Open access peer-reviewed chapter - ONLINE FIRST

An Insight on Protein Kinases and Their Therapeutic Perspective

Written By

Ajit Kumar Dhal and Prajna Ritaparna

Submitted: 07 July 2023 Reviewed: 25 September 2023 Published: 05 July 2024

DOI: 10.5772/intechopen.113285

Metabolism - Annual Volume 2024 IntechOpen
Metabolism - Annual Volume 2024 Authored by Yannis Karamanos

From the Annual Volume

Metabolism - Annual Volume 2024 [Working Title]

Prof. Yannis Karamanos

Chapter metrics overview

2 Chapter Downloads

View Full Metrics

Abstract

This book chapter investigates protein kinases, pivotal enzymes governing cellular signaling, and regulation. Constituting around 2% of human genes, protein kinases play a vital role in phosphorylation, a crucial post-translational modification dictating cellular functions. Emphasizing their dynamic nature as molecular switches, the chapter explores their structural intricacies and regulatory mechanisms. It classifies protein kinases into five families based on evolutionary and structural resemblances, each contributing to diverse signaling pathways governing cell growth, metabolism, and immune responses. Dysregulation of these kinases is implicated in various diseases. The chapter discusses the significance of protein kinases in cancer therapy, highlighting targeted treatments such as small molecule inhibitors and monoclonal antibodies. It further explores their role in neurodegenerative disorders such as Alzheimer’s and Parkinson’s diseases, as well as their involvement in cardiovascular diseases, emphasizing their potential as therapeutic targets. Additionally, it sheds light on the regulatory role of protein kinases in inflammatory conditions, suggesting kinase inhibitors and monoclonal antibodies as promising strategies for managing diseases such as rheumatoid arthritis and inflammatory bowel disease. Overall, the chapter provides a comprehensive overview of protein kinases, underlining their structural diversity, regulatory mechanisms, and pivotal roles in physiological and pathological contexts, thereby highlighting their potential as promising targets for personalized medicine interventions across various diseases.

Keywords

  • protein kinases
  • serine/threonine kinases
  • tyrosine kinases
  • histidine-specific kinases
  • dual-specificity kinases
  • and aspartic acid/glutamic acid-specific kinases

1. Introduction

Protein kinases are a class of enzyme that is essential for cellular signaling and control. They are in charge of the phosphorylation process, which involves the transfer of phosphate groups from adenosine triphosphate (ATP) to certain target proteins, as shown in Figure 1. This post-translational alteration has the potential to significantly alter the structure, function, and location of target proteins, altering a variety of cellular processes. Protein kinases, comprising approximately 2% of all human genes, are diverse and abundant in the human genome, with around 518 kinase genes. They play a crucial role in cellular signaling, regulating a wide range of pathways. With over 280 human protein kinase structures characterized, their significance in modulating up to 30% of human proteins highlights their fundamental importance in cellular function and communication [2]. These are found in all living creatures, from bacteria to humans, and have a variety of roles in physiological processes such as cell development, proliferation, differentiation, metabolism, and apoptosis. Examples can be seen in Figure 2. They play an important role in the regulation of intracellular signaling pathways, conveying signals from external stimuli to the cell’s interior and coordinating proper physiological responses [4, 5]. Every protein kinase shares a common structural motif comprising two distinct lobes, as shown in Figure 3. To show the catalytic cycle of protein phosphorylation by a protein kinase, another simple illustration is given by using Figure 4.

Figure 1.

Protein kinases and phosphatases are pivotal enzymatic regulators within biological systems. They orchestrate the intricate dance of cellular signaling by catalyzing phosphorylation and dephosphorylation reactions. Protein kinases facilitate the amplification and propagation of incoming signals by activating specific substrates through phosphorylation. Conversely, the termination of signal transmission occurs through the dephosphorylation (inactivation) of substrates, a process meticulously orchestrated by protein phosphatases [1].

Figure 2.

Allosteric regulation mechanism of Arabidopsis serine/threonine kinase 1 (SIK1) via phosphorylation [3].

Figure 3.

Protein kinases: A unified mechanism and structural basis [6]. Source: (a) Illustration of the core catalytic cycle governing substrate phosphorylation by kinases. Commencing at the top left, ATP engages the kinase’s active site, followed by substrate binding to the same site. Subsequently, the γ-phosphate of ATP (depicted in red) is transferred to a Ser, Thr, or Tyr residue on the substrate. After phosphorylation, the substrate dissociates from the kinase. The final step portrays the release of ADP from the active site. The sequence of these steps and the rate-limiting stage can vary among different kinases. (b) Every protein kinase shares a common structural motif comprising two distinct lobes: one predominantly composed of β-sheet structures (blue) and the other featuring α-helices (green, orange, and yellow). This dual-lobed architecture forms an ATP-binding cleft, which constitutes the active site. The crystal structure of cyclin-dependent kinase-2 (CDK2) (Protein Data Bank (PDB) ID: 1QMZ) serves as a representative example. ATP is depicted as bound within the cleft (red ball-and-stick model). (c) Despite their shared structural fold, kinases target and phosphorylate diverse protein substrates, largely influenced by variations in the charge and hydrophobicity of surface residues. Electrostatic surface representations are presented for four distinct kinases: three Ser/Thr kinases—CDK2, PKA (cAMP-dependent protein kinase, PDB ID: 1ATP), and PHK (phosphorylase kinase, PDB ID: 2PHK)—as well as the tyrosine kinase domain of the insulin receptor (IRK, PDB ID: 1IRK). Positively charged surfaces are depicted in blue, while negatively charged surfaces are shown in red.

Figure 4.

The catalytic cycle of protein phosphorylation by a protein kinase: depiction of phosphate group transfer with red circles [7].

