sumner SJ, Li S, Barnes S, Du X. Detailed investigation and comparison of the XCMS and MZmine 2 chromatogram construction and chromatographic peak detection methods for Preprocessing mass spectrometry metabolomics data. Analytical Chemistry. 2017;2017:8689-8695. DOI: 10.1021/acs.analchem.7b01069
  27. 27. Moss DJH, Tabrizi SJ, Mead S, Lo K, Pardiñas AF, Holmans P, et al. Identification of genetic variants associated with Huntington’s disease progression: A genome-wide association study. Lancet Neurology. 2017;16:701-711. DOI: 10.1016/S1474-4422(17)30161-8
  28. 28. Li S, Tollefsbol TO. DNA Methylation Methods: Global DNA Methylation and Methylomic Analyses. Vol. 187. Amsterdam, The Netherlands: Elsevier Inc; 2021 ISBN 2059344573
  29. 29. Tang B, Seredenina T, Coppola G, Kuhn A, Geschwind DH, Luthi-Carter R, et al. Gene expression profiling of R6/2 transgenic mice with different CAG repeat lengths reveals genes associated with disease onset and progression in Huntington’s disease. Neurobiology of Disease. 2011;42:459-467. DOI: 10.1016/j.nbd.2011.02.008
  30. 30. Saiki K, David GH, Susanne S, Stephen SJ, Russell H, Glenn HT, et al. Henry primer-directed enzymatic amplification of DNA with thermostable DNA polymerase. Science. 1979;1988(239):487-491
  31. 31. Behjati S, Tarpey PS. What is next generation sequencing? Archives of Disease in Childhood. Education and Practice Edition. 2013;98:236-238. DOI: 10.1136/archdischild-2013-304340
  32. 32. Rossi MJ, Kuntala PK, Lai WKM, Yamada N, Badjatia N, Mittal C, et al. A high-resolution protein architecture of the budding yeast genome. Nature. 2021;592:309-314. DOI: 10.1038/s41586-021-03314-8
  33. 33. Roth SC. What is genomic medicine? Journal of the Medical Library Association. 2019;107:442-448. DOI: 10.5195/jmla.2019.604
  34. 34. Khodadadian A, Darzi S, Haghi-Daredeh S, Eshaghi FS, Babakhanzadeh E, Mirabutalebi SH, et al. Genomics and transcriptomics: The powerful Technologies in Precision Medicine. International Journal of Genetic Medicine. 2020;13:627-640
  35. 35. Marchina E, Misasi S, Bozzato A, Ferraboli S, Agosti C, Rozzini L, et al. Gene expression profile in fibroblasts of Huntington’s disease patients and controls. Journal of the Neurological Sciences. 2014;337:42-46. DOI: 10.1016/j.jns.2013.11.014
  36. 36. Wright GEB, Caron NS, Ng B, Casal L, Casazza W, Xu X, et al. Gene expression profiles complement the analysis of genomic modifiers of the clinical onset of Huntington disease. Human Molecular Genetics. 2020;29:2788-2802. DOI: 10.1093/hmg/ddaa184
  37. 37. Langfelder P, Cantle JP, Chatzopoulou D, Wang N, Gao F, Al-Ramahi I, et al. Integrated genomics and proteomics define huntingtin CAG length-dependent networks in mice. Nature Neuroscience. 2016;19:623-633. DOI: 10.1038/nn.4256
  38. 38. Choudhury KR, Das S, Bhattacharyya NP. Differential proteomic and genomic profiling of mouse striatal cell model of Huntington’s disease and control; probable implications to the disease biology. Journal of Proteomics. 2016;132:155-166. DOI: 10.1016/j.jprot.2015.11.007
  39. 39. Alcalá-Vida R, Seguin J, Lotz C, Molitor AM, Irastorza-Azcarate I, Awada A, et al. Age-related and disease locus-specific mechanisms contribute to early remodelling of chromatin structure in Huntington’s disease mice. Nature Communications. 2021;12:1-16. DOI: 10.1038/s41467-020-20605-2
  40. 40. Lin L, Park JW, Ramachandran S, Zhang Y, Tseng YT, Shen S, et al. Transcriptome sequencing reveals aberrant alternative splicing in Huntington’s disease. Human Molecular Genetics. 2016;25:3454-3466. DOI: 10.1093/hmg/ddw187