Advertisement

2. Domain

Protein kinases consist of three types of domains: a kinase domain, one or more regulatory domains (referred to as second domains), and additional domains that do not affect the kinase domain. Typically, protein kinases have a catalytic domain that binds ATP and phosphorylates the target proteins together with regulatory domains that control their activity. A conserved amino acid sequence pattern called the kinase domain is present in the catalytic domain and provides the enzymatic activity required for phosphorylation. The regulatory domains may contain areas involved in allosteric control, protein-protein interactions, or the detection of particular biological signals. Numerous protein kinases, each with a unique substrate specificity and biological function, are encoded in the human genome [8]. The preferred configuration is a polypeptide with a kinase domain and exactly one-second domain. However, other variations are also possible, such as polypeptides with different combinations of kinase domains, second domains, and additional domains [9]. The term “polypeptide” encompasses a chain of amino acids, typically the 20 naturally occurring proteinogenic alpha amino acids, and includes molecules where the kinase domain and second domain are linked by a linker, which may be peptidic or non-peptidic. Non-peptidic linkers can be polyethylene or PEG linkers, with a preferred length of less than 50, 40, 30, 20, or 10 atoms (excluding hydrogens). Alternatively, the polypeptide can use the naturally occurring linker found in the wild-type counterpart or a different peptidic linker, such as oligo glycine stretches. Polypeptides containing at least one protein kinase domain are also referred to as “Kinases,” with the preferred kinases having at least one-second domain as described above (Figures 5 and 6) [12].

Figure 5.

Structure of a prototypical protein kinase domain revealing the ATP binding site and surrounding conserved elements (INSR kinase, PDB ID: 1GAG) [10].

Figure 6.

Regulatory domains of protein kinase C: deciphering diverse membrane Signaling events [11].

Advertisement

3. Structure and regulation of protein kinases

Over the past two decades, our understanding of protein kinases has significantly evolved. We now recognize that protein kinases are dynamic molecules, specifically designed as highly regulated molecular switches rather than just efficient catalysts [13]. Initially, we recognized the regulatory potential of phosphorylation, but we underestimated the complexity of the protein kinase family and the vast biological networks they regulate. Sequence analysis revealed scattered conserved motifs throughout the protein kinase core, highlighting their dynamic and regulated nature. The first solved protein kinase structure, PKA, confirmed the conserved fold and revealed the active site characteristics, including the unique positioning of the ATP-binding site and the role of the Activation Loop in controlling the active site’s accessibility [14]. Because all eukaryotic protein kinases share a conserved catalytic core, the crystal structure of cAMP-dependent protein kinase (PKA) allowed us for the first time to begin understanding the molecular basis for the functioning of one protein kinase. This discovery shed light on the entire family of eukaryotic protein kinases [15]. Eukaryote protein kinases (EPKs) are dynamic and flexible molecules that have evolved as molecular switches, transitioning between “on” and “off” states. The integration of sequence, structure, and dynamics is crucial to understand their function and regulation. The landmark crystal structure of the first protein kinase, PKA, provided insights into the conserved structural core of kinases, featuring N- and C-lobes connected by a hinge. The ATP-binding site is located at the interface between these lobes, with surfaces formed by β-sheets. The lobes are connected by the hinge, and the interface encompasses key structural elements like the C-helix, activation loop (A-loop), and substrate recognition sites. Understanding the different structural and functional states of protein kinases aids in the development of kinase inhibitors [16]. Protein kinases function as molecular switches that can be toggled between an “off” state and an “on” state in response to signals. One regulatory mechanism involves pseudo substrate inhibition, where a sequence from the kinase itself or another protein occupies the protein substrate-binding site, preventing substrate binding and inhibiting kinase activity. Inactive kinases may also have key active site residues displaced from their proper positions. In the active state, kinases exhibit a specific conformation characterized by the alignment of catalytic and regulatory residues, forming a regulatory R-spine and a catalytic C-spine. The active conformation also stabilizes the A-loop, which acts as a platform for protein substrate binding. In contrast, inactive kinase structures display a range of conformations, often involving disruption of interactions between the C-helix and the A-loop. The C-helix, a critical structural element, contributes to the allosteric behavior of kinases and is involved in interactions that define kinase activity. Kinases can transition between an active conformation (C-helix in) and an inactive conformation (C-helix out). Autoinhibitory features, such as refolded conformations of the A-loop or specific interactions involving the C-helix, contribute to stabilizing the inactive states. The αC-β4 loop, a highly ordered structure, serves to connect the C-helix and β4 and plays a role in regulating kinase activity and forming dimer interfaces. The confirmation of the A-loop is also influenced by a tripeptide motif known as the DFG motif, which acts as a pivot point. Kinases can adopt different DFG conformations, including DFG-in, DFG-out, and intermediate conformations. The conformational diversity of kinases, including the positions of the C-helix and the DFG motif, affects the geometry of the ATP-binding site and provides opportunities for designing kinase inhibitors that target specific pockets within the kinase structure. Understanding these regulatory mechanisms and conformational changes is crucial for developing effective kinase inhibitors and advancing our knowledge of kinase biology (Figures 57) [18].

Figure 7.

Structure and subdomains of a prototypical protein kinase domain [17]. Source: (A) The architecture of a representative protein kinase domain is depicted, with the protein backbone visualized in a gray cartoon representation, and the peptide substrate displayed in orange. The non-hydrolyzable ATP analog, AMP-PNP, is illustrated in yellow. This structural representation is derived from the PDB structure 1ir3. (B) Schematic delineation of the ATP binding site, partitioned into distinct subdomains.

Advertisement

4. Types of protein kinases

Based on their evolutionary connections and structural resemblances, Protein Kinases are divided into five families. Those are serine/threonine kinases, tyrosine kinases, aspartic acid/glutamic acid-specific, dual-specificity kinases and histidine-specific kinases. These are a few well-known kinase families. Each family controls different signaling pathways and target proteins. Sometimes kinases work in a series of cellular signals forming a phosphorylation cascade [19]. Cancer, neurological illnesses, and autoimmune diseases have all been linked to the dysregulation of protein kinase activity. As a result, many kinase inhibitors have been created and used in targeted therapy. Protein kinases have thus become desirable targets for therapeutic research. Serine/threonine protein kinases (ST-PKs) perform critical roles in controlling physiological processes such as cell cycle progression, signal transduction, and protein synthesis. They phosphorylate serine and threonine residues in target proteins. Tyrosine protein kinases (TKs), which are engaged in important signaling pathways affecting cell growth, differentiation, and metabolism, have as their sole target tyrosine residues. For bacteria to recognize and react to environmental changes, signal transduction, which is made possible by histidine-specific kinases, is crucial. Having the capacity to phosphorylate both serine/threonine and tyrosine residues, dual-specificity protein kinases are involved in a variety of biological functions, such as cell cycle regulation and stress responses. Finally, aspartic acid/glutamic acid-specific protein kinases phosphorylate aspartic acid and glutamic acid residues; however, they are less well-studied than the other groups in terms of their roles and regulation mechanisms. Collectively, these many protein kinases help to regulate physiological activities in a complex way and give phosphorylation events within signaling networks a certain level of specificity. Kinases fall under EC number 2.7, which is assigned by the enzyme commission [20].