  41. 41. Neueder A, Kojer K, Hering T, Lavery DJ, Chen J, Birth N, et al. Abnormal molecular signatures of inflammation, energy metabolism, and vesicle biology in human Huntington disease peripheral tissues. Genome Biology. 2022;23:1-21. DOI: 10.1186/s13059-022-02752-5
  42. 42. Miller JRC, Lo KK, Andre R, Hensman Moss DJ, Träger U, Stone TC, et al. RNA-Seq of Huntington’s disease patient myeloid cells reveals innate transcriptional dysregulation associated with proinflammatory pathway activation. Human Molecular Genetics. 2016;25:2893-2904. DOI: 10.1093/hmg/ddw142
  43. 43. Moily NS, Ormsby AR, Stojilovic A, Ramdzan YM, Diesch J, Hannan RD, et al. Transcriptional profiles for distinct aggregation states of mutant huntingtin exon 1 protein unmask New Huntington’s disease pathways. Molecular and Cellular Neuroscience. 2017;83:103-112. DOI: 10.1016/j.mcn.2017.07.004
  44. 44. Seefelder M, Kochanek S. A Meta-analysis of transcriptomic profiles of Huntington’s disease patients. PLoS One. 2021;16. DOI: 10.1371/journal.pone.0253037
  45. 45. Sinha M, Mukhopadhyay S, Bhattacharyya NP. Mechanism(s) of alteration of Micro Rna expressions in Huntington’s disease and their possible contributions to the observed cellular and molecular dysfunctions in the disease. Neuromolecular Medicine. 2012;14:221-243
  46. 46. Mastrokolias A, Ariyurek Y, Goeman JJ, Van Duijn E, Roos RAC, Van Der Mast RC, et al. Huntington’s disease biomarker progression profile identified by transcriptome sequencing in peripheral blood. European Journal of Human Genetics. 2015;23:1349-1356. DOI: 10.1038/ejhg.2014.281
  47. 47. Mastrokolias A, Pool R, Mina E, Hettne KM, van Duijn E, van der Mast RC, et al. Integration of targeted metabolomics and transcriptomics identifies deregulation of phosphatidylcholine metabolism in Huntington’s disease peripheral blood samples. Metabolomics. 2016;12. DOI: 10.1007/s11306-016-1084-8
  48. 48. Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, Rosas HD, et al. Genome-wide expression profiling of human blood reveals biomarkers for Huntington’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:11023-11028. DOI: 10.1073/pnas.0504921102
  49. 49. Cha JHJ. Transcriptional signatures in Huntington’s disease
  50. 50. Mehta SR, Tom CM, Wang Y, Bresee C, Rushton D, Mathkar PP, et al. Human Huntington’s disease IPSC-derived cortical neurons display altered transcriptomics, morphology, and maturation. Cell Reports. 2018;25:1081-1096.e6. DOI: 10.1016/j.celrep.2018.09.076
  51. 51. Stopa EG, Tanis KQ , Miller MC, Nikonova EV, Podtelezhnikov AA, Finney EM, et al. Comparative transcriptomics of choroid plexus in Alzheimer’s disease, frontotemporal dementia and Huntington’s disease: Implications for CSF homeostasis. Fluids Barriers CNS. 2018;15. DOI: 10.1186/s12987-018-0102-9
  52. 52. Runne H, Kuhn A, Wild EJ, Pratyaksha W, Kristiansen M, Isaacs JD, et al. Analysis of potential transcriptomic biomarkers for huntington’s disease in peripheral blood. Proceedings of the National Academy of Sciences of the United States of America. 4 Sep 2007;104(36):1-6. DOI: 10.1073/pnas.0703652104
  53. 53. Jin J, Cheng Y, Zhang Y, Wood W, Peng Q , Hutchison E, et al. Interrogation of brain MiRNA and MRNA expression profiles reveals a molecular regulatory network that is perturbed by mutant huntingtin. Journal of Neurochemistry. 2012;123:477-490. DOI: 10.1111/j.1471-4159.2012.07925.x
  54. 54. Hervás-Corpión I, Guiretti D, Alcaraz-Iborra M, Olivares R, Campos-Caro A, Barco Á, et al. Early alteration of epigenetic-related transcription in Huntington’s disease mouse models. Scientific Reports. 2018;8. DOI: 10.1038/s41598-018-28185-4
  55. 55. Bensalel J, Xu H, Lu ML, Capobianco E, Wei J. RNA-Seq analysis reveals significant transcriptome changes in huntingtin-null human neuroblastoma cells. BMC Medical Genomics. 2021;14. DOI: 10.1186/s12920-021-01022-w
  56. 56. Dickson E, Dwijesha AS, Andersson N, Lundh S, Björkqvist M, Petersén Å, et al. Microarray profiling of hypothalamic gene expression changes in Huntington’s disease mouse models. Frontiers in Neuroscience. 2022;16. DOI: 10.3389/fnins.2022.1027269
  57. 57. Reyes-Ortiz AM, Abud EM, Burns MS, Wu J, Hernandez SJ, McClure N, et al. Single-nuclei transcriptome analysis of Huntington disease IPSC and mouse astrocytes implicates maturation and functional deficits. iScience. 2023;26. DOI: 10.1016/j.isci.2022.105732
  58. 58. Chaves G, Özel RE, Rao NV, Hadiprodjo H, Da Costa Y, Tokuno Z, et al. Metabolic and transcriptomic analysis of Huntington’s disease model reveal changes in intracellular glucose levels and related genes. Heliyon. 2017;3:e00381. DOI: 10.1016/j.heliyon.2017
  59. 59. Huang L, Fang L, Liu Q , Torshizi AD, Wang K. Integrated analysis on transcriptome and Behaviors defines HTT repeat-dependent network modules in Huntington’s disease. Genes Diseases. 2022;9:479-493. DOI: 10.1016/j.gendis.2021.05.004
  60. 60. Al-Amrani S, Al-Jabri Z, Al-Zaabi A, Alshekaili J, Al-Khabori M. Proteomics: Concepts and applications in human medicine. World Journal of Biological Chemistry. 2021;12:57-69. DOI: 10.4331/wjbc.v12.i5.57
  61. 61. Fang Q , Strand A, Law W, Faca VM, Fitzgibbon MP, Hamel N, et al. Brain-specific proteins decline in the cerebrospinal fluid of humans with Huntington disease. Molecular and Cellular Proteomics. 2009;8:451-466. DOI: 10.1074/mcp.M800231-MCP200
  62. 62. Chen S, Lu FF, Seeman P, Liu F. Quantitative proteomic analysis of human substantia Nigra in Alzheimer’s disease, Huntington’s disease and multiple sclerosis. Neurochemical Research. 2012;37:2805-2813. DOI: 10.1007/s11064-012-0874-2
  63. 63. Schönberger SJ, Jezdic D, Faull RLM, Cooper GJS. Proteomic analysis of the human brain in Huntington’s disease indicates pathogenesis by molecular processes linked to other neurodegenerative diseases and to Type-2 diabetes. Journal of Huntingtons Diseases. 2013;2:89-99. DOI: 10.3233/JHD-120044
  64. 64. Sorolla MA, Reverter-Branchat G, Tamarit J, Ferrer I, Ros J, Cabiscol E. Proteomic and oxidative stress analysis in human brain samples of Huntington disease. Free Radical Biology & Medicine. 2008;45:667-678. DOI: 10.1016/j.freeradbiomed.2008.05.014
  65. 65. Chae J et al. Quantitative proteomic analysis of induced pluripotent stem cells derived from a human Huntington’s disease patient. Biochemical Journal. 2012;446:359-371. DOI: 10.1042/BJ20111495
  66. 66. McQuade LR, Balachandran A, Scott HA, Khaira S, Baker MS, Schmidt U. Proteomics of Huntington’s disease-affected human embryonic stem cells reveals an evolving pathology involving mitochondrial dysfunction and metabolic disturbances. Journal of Proteome Research. 2014;13:5648-5659. DOI: 10.1021/pr500649m
  67. 67. Dalrymple A, Wild EJ, Joubert R, Sathasivam K, Björkqvist M, Petersén Å, et al. Proteomic profiling of plasma in Huntington’s disease reveals neuroinflammatory activation and biomarker candidates. Journal of Proteome Research. 2007;6:2833-2840. DOI: 10.1021/pr0700753