4.1 Serine/threonine protein kinases

Serine/threonine protein kinases (STKs) are a diverse group of enzymes that phosphorylate the hydroxyl (OH) group of serine or threonine residues in target proteins. They play a crucial role in regulating various cellular processes and are involved in important post-translational modifications. There are several types of STKs, including CK2 (casein kinase 2), which is known for its involvement in diverse cellular functions. Protein kinase A (PKA) consists of two domains and is responsible for phosphorylating specific substrates. Protein kinase C (PKC) is a family of kinases divided into three subfamilies based on their second messenger requirements. Mos/Raf kinases are part of the MAPKK kinase family and are involved in cell growth stimulation. Mitogen-activated protein kinases (MAPKs) respond to extracellular stimuli and regulate various cellular activities. Ca2+/calmodulin-dependent protein kinases (CAMKs) are primarily regulated by the Ca2+/calmodulin complex. Protein kinase B (AKT) is an important kinase involved in cell proliferation and insulin actions. Cyclin-dependent kinases (CDKs) play a central role in cell cycle regulation, while protein kinase D (PKD) is activated by protein kinase C and regulates various cellular functions such as cell growth, migration, and apoptosis. PKD is also implicated in cardiovascular, neuronal, immune, and tumor-related processes, highlighting its multifaceted roles in different biological contexts. Overall, the diverse types of serine/threonine protein kinases contribute to the intricate regulation of cellular processes and serve as crucial signaling components in various physiological and pathological conditions [2, 20].

4.2 Tyrosine kinases

Tyrosine kinases play a pivotal role in cellular functions by phosphorylating tyrosine residues and acting as molecular switches. They comprise a diverse group of enzymes involved in various signaling cascades, with NRTKs, RTKs, and nuclear tyrosine protein kinases collectively contributing to the intricate landscape of tyrosine kinase-mediated signaling pathways. Tyrosine kinases encompass three major categories: non-receptor tyrosine protein kinases (NRTKs), receptor tyrosine kinases (RTKs), and nuclear tyrosine protein kinases. NRTKs possess crucial domains such as SH1, SH2, SH3, PH, and PTB, which regulate catalytic reactions, enzyme localization, activity, and molecular interactions. With over 30 members across 11 families, NRTKs mediate signal transduction from growth factor receptors, cytokine receptors, lymphocyte antigen receptors, and integrins. Examples include the SRC kinase family, JAK kinase family, and Sky/ZAP-70 family, each involved in distinct signaling pathways and cellular functions. On the other hand, RTKs are transmembrane proteins that transmit signals upon ligand binding, regulating cell growth, differentiation, and development. Nuclear tyrosine protein kinases participate in transcriptional processes and cell cycle regulation [20, 21].

4.3 Histidine protein kinases

Histidine protein kinases phosphorylate histidine residues in substrate proteins. They can be categorized into two main types: two-component histidine protein kinases and two-component mammalian histidine protein kinases. Two-component histidine protein kinases are involved in regulating responses to environmental stimuli. In prokaryotes, histidine kinases are sensor proteins that initiate signaling pathways known as two-component systems (TCSs) in response to various environmental cues. TCSs typically consist of a histidine kinase and a corresponding response regulator that often acts as a transcription factor. The enzymatic activity of a histidine kinase involves the transfer of a phosphate group from ATP to a protein, resulting in the conversion of ATP to ADP and the phosphorylation of histidine. Most histidine kinases are homodimers and possess auto-kinase, phosphotransferase, and phosphatase activities. Histidine kinases are widely present in various organisms such as bacteria, archaea, slime molds, fungi, and plants. They play a crucial role in two-component signal transduction pathways. Examples of these pathways include the bacterial phosphoenolpyruvate: the sugar phosphotransferase system (PTS) and the histidine kinases involved in different signal transduction processes [20, 22].

4.4 Dual-specificity protein kinases

Dual-specific protein kinases exhibit kinase activity toward both serine/threonine and tyrosine residues. They are capable of acting as both serine/threonine kinases and tyrosine kinases. MEKs, which are involved in MAP pathways, serve as prominent examples of dual-specificity kinases. Additionally, there are several other well-known dual-specificity kinases, such as ADK1, CLK1, CLK2, CLK3, CLK4, DSTYK, DYRK1A, DYRK1B, DYRK2, DYRK3, DYRK4, Mps1p, TESK1, TESK2, and TTK. An instance of a dual-specificity protein kinase is MEK5, which phosphorylates the TEY activation motif to activate ERK5 [20, 23, 24].

4.5 Aspartic acid/glutamic acid-specific protein kinases

Protein kinases that are specific to aspartic acid/glutamic acid can phosphorylate aspartate/glutamate. The phosphorus receptor is a part of the protein’s acyl group [20], for example, MEK1 protein kinase. Through the activation of several regulatory kinases, the dual-specificity kinase MEK1 regulates Golgi disassembly. MEK1 is an essential component of the tripeptide signaling module that makes up the MAP kinase signal transduction pathway [24].

Advertisement

5. Protein kinases in therapeutics

Protein kinases, which play pivotal roles in regulating numerous cellular processes, are commonly linked to various diseases, serving as either causal factors or potential targets for therapeutic intervention. Here are some diseases along with the protein kinases associated with them. Some examples are shown in Figure 8.

Figure 8.

Protein kinases: an examination of their overview, classification, and therapeutic prospects [25].

5.1 Targeting protein kinases in cancer therapeutics

Dysregulated protein kinase activity is a hallmark of many cancers. Oncogenic protein kinases drive tumor growth, angiogenesis, and metastasis. Targeting these kinases has shown significant promise in cancer therapeutics. Small molecule inhibitors, such as Imatinib targeting BCR-ABL [26] in chronic myeloid leukemia, have revolutionized cancer treatment. Monoclonal antibodies, such as Trastuzumab targeting HER2 in breast cancer [27], have been successful in blocking kinase activity and inhibiting tumor growth. Gene therapy approaches, such as using viral vectors to deliver RNA interference against oncogenic kinases, hold potential in personalized cancer treatments. An example is explained in Figure 9.

Figure 9.

Serine/threonine phosphorylation in cytokine-mediated signal transduction pathways | Leukemia research [28].

5.2 Protein kinases in neurodegenerative disorders

Protein kinases are implicated in the pathogenesis of neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases. Abnormal phosphorylation of tau protein by kinases like glycogen synthase kinase-3β (GSK-3β) contributes to neurofibrillary tangle formation in Alzheimer’s disease. Inhibitors targeting GSK-3β [29] have shown promise in preclinical studies. Similarly, aberrant protein kinase activity, including that of leucine-rich repeat kinase 2 (LRRK2), is implicated in Parkinson’s disease. Inhibitors targeting LRRK2 have been developed and are being evaluated in clinical trials. Additional neurological disorders are elucidated in Figures 1012.

Figure 10.

Impact of protein kinases (PKA, PKB, and PKC) on postischemic brain injury progression and associated cell Signaling events [30]. This figure delineates the pivotal roles played by three crucial protein kinases—protein kinase A (PKA), protein kinase B (PKB), and protein kinase C (PKC)—in the progression of postischemic brain injury, as well as their associated cell signaling cascades. The key molecular components involved in these pathways are identified: BAD (Bcl-2-associated death promoter), cAMP (cyclic adenosine monophosphate), DAG (diacylglycerol), GSK-3 (glycogen synthase kinase three), IKK alpha (I kappa B kinase subunit alpha), IP3 (inositol triphosphate), mTOR (mechanistic target of rapamycin), PIP2 (phosphatidylinositol biphosphate), PKA (protein kinase A), PKC (protein kinase C), PLC (phospholipase C), RACK (receptor for activated C-kinase), and S6K1 (protein S6 kinase one).

Figure 11.

Regulation of brain-type creatine kinase (CKB) in Huntington’s disease (HD) [31]. This figure caption provides a concise overview of potential mechanisms underlying the regulation of the creatine kinase (CK)/phosphocreatine (PCr) system by mutant huntingtin (Htt) in Huntington’s disease (HD). It highlights the downregulation of both CKB expression and transcript levels in HD. Solid lines indicate pathways supported by experimental evidence, while dotted lines represent hypothesized pathways. It is known that mutant Htt interacts with and activates p53, which, in turn, suppresses the CKB promoter.

Figure 12.

Targets of CK2 in neurodegenerative disorders. This figure highlights the involvement of CK2 in various neurodegenerative diseases, indicating its impact on disease-related processes using color-coded shapes. Red shapes represent CK2 targets that contribute to pathological functions, blue shapes represent proteins through which CK2 may exert protective effects against the disease, and gray shapes denote CK2-dependent phosphorylation events that do not have a direct disease-related effect [32]. Source: (a) Parkinson’s Disease (PD): CK2 promotes α-synuclein aggregation in Lewis bodies (LB) by phosphorylating Ser129, although this site is also targeted by other protein kinases. (b) Alzheimer’s Disease (AD): CK2 plays a role in the 5-HT4 receptor-stimulated induction of α-secretase activity, reducing Aβ production through the non-amyloidogenic pathway. However, CK2 induces tau hyperphosphorylation by phosphorylating SET, an inhibitor of the PP2A phosphatase, leading to its cytosolic localization and binding to PP2A. CK2 also phosphorylates KLC1, causing FAT impairment. CK2 targets PS-2, but this phosphorylation does not affect APP processing. (c) Huntington’s Disease (HD): CK2 increases phosphorylation at Ser13 and Ser16 of HTT, which are hypo-phosphorylated in the polyQ-HTT mutant, thereby reducing cellular toxicity. (d) Spinocerebellar Ataxia Type 3 (SCA3): CK2 associates with and phosphorylates ataxin-3, promoting its nuclear localization and stabilization, and enhancing inclusion formation. (e) Amyotrophic Lateral Sclerosis (ALS): CK2 is a potential kinase of TDP43, a major component of protein aggregates in motor neurons. Phosphorylation by CK2 decreases TDP43’s propensity to aggregate. CK2 also phosphorylates cyclin F, negatively regulating the E3 ligase activity of the SKP1/cullin1/F-box (SCF)-E3 ligase complex and reducing aberrant protein ubiquitination typically observed in ALS.

5.3 Protein kinases and cardiovascular diseases

Protein kinases play a critical role in cardiovascular functions and are involved in the pathogenesis of cardiovascular diseases. For instance, protein kinase C (PKC) isoforms are known to regulate cardiac contractility, hypertrophy, and fibrosis. PKC inhibitors, such as Enzastaurin, have shown the potential to reduce myocardial damage in heart failure. In addition, the mitogen-activated protein kinase (MAPK) pathway is involved in vascular smooth muscle cell proliferation and migration. Inhibitors targeting MAPK kinases have shown promise in preventing restenosis after angioplasty [33].

5.4 Protein kinases in inflammatory conditions

Inflammation is tightly regulated by protein kinases, which modulate the activity of key transcription factors and inflammatory mediators. Dysregulated kinase signaling can lead to chronic inflammation and contribute to the pathogenesis of inflammatory diseases. Small molecule inhibitors targeting kinases, such as Janus kinases (JAKs) and spleen tyrosine kinase (SYK), have shown efficacy in inflammatory conditions like rheumatoid arthritis and inflammatory bowel disease. Monoclonal antibodies targeting cytokine receptors and downstream kinases have also emerged as effective therapeutics in various inflammatory diseases [34]. A comprehensive list of protein kinases with their descriptions and associated diseases is depicted in Table 1.