  68. 68. Agrawal S, Fox JH. Novel proteomic changes in brain mitochondria provide insights into mitochondrial dysfunction in mouse models of Huntington’s disease. Mitochondrion. 2019;47:318-329. DOI: 10.1016/j.mito.2019.03.004
  69. 69. Mees I, Li S, Tran H, Ang CS, Williamson NA, Hannan AJ, et al. Phosphoproteomic dysregulation in Huntington’s disease mice is rescued by environmental enrichment. Brain Communication. 2022;4. DOI: 10.1093/braincomms/fcac305
  70. 70. Liu X, Miller BR, Rebec GV, Clemmer DE. Protein expression in the striatum and cortex regions of the brain for a mouse model of Huntington’s disease. Journal of Proteome Research. 2007;6:3134-3142. DOI: 10.1021/pr070092s
  71. 71. Perluigi M, Poon HF, Maragos W, Pierce WM, Klein JB, Calabrese V, et al. Proteomic analysis of protein expression and oxidative modification in R6/2 transgenic mice: A model of Huntington disease. Molecular and Cellular Proteomics. 2005;4:1849-1861. DOI: 10.1074/mcp.M500090-MCP200
  72. 72. Deschepper M, Hoogendoorn B, Brooks S, Dunnett SB, Jones L. Proteomic changes in the brains of Huntington’s disease mouse models reflect pathology and implicate mitochondrial changes. Brain Research Bulletin. 2012;88:210-222. DOI: 10.1016/j.brainresbull.2011.01.012
  73. 73. Culver BP, Savas JN, Park SK, Choi JH, Zheng S, Zeitlin SO, et al. Proteomic analysis of Wild-type and mutant huntingtin-associated proteins in mouse brains identifies unique interactions and involvement in protein synthesis. Journal of Biological Chemistry. 2012;287:21599-21614. DOI: 10.1074/jbc.M112.359307
  74. 74. Cozzolino F, Landolfi A, Iacobucci I, Monaco V, Caterino M, Celentano S, et al. New label-free methods for protein relative quantification applied to the investigation of an animal model of Huntington disease. PLoS One. 2020;15:1-20. DOI: 10.1371/journal.pone.0238037
  75. 75. Vodicka P, Mo S, Tousley A, Green KM, Sapp E, Iuliano M, et al. Mass spectrometry analysis of Wild-type and Knock-in Q140/Q140 Huntington’s disease mouse brains reveals changes in glycerophospholipids including alterations in phosphatidic acid and Lyso-phosphatidic acid. Journal of Huntingtons Diseases. 2015;4:187-201. DOI: 10.3233/JHD-150149
  76. 76. Sap KA, Guler AT, Bezstarosti K, Bury AE, Juenemann K, Demmers JAA, et al. Global proteome and ubiquitinome changes in the soluble and insoluble fractions of Q175 Huntington mice brains. Molecular and Cellular Proteomics. 2019;18:1705-1720. DOI: 10.1074/mcp.RA119.001486
  77. 77. Ratovitski T, Chighladze E, Arbez N, Boronina T, Herbrich S, Cole RN, et al. Huntingtin protein interactions altered by polyglutamine expansion as determined by quantitative proteomic analysis. Cell Cycle. 2012;11:2006-2021. DOI: 10.4161/cc.20423
  78. 78. Zabel C, Klose J. Influence of huntington’s disease on the human and mouse proteome. International Review of Neurobiology. 2004;61:241-283. DOI: 10.1016/S0074-7742(04)61010-5
  79. 79. Clish CB. Metabolomics: An emerging but powerful tool for precision medicine. Molecular Case Studies. 2015;1:a000588. DOI: 10.1101/mcs.a000588
  80. 80. Rosas HD, Doros G, Bhasin S, Thomas B, Gevorkian S, Malarick K, et al. A systems-level “Misunderstanding”: The plasma metabolome in Huntington’s disease. Annals of Clinical Translational Neurology. 2015;2:756-768. DOI: 10.1002/acn3.214
  81. 81. Herman S, Niemelä V, Emami Khoonsari P, Sundblom J, Burman J, Landtblom AM, et al. Alterations in the tyrosine and phenylalanine pathways revealed by biochemical profiling in cerebrospinal fluid of Huntington’s disease subjects. Scientific Reports. 2019;9:1-13. DOI: 10.1038/s41598-019-40186-5