Protein kinase groupProtein kinase familyCellular functionSome examplesAssociated diseasesReferences
Serine threonine protein kinasesCyclin-dependent kinase (CDK)Regulation of cell cycleCDK1Polyploidy and retinoblastoma[35]
CDK2Colon adenocarcinoma, lung adenocarcinoma, breast invasive ductal carcinoma, conventional glioblastoma multiforme, and endometrial endometrioid adenocarcinoma[36]
CDK4Breast cancer[37]
CDK5Lissencephaly 7 with cerebellar hypoplasia, medullary thyroid cancer (MTC), hepatocellular carcinoma, Alzheimer’s Disease, Parkinson’s disease, Huntington’s disease[38, 39, 40]
CDK6Primary microcephaly 12, prostate cancer, breast cancer[38]
CDK8Pancreatic cancer, uterine leiomyosarcoma, sporadic pituitary adenomas, intellectual developmental disorder with hypotonia and behavioral abnormalities[38, 41, 42, 43]
CDK10Gastrointestinal cancer, Al Kaissi syndrome[38, 44]
CDK13Congenital heart defects, dysmorphic facial features, intellectual developmental disorder[38]
CDK19Hepatocellular carcinoma, prostate cancer, bilateral congenital retinal folds, microcephaly, and mild mental retardation[45, 46, 47]
Mitogen-Activated Protein Kinase (MAPK)Embryogenesis, cell death, cell proliferation, and cell differentiationERK1/2Cancer, asthma, stroke, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, leishmaniasis, Chagas disease[48, 49]
JNK1/2/3Obesity and diabetes, immune disorders, skin cancer, colorectal cancer, prostate cancer, and various respiratory diseases[50, 51]
p38 proteinBreast cancer, prostate cancer, bladder cancer, liver cancer, lung cancer, colon cancer, thyroid cancer, leukemia, rheumatoid arthritis, Type 2 Diabetes, inflammatory diseases, chronic obstructive pulmonary disease[52, 53]
ERK5Ischaemia, chronic pain, prostate cancer, bladder cancer, breast cancer, esophageal squamous cell carcinoma, nasopharyngeal carcinoma, acute myeloid leukemia, T-cell leukemia, B-cell malignancies, hypertension[54, 55]
Protein Kinase CCell death and proliferation, gene transcription and translation, alteration of cell shape and migration, regulation of ion channels and receptorsPKCα/βDiabetes, atherosclerosis, endothelial dysfunction,[56]
Protein kinase DCell Golgi reverse membrane transport, cell growth, proliferation, migration, differentiation and apoptosisPKD1/2/3Cancer, metabolic disorders, inflammatory diseases, neuronal dysfunctions, immune dysregulation, cardiac hypertrophy,[57, 58]
DNA-dependent protein kinaseGoverns DNA damage and repairSevere combined immunodeficiency, ataxia-telangiectasia, chronic kidney disease, thyroid cancer[59, 60, 61]
Aurora protein kinasesCell proliferationAURKA/B/CBreast cancer, ovarian cancer, gastric/gastrointestinal cancer, colorectal cancer, esophageal squamous cell carcinoma, lung cancer, cervical cancer, prostate cancer, glioma, acute myeloid leukemia, oral cancer[62]
Pancreatic KininogenaseInvolved in neurological functions, inhibition of platelet aggressionType 2 diabetes, renal fibrosis,[20, 63]
Tyrosine kinasesNon-receptor tyrosine protein kinasesCatalytic reaction of the kinase, enzyme localization, signal transduction of various growth factor receptors, cytokine receptors, lymphocyte antigen receptors, and adhesion molecule integrins.SRCChronic kidney disease, solid tumor, hematologic malignancies[64, 65]
Sky/ZAP-70Severe combined immunodeficiency disorder, lymphocytic leukemia, inflammatory bowel diseases[66]
JAK1/2/3Immunodeficiencies, myeloproliferative disorders, cancers, gynecologic tumors, hepatocellular carcinoma, invasive ductal carcinoma, putative primary erythrocytosis, thrombocythemia[67, 68]
ABLCancer, neurodegenerative diseases[69, 70]
Receptor Tyrosine Protein KinasesCell surface receptors for many polypeptide growth factors, cytokines, and hormonesRTKHematological disorder, inherited diseases (Dwarfism, craniosynostosis, heritable cancer susceptibility, venous malformation, and Piebaldism),[71]
EGFRLung cancer, cholangiocarcinoma[72]
PDGFRPDGFRA-associated chronic eosinophilic leukemia, gastrointestinal stromal tumor,[73]
FGFRCongenital craniosynostosis, Dwarfism syndromes, chronic kidney disease (CKD), obesity, insulin resistance, Tumors[74, 75]
NGFRAlzheimer’s disease, depression, schizophrenia, and antidepressant efficacy[76, 77]
Histidine-specific protein kinasesHistidine phosphorylationBCKDKNeurobehavioral deficit, autism, epilepsy[78, 79]
Dual-specificity protein kinasesDYRKDown syndrome, Alzheimer’s disease, Parkinson’s disease, mental retardation disease 7, viral infections, pancreatic cancer, brain tumor, acute megakaryoblastic leukemia, lymphoblastic leukemia[80]
CLKGlioblastoma, HIV-1, knee osteoarthritis, hepatocellular carcinoma, prostate cancer, cholangiocarcinoma[80]

Table 1.

A comprehensive list of protein kinases-associated diseases and their descriptions.

Advertisement

6. Conclusion

In conclusion, protein kinases are pivotal players in cellular signaling and control, regulating a wide array of physiological processes through post-translational phosphorylation. The diversity and abundance of protein kinases in the human genome underscore their fundamental importance in cellular function and communication. Protein kinases are dynamic molecules, serving as highly regulated molecular switches rather than mere catalysts. Their structural and functional diversity allows for precise control of cellular responses, making them attractive targets for therapeutic research.

The classification of protein kinases into distinct families, including serine/threonine kinases, tyrosine kinases, histidine-specific kinases, dual-specificity kinases, and aspartic acid/glutamic acid-specific kinases, highlights the intricate nature of phosphorylation events within signaling networks. These kinases play critical roles in a wide range of physiological and pathological conditions, including cancer, neurodegenerative disorders, cardiovascular diseases, and inflammatory conditions.

The development of kinase inhibitors, monoclonal antibodies, and gene therapy approaches has shown significant promise in cancer therapeutics, offering personalized treatment options. In neurodegenerative disorders, targeting specific kinases involved in abnormal phosphorylation pathways holds potential for disease-modifying interventions. Protein kinases also play a crucial role in cardiovascular functions, and kinase inhibitors have the potential to mitigate myocardial damage and prevent restenosis after angioplasty. Furthermore, in inflammatory conditions, kinase inhibitors and monoclonal antibodies have emerged as effective therapeutics, providing new avenues for managing chronic inflammation.

Overall, the complex and versatile nature of protein kinases positions them as central regulators of cellular processes and attractive targets for therapeutic interventions across a spectrum of diseases. Continued research into the structure, function, and regulation of protein kinases is essential for the development of innovative treatments and a deeper understanding of cellular biology.

Advertisement

Conflicts of interest

The authors declare that they have no conflict of interest.

References

  1. 1. Hillertz P. Advances in Fragment-Based Drug Discovery: Studies of cAMP-dependent protein-kinase A using X-ray-crystallography, surface-plasmon-resonance and high compound concentration assays (Doctoral dissertation). DOI: 10.11588/heidok.00010665
  2. 2. Modi V, Dunbrack Jr RL. A structurally-validated multiple sequence alignment of 497 human protein kinase domains. Scientific Reports. 24 Dec 2019;9(1):19790. DOI: 10.1038/s41598-019-56499-4
  3. 3. Mu J, Zhou J, Gong Q, Xu Q. An allosteric regulation mechanism of Arabidopsis serine/threonine kinase 1 (SIK1) through phosphorylation. Computational and Structural Biotechnology Journal. 2022;20:368-379. DOI: 10.1016/j.csbj.2021.12.033
  4. 4. Martin J, Anamika K, Srinivasan N. Classification of protein kinases on the basis of both kinase and non-kinase regions. PLoS One. 2010;5(9):1-12. DOI: 10.1371/journal.pone.0012460
  5. 5. Ardito F, Giuliani M, Perrone D, Troiano G, Lo Muzio L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (review). International Journal of Molecular Medicine. 2017;40(2):271-280. DOI: 10.3892/ijmm.2017.3036
  6. 6. Ubersax JA, Ferrell JE. Mechanisms of specificity in protein phosphorylation. Nature Reviews Molecular Cell Biology. 2007;8(7):530-541. DOI: 10.1038/nrm2203
  7. 7. Li T et al. A systematic review of recently reported marine-derived natural product kinase inhibitors. Marine Drugs. 23 Aug 2019;17(9):493. DOI: 10.3390/md17090493
  8. 8. Cheng HC, Qi RZ, Paudel H, Zhu HJ. Regulation and function of protein kinases and phosphatases. Enzyme Research. 2011;1:2011. DOI: 10.4061/2011/794089
  9. 9. Kobe B, Kemp BE. Principles of kinase regulation. Handbook of Cell Signaling. 2003;1–3:539-542. DOI: 10.1016/B978-012124546-7/50450-2
  10. 10. Modi V, Dunbrack Jr RL. Defining a new nomenclature for the structures of active and inactive kinases. Proceedings of the National Academy of Sciences. 2 Apr 2019;116(14):6818-6827. DOI: 10.81/zenodo.2580462
  11. 11. Corbalán-García S, Gómez-Fernández JC. Protein kinase C regulatory domains: The art of decoding many different signals in membranes. Biochimica et Biophysica Acta - Molecular and Cell Biology of Lipids. 2006;1761(7):633-654. DOI: 10.1016/j.bbalip.2006.04.015
  12. 12. Reddy Chichili VP, Kumar V, Sivaraman J. Linkers in the structural biology of protein-protein interactions. Protein Science. 2013;22(2):153-167. DOI: 10.1002/pro.2206
  13. 13. Cormier KW, Woodgett JR. Protein kinases: Physiological roles in cell signalling. In: eLS. Wiley; 2016. pp. 1-9. DOI: 10.1002/9780470015902.a0002710.pub3
  14. 14. Taylor SS, Zhang P, Steichen JM, Keshwani MM, Kornev AP. PKA: Lessons learned after twenty years. Biochimica et Biophysica Acta - Proteins and Proteomics. 2013;1834(7):1271-1278. DOI: 10.1016/j.bbapap.2013.03.007
  15. 15. Serota TS, Elzbieta RA. Three Protein Kinase Structures Define a Common Motif. Structure-Cell Press. 1994;2(5):P345-355. DOI: 10.1016/ s0969-2126(00)00036-8
  16. 16. McClendon CL, Kornev AP, Gilson MK, Taylora SS. Dynamic architecture of a protein kinase. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(43):E4623-E4631. DOI: 10.1073/pnas.1418402111
  17. 17. Macías ÁL. Design, Synthesis and Biological Evaluation of 6-Alkoxypurine Derivatives as Kinase Inhibitors a Dissertation Presented [Online]. 2019. Available from: http://hdl.handle.net/10481/57973
  18. 18. Arter C, Trask L, Ward S, Yeoh S, Bayliss R. Structural features of the protein kinase domain and targeted binding by small-molecule inhibitors. Journal of Biological Chemistry. 2022;298(8). DOI: 10.1016/j.jbc.2022.102247
  19. 19. Park G, Pan S, Borkovich KA. Mitogen-activated protein kinase cascade required for regulation of development and secondary metabolism in Neurospora crassa. Eukaryotic Cell. 2008;7(12):2113-2122. DOI: 10.1128/EC.00466-07
  20. 20. CUSABIO Team. Protein Kinases Overview: Definition, Types, Function, Activation Mechanism, Etc. [Online]. Available from: https://www.cusabio.com/c-16643.html [Accessed: July 29, 2023]
  21. 21. Hubbard SR, Till JH. Protein Tyrosine Kinase Structure and Function. 2000. Available from: www.annualreviews.org
  22. 22. Lembke HK, Carlson EE. Activity-based ATP analog probes for bacterial histidine kinases. Methods in Enzymology. 2022;664:59-84. DOI: 10.1016/BS.MIE.2022.01.003
  23. 23. Protein Kinases: Overview, Classification and Therapeutic Potential [Online]. Available from: https://www.assaygenie.com/blog/protein-kinases [Accessed: July 30, 2023]
  24. 24. Colanzi A, Sütterlin C. Signaling at the golgi during mitosis. Methods in Cell Biology. 2013;118:383-400. DOI: 10.1016/B978-0-12-417164-0.00023-9
  25. 25. Assaygenie Team. Protein Kinases: Overview, Classification and Therapeutic Potential [Online]. Available from: https://www.assaygenie.com/blog/protein-kinases [Accessed: July 26, 2023]
  26. 26. American Cancer Society. Targeted Therapies for Chronic Myeloid Leukemia [Online]. Available from: https://www.cancer.org/cancer/types/chronic-myeloid-leukemia/treating/targeted-therapies.html [Accessed: July 29, 2023]
  27. 27. HER2-Positive Breast Cancer [Online]. Available from: https://www.webmd.com/breast-cancer/her2 [Accessed: July 29, 2023]