  82. 82. McGarry A, Gaughan J, Hackmyer C, Lovett J, Khadeer M, Shaikh H, et al. Cross-sectional analysis of plasma and CSF metabolomic markers in Huntington’s disease for participants of varying functional disability: A pilot study. Scientific Reports. 2020;10:1-13. DOI: 10.1038/s41598-020-77526-9
  83. 83. Cheng ML, Chang KH, Wu YR, Chen CM. Metabolic disturbances in plasma as biomarkers for Huntington’s disease. Journal of Nutritional Biochemistry. 2016;31:38-44. DOI: 10.1016/j.jnutbio.2015.12.001
  84. 84. Graham SF, Pan X, Yilmaz A, Macias S, Robinson A, Mann D, et al. Targeted biochemical profiling of brain from Huntington’s disease patients reveals novel metabolic pathways of interest. Biochimica et Biophysica Acta - Molecular Basis of Disease. 2018;1864:2430-2437. DOI: 10.1016/j.bbadis.2018.04.012
  85. 85. Graham SF, Kumar P, Bahado- Singh RO, Robinson A, Mann D, Green BD. Novel metabolite biomarkers of Huntington’s disease as detected by high-resolution mass spectrometry. Journal of Proteome Research. 2016;15:1592-1601. DOI: 10.1021/acs.jproteome.6b00049
  86. 86. Patassini S, Begley P, Xu J, Church SJ, Reid SJ, Kim EH, et al. Metabolite mapping reveals severe widespread perturbation of multiple metabolic processes in Huntington’s disease human brain. Biochimica et Biophysica Acta - Molecular Basis of Disease. 2016;1862:1650-1662. DOI: 10.1016/j.bbadis.2016.06.002
  87. 87. Skene DJ, Middleton B, Fraser CK, Pennings JLA, Kuchel TR, Rudiger SR, et al. Metabolic profiling of Presymptomatic Huntington’s disease sheep reveals novel biomarkers. Scientific Reports. 2017;7:1-16. DOI: 10.1038/srep43030
  88. 88. Andersen JV, Skotte NH, Aldana BI, Nørremølle A, Waagepetersen HS. Enhanced cerebral branched-chain amino acid metabolism in R6/2 mouse model of Huntington’s disease. Cellular and Molecular Life Sciences. 2019;76:2449-2461. DOI: 10.1007/s00018-019-03051-2
  89. 89. Chang KL, New LS, Mal M, Goh CW, Aw CC, Browne ER, et al. Metabolic profiling of 3-Nitropropionic acid early-stage Huntingtons disease rat model using gas chromatography time-of-flight mass spectrometry. Journal of Proteome Research. 2011;10:2079-2087. DOI: 10.1021/pr2000336
  90. 90. Bertrand M, Decoville M, Meudal H, Birman S, Landon C. Metabolomic nuclear magnetic resonance studies at Presymptomatic and symptomatic stages of Huntington’s disease on a Drosophila model. Journal of Proteome Research. 2020;19:4034-4045. DOI: 10.1021/acs.jproteome.0c00335
  91. 91. Verwaest KA, Vu TN, Laukens K, Clemens LE, Nguyen HP, Van Gasse B, et al. 1H NMR based metabolomics of CSF and blood serum: A metabolic profile for a transgenic rat model of Huntington disease. Biochimica et Biophysica Acta - Molecular Basis of Disease. 2011;1812:1371-1379. DOI: 10.1016/j.bbadis.2011.08.001
  92. 92. Hashimoto M, Watanabe K, Miyoshi K, Koyanagi Y, Tadano J, Miyawaki I. Multiplatform metabolomic analysis of the R6/2 mouse model of Huntington’s disease. FEBS Open Bio. 2021;11:2807-2818. DOI: 10.1002/2211-5463.13285
  93. 93. Kumar KK, Goodwin CR, Uhouse MA, Bornhorst J, Schwerdtle T, Aschner M, et al. Untargeted metabolic profiling identifies interactions between Huntington’s disease and neuronal manganese status. Metallomics. 2015;7:363-370. DOI: 10.1039/c4mt00223g
  94. 94. Tsang TM, Woodman B, Mcloughlin GA, Griffin JL, Tabrizi SJ, Bates GP, et al. Metabolic characterization of the R6/2 transgenic mouse model of Huntington’s disease by high-resolution MAS 1H NMR spectroscopy. Journal of Proteome Research. 2006;5:483-492. DOI: 10.1021/pr050244o
  95. 95. Dimitriu MA, Lazar-Contes I, Roszkowski M, Mansuy IM. Single-cell multiomics techniques: From conception to applications. Front cell. Developmental Biology. 2022;10:1-16
  96. 96. Maiuri T, Truant R. Single cell technologies define New therapeutic avenues for Huntington’s disease. Neuron. 2020;107:768-769
  97. 97. Malaiya S, Cortes-Gutierrez M, Herb BR, Coffey SR, Legg SRW, Cantle JP, et al. Single-nucleus RNA-Seq reveals dysregulation of striatal cell identity due to Huntington’s disease mutations. Journal of Neuroscience. 2021;41:5334-5352. DOI: 10.1523/JNEUROSCI.2074-20.2021
  98. 98. Al-Dalahmah O, Sosunov AA, Shaik A, Ofori K, Liu Y, Vonsattel JP, et al. Single-nucleus RNA-Seq identifies Huntington disease astrocyte states. Acta Neuropathologica Communications. 2020;8:1-21. DOI: 10.1186/s40478-020-0880-6
  99. 99. Lim RG, Al-Dalahmah O, Wu J, Gold MP, Reidling JC, Tang G, et al. Huntington disease oligodendrocyte maturation deficits revealed by single-nucleus RNAseq are rescued by thiamine-biotin supplementation. Nature Communications. 2022;13:1-23. DOI: 10.1038/s41467-022-35388-x
  100. 100. Wang D, Bodovitz S. Single cell analysis: The new frontier in “Omics.”. Trends in Biotechnology. 2010;28:281-290
  101. 101. Clough E, Barrett T. The gene expression omnibus database. In: Methods in Molecular Biology. Vol. 1418. Humana Press Inc.; 2016. pp. 1-18
  102. 102. Papatheodorou I, Moreno P, Manning J, Fuentes AMP, George N, Fexova S, et al. Expression atlas update: From tissues to single cells. Nucleic Acids Research. 2020;48:D77-D83. DOI: 10.1093/nar/gkz947
  103. 103. Slavov N. Driving single cell proteomics forward with innovation. Journal of Proteome Research. 2021;2021:1-9
  104. 104. Deutsch EW, Csordas A, Sun Z, Jarnuczak A, Perez-Riverol Y, Ternent T, et al. The ProteomeXchange consortium in 2017: Supporting the cultural change in proteomics public data deposition. Nucleic Acids Research. 2017;45:D1100-D1106. DOI: 10.1093/nar/gkw936
  105. 105. Deutsch EW, Bandeira N, Sharma V, Perez-Riverol Y, Carver JJ, Kundu DJ, et al. The ProteomeXchange consortium in 2020: Enabling “big data” approaches in proteomics. Nucleic Acids Research. 2020;48:D1145-D1152. DOI: 10.1093/nar/gkz984
  106. 106. Sud M, Fahy E, Cotter D, Azam K, Vadivelu I, Burant C, et al. Metabolomics workbench: An international repository for metabolomics data and metadata, metabolite standards, protocols, tutorials and training, and analysis tools. Nucleic Acids Research. 2016;44:D463-D470. DOI: 10.1093/nar/gkv1042
  107. 107. Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, et al. Integrated analysis of multimodal single-cell data. Cell. 2021;184:3573-3587.e29. DOI: 10.1016/j.cell.2021.04.048
  108. 108. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888-1902.e21. DOI: 10.1016/j.cell.2019.05.031
  109. 109. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nature Biotechnology. 2015;33:495-502. DOI: 10.1038/nbt.3192
  110. 110. Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, et al. Inference and analysis of cell-cell communication using CellChat. Nature Communications. 2021;12:1-20. DOI: 10.1038/s41467-021-21246-9
  111. 111. Raredon MSB, Yang J, Kothapalli N, Lewis W, Kaminski N, Niklason LE, et al. Comprehensive visualization of cell-cell interactions in single-cell and spatial transcriptomics with NICHES. Bioinformatics. 2023;39:1-3. DOI: 10.1093/bioinformatics/btac775