  28. 28. Mccubrey JA, Stratford May W, Duronio V, Mufson A. Mini-review and Meeting Report Serine/Threonine Phosphorylation in Cytokine Signal Transduction [Online]. 2000. Available from: www.nature.com/leu
  29. 29. Shri SR, Manandhar S, Nayak Y, Pai KSR. Role of GSK-3β Inhibitors: New Promises and Opportunities for Alzheimer’s Disease. Advanced Pharmaceutical Bulletin. 2023;13(4):688. DOI: 10.34172/apb.2023.071
  30. 30. Raval AP, Perez-Pinzon MA, Dave KR. Protein Kinases in Cerebral Ischemia. In: Primer on Cerebrovascular Diseases. Academic Press; 1 Jan 2017. pp. 246-250. DOI: 10.1016/B978-0-12-803058-5.00050-3
  31. 31. Ju TC, Lin YS, Chern Y. Energy dysfunction in Huntington’s disease: Insights from PGC-1α, AMPK, and CKB. Cellular and Molecular Life Sciences. 2012;69(24):4107-4120. DOI: 10.1007/s00018-012-1025-2
  32. 32. Borgo C, D’Amore C, Sarno S, Salvi M, Ruzzene M. Protein kinase CK2: A potential therapeutic target for diverse human diseases. Signal Transduction and Targeted Therapy. 2021;6(-1):183. DOI: 10.1038/s41392-021-00567-7
  33. 33. Del Buono MG, Moroni F, Montone RA, Azzalini L, Sanna T, Abbate A. Ischemic cardiomyopathy and heart failure after acute myocardial infarction. Current Cardiology Reports. 2022;24(10):1505-1515. DOI: 10.1007/s11886-022-01766-6
  34. 34. JAK Inhibitors for Rheumatoid Arthritis [Online]. Available from: https://www.webmd.com/rheumatoid-arthritis/jak-inhibitors-rheumatoid-arthritis [Accessed: July 30, 2023]
  35. 35. Gene Cards Team. CDK1 gene [Online]. Available from: https://www.genecards.org/cgi-bin/carddisp.pl?gene=CDK1#:∼:text=CDK1%20(Cyclin%20Dependent%20Kinase%201,of%20Nlp%20from%20mitotic%20centrosomes [Accessed: October 25, 2023]
  36. 36. My Cancer Genom Team. CDK2 [Online]. Available from: https://www.mycancergenome.org/content/gene/cdk2/#:%20:text=CDK2%20is%20altered%20in%200.16,prevalence%20%20f%20alterations%20%5B3%5D[Accessed: October 25, 2023]
  37. 37. Baker SJ, Reddy EP. CDK4: A key player in the cell cycle, development, and cancer. Genes and Cancer. 2012;3(11–12):658-669. DOI: 10.1177/1947601913478972
  38. 38. Colas P. Cyclin-dependent kinases and rare developmental disorders. Orphanet Journal of Rare Diseases. 2020;15(1):1-14. DOI: 10.1186/s13023-020-01472-y
  39. 39. Sabbir MG. Loss of Ca2+/calmodulin dependent protein kinase kinase 2 leads to aberrant transferrin phosphorylation and trafficking: A potential biomarker for Alzheimer’s disease. Frontiers in Molecular Biosciences. 2018;5(November):99. DOI: 10.3389/fmolb.2018.00099
  40. 40. Łukasik P, Załuski M, Gutowska I. Cyclin-dependent kinases (Cdk) and their role in diseases development–review. International Journal of Molecular Sciences. 2021;22(6):1-33. DOI: 10.3390/ijms22062935
  41. 41. Yasutake N et al. Cyclin-dependent kinase 8 is an independent prognosticator in uterine leiomyosarcoma. Pathology, Research and Practice. 2022;235:153920. DOI: 10.1016/J.PRP.2022.153920
  42. 42. Wei R et al. CDK8 regulates the angiogenesis of pancreatic cancer cells in part via the CDK8-β-catenin-KLF2 signal axis. Experimental Cell Research. 2018;369(2):304-315. DOI: 10.1016/J.YEXCR.2018.05.033
  43. 43. Gaspar LM et al. A common variant in the CDK8 gene is associated with sporadic pituitary adenomas in the Portuguese population: A case-control study. International Journal of Molecular Sciences. 2022;23(19):11749. DOI: 10.3390/ijms231911749
  44. 44. Bazzi ZA, Tai IT. CDK10 in gastrointestinal cancers: Dual roles as a tumor suppressor and oncogene. Frontiers in Oncology. 2021;11:655479. DOI: 10.3389/fonc.2021.655479
  45. 45. Chen X et al. CDK6 is upregulated and may be a potential therapeutic target in enzalutamide-resistant castration-resistant prostate cancer. European Journal of Medical Research. 2022;27(1):1-3. DOI: 10.1186/s40001-022-00730-y
  46. 46. Cai X, Deng J, Zhou J, Cai H, Chen Z. Cyclin-dependent kinase 19 upregulation correlates with an unfavorable prognosis in hepatocellular carcinoma. BMC Gastroenterology. 2021;21(1). DOI: 10.1186/s12876-021-01962-8
  47. 47. Becker F et al. Increased mediator complex subunit CDK19 expression associates with aggressive prostate cancer. International Journal of Cancer. 2020;146(2):577-588. DOI: 10.1002/ijc.32551
  48. 48. Soares-Silva M, Diniz FF, Gomes GN, Bahia D. The mitogen-activated protein kinase (MAPK) pathway: Role in immune evasion by trypanosomatids. Frontiers in Microbiology. 2016;7(FEB):183. DOI: 10.3389/fmicb.2016.00183
  49. 49. Kim EK, Choi EJ. Compromised MAPK signaling in human diseases: An update. Archives of Toxicology. 2015;89(6):867-882. DOI: 10.1007/s00204-015-1472-2
  50. 50. Hammouda MB, Ford AE, Liu Y, Zhang JY. The JNK Signaling pathway in inflammatory skin disorders and cancer. Cell. 2020;9-4:857. DOI: 10.3390/cells9040857
  51. 51. Chen J et al. The roles of c-Jun n-terminal kinase (Jnk) in infectious diseases. International Journal of Molecular Sciences. 2021;22(17) MDPI. DOI: 10.3390/ijms22179640
  52. 52. Gupta J, Nebreda AR. Roles of p38α mitogen-activated protein kinase in mouse models of inflammatory diseases and cancer. FEBS Journal. 2015;282(10):1841-1857. DOI: 10.1111/febs.13250
  53. 53. Burton JC, Antoniades W, Okalova J, Roos MM, Grimsey NJ. Atypical p38 signaling, activation, and implications for disease. International Journal of Molecular Sciences. 2021;22(8):4183. DOI: 10.3390/ijms22084183