  112. 112. Shan N, Lu Y, Guo H, Li D, Jiang J, Yan L, et al. CITEdb: A manually curated database of cell-cell interactions in human. Bioinformatics. 2022;38:5144-5148. DOI: 10.1093/bioinformatics/btac654
  113. 113. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: Inferring cell–cell communication from combined expression of multi-subunit ligand–receptor complexes. Nature Protocols. 2020;15:1484-1506. DOI: 10.1038/s41596-020-0292-x
  114. 114. Diniz WJS, Canduri F. Bioinformatics: An overview and its applications. Genetics and Molecular Research. 2017;16:1-21
  115. 115. Petryszak R, Fonseca AN, et al. Brazma Alvix RNA-Seq gene profiling - A systematic empirical comparison. Binformatics. 2017;12:1-10. DOI: 10.1371/journal
  116. 116. Parkinson H, Kapushesky M, Shojatalab M, Abeygunawardena N, Coulson R, Farne A, et al. ArrayExpress – A public database of microarray experiments and gene expression profiles. Nucleic Acids Research. 2007;35:747-750. DOI: 10.1093/nar/gkl995
  117. 117. Cezard T, Cunningham F, Hunt SE, Koylass B, Kumar N, Saunders G, et al. The European variation archive: A FAIR resource of genomic variation for all species. Nucleic Acids Research. 2022;50:D1216-D1220. DOI: 10.1093/nar/gkab960
  118. 118. Hubbard T, Barker D, Birney E, Cameron G, Chen Y, Clark L, et al. The Ensembl Genome Database Project. 2002;30:38-41
  119. 119. Baxi EG, Thompson T, Li J, Kaye JA, Lim RG, Wu J, et al. Answer ALS, a large-scale resource for sporadic and familial ALS combining clinical and multi-omics data from induced pluripotent cell lines. Nature Neuroscience. 2022;25:226-237. DOI: 10.1038/s41593-021-01006-0
  120. 120. Sathe S, Ware J, Levey J, Neacy E, Blumenstein R, Noble S, et al. Enroll-HD: An integrated clinical research platform and worldwide observational study for Huntington’s disease. Frontiers in Neurology. 2021;12:1-10
  121. 121. Marek K, Chowdhury S, Siderowf A, Lasch S, Coffey CS, Caspell-Garcia C, et al. The Parkinson’s progression markers initiative (PPMI) – Establishing a PD biomarker cohort. Annals of Clinical Translational Neurology. 2018;5:1460-1477. DOI: 10.1002/acn3.644
  122. 122. Petersen RC, Aisen PS, Beckett LA, Donohue MC, Gamst AC, Harvey DJ, et al. Alzheimer’s Disease Neuroimaging Initiative (ADNI). Clinical Characterization. 2010;2010:201-209
  123. 123. Philip J, Richard C. The PRIDE Proteomics Identifications Database: Data Submission, Query, and Dataset Comparison. In: Thompson JD, Ueffing M, Schaeffer-Reiss C, editors. Methods in Molecular Biology. Vol. 484. Totowa, NJ: Humana Press; 2008
  124. 124. Moriya Y, Kawano S, Okuda S, Watanabe Y, Matsumoto M, Takami T, et al. The Jpost environment: An integrated proteomics data repository and database. Nucleic Acids Research. 2019;47:D1218-D1224. DOI: 10.1093/nar/gky899
  125. 125. Samaras P, Schmidt T, Frejno M, Gessulat S, Reinecke M, Jarzab A, et al. ProteomicsDB: A multi-omics and multi-organism resource for life science research. Nucleic Acids Research. 2020;48:D1153-D1163. DOI: 10.1093/nar/gkz974
  126. 126. Deutsch EW. The PeptideAtlas project. Methods in Molecular Biology. 2010;604:285-296. DOI: 10.1007/978-1-60761-444-9_19
  127. 127. Haug K, Cochrane K, Nainala VC, Williams M, Chang J, Jayaseelan KV, et al. MetaboLights: A resource evolving in response to the needs of its scientific community. Nucleic Acids Research. 2020;48:D440-D444. DOI: 10.1093/nar/gkz1019
  128. 128. Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vázquez-Fresno R, et al. HMDB 4.0: The human metabolome database for 2018. Nucleic Acids Research. 2018;46:D608-D617. DOI: 10.1093/nar/gkx1089