  54. 54. Roberts R. The extracellular signal-regulated kinase (ERK) pathway: A potential therapeutic target in hypertension. Journal of Experimental Pharmacology. 2012;(8):77-83. DOI: 10.2147/jep.s28907
  55. 55. Nithianandarajah-Jones GN, Wilm B, Goldring CEP, Müller J, Cross MJ. ERK5: Structure, regulation and function. Cellular Signalling. 2012;24(11):2187-2196. DOI: 10.1016/J.CELLSIG.2012.07.007
  56. 56. Lien CF, Chen SJ, Tsai MC, Lin CS. Potential role of protein kinase C in the pathophysiology of diabetes-associated atherosclerosis. Frontiers in Pharmacology. 2021;12(7):716332. DOI: 10.3389/fphar.2021.716332
  57. 57. Simsek Papur O, Sun A, Glatz JFC, Luiken JJFP, Nabben M. Acute and chronic effects of protein kinase-D Signaling on cardiac energy metabolism. Frontiers in Cardiovascular Medicine. 2018;5(6):65. DOI: 10.3389/fcvm.2018.00065
  58. 58. Protein Kinase D [Online]. Available from: https://encyclopedia.pub/entry/8697 [Accessed: October 25, 2023]
  59. 59. Yang Y et al. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) drives chronic kidney disease progression in male mice. Nature Communications. 2023;14(1):1334. DOI: 10.1038/s41467-023-37043-5
  60. 60. Goodwin JF, Knudsen KE. Beyond DNA repair: DNA-PK function in cancer. Cancer Discovery. 2014;4(10):1126-1139. DOI: 10.1158/2159-8290.CD-14-0358
  61. 61. Ihara M, Ashizawa K, Shichijo K, Kudo T. Expression of the DNA-dependent protein kinase catalytic subunit is associated with the radiosensitivity of human thyroid cancer cell lines. Journal of Radiation Research. 2019;60(2):171-177. DOI: 10.1093/jrr/rry097
  62. 62. Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora Kinases: Novel Therapy Targets in Cancers [Online]. 2017. Available from: www.impactjournals.com/oncotarget/
  63. 63. Zhu D, Zhang L, Cheng L, Ren L, Tang J, Sun D. Pancreatic Kininogenase ameliorates renal fibrosis in Streptozotocin induced-diabetic nephropathy rat. Kidney & Blood Pressure Research. 2016;41(1):9-17. DOI: 10.1159/000368542
  64. 64. Wang J, Zhuang S. Src family kinases in chronic kidney disease. American Journal of Physiology. Renal Physiology. 2017;313:721-728. DOI: 10.1152/ajprenal.00141.2017.-Src
  65. 65. Alvarez RH, Kantarjian HM, Cortes JE. The role of Src in solid and hematologic malignancies: Development of new-generation src inhibitors. Cancer. 2006;107(8):1918-1929. DOI: 10.1002/cncr.22215
  66. 66. NCBI. ZAP70 Zeta Chain of T Cell Receptor-Associated Protein Kinase 70 [Online]. Available from: https://www.ncbi.nlm.nih.gov/gene/7535 [Accessed: October 24, 2023]
  67. 67. Laurence A, Pesu M, Silvennoinen O, O’shea J. JAK Kinases in Health and Disease: An Update. 2012
  68. 68. Hammarén HM, Virtanen AT, Raivola J, Silvennoinen O. The regulation of JAKs in cytokine signaling and its breakdown in disease. Cytokine. 2019;118:48-63. DOI: 10.1016/J.CYTO.2018.03.041
  69. 69. Team Cell Signaling Technology. Kinase-Disease Associations [Online]. Available from: https://www.cellsignal.com/learn-and-support/reference-tables/kinasedisease-associations [Accessed: October 24, 2023]
  70. 70. Khatri A, Wang J, Pendergast AM. Multifunctional Abl kinases in health and disease. Journal of Cell Science. 2016;129(1):9-16. DOI: 10.1242/jcs.175521
  71. 71. Trends in Genetics team, “RTK Mutations and Human Syndromes.”
  72. 72. Medline Plus. EGFR gene [Online]. Available from: https://medlineplus.gov/genetics/gene/egfr/#synonyms [Accessed: October 25, 2023]
  73. 73. Medline Plus. PDGFRA Gene [Online]. Available from: https://medlineplus.gov/genetics/gene/pdgfra/#conditions [Accessed: October 25, 2023]
  74. 74. NCBI. FGFR Craniosynostosis Syndromes Overview [Online]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1455/ [Accessed: October 25, 2023]
  75. 75. Xie Y et al. FGF/FGFR signaling in health and disease. Signal Transduction and Targeted Therapy. 2020;5(1):181. DOI: 10.1038/s41392-020-00222-7
  76. 76. Zhao L et al. NGFR gene and Single nucleotide polymorphisms, rs2072446 and rs11466162, playing roles in psychiatric disorders. Brain Sciences. 2022;12(10):1372. DOI: 10.3390/brainsci12101372
  77. 77. Bruno F, Abondio P, Montesanto A, Luiselli D, Bruni AC, Maletta R. The nerve growth factor receptor (NGFR/p75NTR): A major player in Alzheimer’s disease. International Journal of Molecular Sciences. 2023;24(4):3200. DOI: 10.3390/ijms24043200
  78. 78. Novarino G et al. Mutations in BCKD-kinase lead to a potentially treatable form of autism with epilepsy. Science (1979). 2012;338(6105):394-397. DOI: 10.1126/science.1224631
  79. 79. García-Cazorla A et al. Two novel mutations in the BCKDK (branched-chain Keto-acid dehydrogenase kinase) gene are responsible for a neurobehavioral deficit in two pediatric unrelated patients. Human Mutation. 2014;35(4):470-477. DOI: 10.1002/humu.22513
  80. 80. Lindberg MF, Meijer L. Dual-specificity, tyrosine phosphorylation-regulated kinases (Dyrks) and cdc2-like kinases (clks) in human disease, an overview. International Journal of Molecular Sciences. 2021;22(11). DOI: 10.3390/ijms22116047

Written By

Ajit Kumar Dhal and Prajna Ritaparna

Submitted: 07 July 2023 Reviewed: 25 September 2023 Published: 05 July 2024