  129. 129. Barabási AL, Gulbahce N, Loscalzo J. Network medicine: A network-based approach to human disease. Nature Reviews. Genetics. 2011;12:56-68
  130. 130. Chasman D, Siahpirani AF, Roy S. Network-based approaches for analysis of complex biological systems. Current Opinion in Biotechnology. 2016;39:157-166
  131. 131. Chandrasekaran S, Bonchev D. Network analysis of human post-mortem microarrays reveals novel genes, MicroRNAs, and mechanistic scenarios of potential importance in fighting Huntington’s disease. Computational and Structural Biotechnology Journal. 2016;14:117-130. DOI: 10.1016/j.csbj.2016.02.001
  132. 132. Sneha NP, Dharshini SAP, Taguchi YH, Gromiha MM. Integrative Meta-analysis of Huntington’s disease transcriptome landscape. Genes (Basel). 2022;13:1-20. DOI: 10.3390/genes13122385
  133. 133. Pirhaji L, Milani P, Leidl M, Curran T, Avila-Pacheco J, Clish CB, et al. Revealing disease-associated pathways by network integration of untargeted metabolomics. Nature Methods. 2016;13:770-776. DOI: 10.1038/nmeth.3940
  134. 134. Pradhan SS, Thota SM, Rajaratnam S, Bhagavatham SKS, Pulukool SK, Rathnakumar S, et al. Integrated multi-omics analysis of Huntington disease identifies pathways that modulate protein aggregation. DMM Disease Models and Mechanisms. 2022;15:1-20. DOI: 10.1242/dmm.049492
  135. 135. Xiang C, Cong S, Liang B, Cong S. Bioinformatic gene analysis for potential therapeutic targets of Huntington’s disease in pre-symptomatic and symptomatic stage. Journal of Translational Medicine. 2020;18:1-10. DOI: 10.1186/s12967-020-02549-9
  136. 136. Zhao N, Quicksall Z, Asmann YW, Ren Y. Network approaches for omics studies of neurodegenerative diseases. Frontiers in Genetics. 2022;13:1-6
  137. 137. Fu MH, Li CL, Lin HL, Tsai SJ, Lai YY, Chang YF, et al. The potential regulatory mechanisms of MIR-196a in Huntington’s disease through bioinformatic analyses. PLoS One. 2015;10:1-11. DOI: 10.1371/journal.pone.0137637
  138. 138. Shirasaki DI, Greiner ER, Al-Ramahi I, Gray M, Boontheung P, Geschwind DH, et al. Network Organization of the Huntingtin Proteomic Interactome in mammalian brain. Neuron. 2012;75:41-57. DOI: 10.1016/j.neuron.2012.05.024
  139. 139. Christodoulou CC, Papanicolaou EZ. Integrated bioinformatics analysis of shared genes, MiRNA, biological pathways and their potential role as therapeutic targets in Huntington’s disease stages. International Journal of Molecular Sciences. 2023;24:1-29. DOI: 10.3390/ijms24054873
  140. 140. Kakouri AC, Christodoulou CC, Zachariou M, Oulas A, Minadakis G, Demetriou CA, et al. Revealing clusters of connected pathways through multisource data integration in Huntington’s disease and spastic Ataxia. IEEE Journal of Biomedical and Health Informatics. 2019;23:26-37. DOI: 10.1109/JBHI.2018.2865569
  141. 141. Christodoulou CC, Zachariou M, Tomazou M, Karatzas E, Demetriou CA, Zamba-Papanicolaou E, et al. Investigating the transition of pre-symptomatic to symptomatic Huntington’s disease status based on omics data. International Journal of Molecular Sciences. 2020;21:1-26. DOI: 10.3390/ijms21197414
  142. 142. Onisiforou A, Spyrou GM. Systems bioinformatics reveals possible relationship between COVID-19 and the development of neurological diseases and neuropsychiatric disorders. Viruses. 2022;14:1-28. DOI: 10.3390/v14102270

Written By

Christiana C. Christodoulou and Eleni Zamba Papanicolaou

Submitted: 01 June 2023 Reviewed: 05 June 2023 Published: 29 February 2024