Fact-checked by Grok 2 weeks ago

c-Jun N-terminal kinases

c-Jun N-terminal kinases (JNKs), also known as stress-activated protein kinases (SAPKs), are a subfamily of serine/threonine protein kinases within the mitogen-activated protein kinase (MAPK) superfamily, essential for transducing extracellular signals into cellular responses such as stress adaptation, inflammation, and programmed cell death. Encoded by three distinct genes—MAPK8 (JNK1), MAPK9 (JNK2), and MAPK10 (JNK3)—JNKs produce up to ten isoforms through alternative splicing, ranging from 46 to 55 kDa in size, with JNK1 and JNK2 exhibiting ubiquitous expression while JNK3 is predominantly found in the brain, heart, and testes. These kinases were first identified in the early 1990s for their ability to phosphorylate the transcription factor c-Jun at N-terminal serine residues (Ser63 and Ser73), thereby enhancing its transcriptional activity as part of the AP-1 complex. JNKs are activated through a three-tiered MAPK signaling , where upstream MAP kinase (MKK4 and MKK7) a conserved threonine-proline-tyrosine (TPY) motif in the activation loop (Thr183/Tyr185 in JNK1), in response to diverse stimuli including environmental stresses (e.g., UV radiation, ), cytokines (e.g., TNF-α), and growth factors. This dual phosphorylation enables JNKs to translocate to various subcellular compartments, such as the or mitochondria, where they over 50 known substrates, including transcription factors (c-Jun, ATF2, Elk-1), receptors, and cytoskeletal proteins. The isoform-specific functions are notable: JNK1 and JNK2 often promote pro-inflammatory and proliferative responses, whereas JNK3 is more associated with neuronal and stress-induced neurodegeneration. In biological contexts, JNK signaling is pivotal for regulating cell proliferation, differentiation, migration, and survival, with dysregulation implicated in numerous pathologies including cancer, neurodegenerative diseases (e.g., Alzheimer's via tau hyperphosphorylation), metabolic disorders (e.g., insulin resistance in diabetes), and inflammatory conditions. For instance, persistent JNK activation contributes to amyloid-β-mediated neurotoxicity and tau pathology in Alzheimer's disease models, highlighting its therapeutic potential as a target for inhibitors like SP600125. As of 2025, JNK inhibitors such as CC-90001 are in clinical trials for conditions like idiopathic pulmonary fibrosis, with promising results in reducing fibrosis progression. Overall, the versatility of JNK pathways underscores their role as integrators of stress signals, balancing protective and detrimental cellular outcomes depending on context and duration of activation.

Structure and Isoforms

Gene and Protein Isoforms

The c-Jun N-terminal kinases (JNKs) are encoded by three distinct genes in the human genome: MAPK8 (also known as JNK1), located on chromosome 10q11.22; MAPK9 (JNK2), on chromosome 5q35.3; and MAPK10 (JNK3), on chromosome 4q21.3. Each gene undergoes alternative splicing to generate multiple protein isoforms, resulting in a total of ten primary isoforms across the family. For JNK1 and JNK2, four isoforms are produced per gene: the α and β variants differ in their C-terminal sequences due to alternative exon usage, while the numerical suffixes (1 or 2) indicate the presence or absence of a 9-amino-acid insertion in the kinase subdomain X. Representative examples include JNK1α1, JNK1β1, JNK2α1, and JNK2β1 for the shorter forms, and JNK1α2, JNK1β2, JNK2α2, JNK2β2 for the longer variants. For JNK3, two main isoforms are typically expressed: JNK3α1 and JNK3α2, though up to three have been characterized, reflecting less extensive splicing compared to JNK1 and JNK2. The molecular weights of JNK isoforms vary based on splicing, ranging from approximately 46 for the shorter forms (e.g., JNK1α1, JNK1β1, JNK2α1, JNK2β1, and the 46 JNK3 variant) to 55 for the longer forms (e.g., JNK1α2, JNK1β2, JNK2α2, JNK2β2, and the 55 JNK3 variant), owing to the additional insertion. These isoforms exhibit differential expression patterns that contribute to the family's functional diversity. JNK1 and JNK2 are ubiquitously expressed across human tissues, with particularly high levels in the and , enabling broad involvement in cellular responses to . In contrast, JNK3 shows tissue-restricted expression, predominantly in the (e.g., neurons), testis, and heart, with lower levels in and minimal presence elsewhere. The JNK family demonstrates strong evolutionary conservation across mammals, with high sequence homology (over 85% identity among isoforms) to other mitogen-activated protein kinases (MAPKs), such as p38 and ERK, reflecting a shared ancestral role in stress signaling pathways. This conservation extends beyond mammals to invertebrates, including , where a single JNK ortholog () performs analogous functions, underscoring the ancient origins of the JNK signaling module.

Domain Architecture and Biochemical Properties

c-Jun N-terminal kinases (JNKs) are serine/ protein belonging to the (MAPK) family, characterized by a conserved N-terminal that spans approximately 300 and is responsible for ATP binding and substrate . This catalytic features the canonical bilobal architecture typical of protein , with an N-terminal lobe containing β-sheets for ATP coordination via a conserved residue and a C-terminal lobe housing the substrate-binding cleft. The is flanked by an activation loop, containing a Thr-Pro-Tyr (TPY) (Thr183-Pro184-Tyr185 in JNK1), which undergoes dual to induce a conformational change that aligns catalytic residues for optimal activity. Unlike other MAPKs such as ERK, JNKs possess a unique C-terminal extension of about 50-100 residues, which is intrinsically disordered and contributes to isoform-specific regulatory interactions without directly participating in . JNKs exhibit dual-specificity activity, preferentially phosphorylating substrates at a consensus of (Pro)-X-Ser/Thr-Pro [(P)-X-S/T-P], where the residues flanking the phosphorylatable serine or enhance recognition and efficiency. This preference distinguishes JNKs from other proline-directed s and is evident in substrates like c-Jun, where at Ser63 and Ser73 within such sequences amplifies transcriptional activation. JNKs can form dimers, a process facilitated by their domain and influenced by the C-terminal extension, which may promote autophosphorylation or stabilize active conformations in certain isoforms like JNK2α2. Additionally, JNKs engage in scaffold interactions, such as with the JNK-interacting protein 1 (JIP1), which binds via a to the domain's edge, enhancing specificity and localizing JNK signaling without requiring dimerization for basic activity. Crystal structures of JNK1, such as PDB entry 1UKI in complex with a JIP1 , reveal the active site's conformation, showing how the ATP-binding pocket accommodates inhibitors like SP600125 and how the loop's TPY repositions upon to open the substrate cleft. These structures highlight conserved residues like Asp169 in the catalytic loop and the Met111, which influence inhibitor selectivity and underscore the domain's adaptability for stress-responsive signaling. Isoform variations, such as longer C-terminal extensions in JNK1α versus JNK1β, subtly modulate these interactions but preserve core domain functionality.

Activation and Regulation

Upstream Kinase Cascade

The c-Jun N-terminal kinases (JNKs) function as the terminal kinases in a (MAPK) signaling cascade, where they are activated through dual on 183 (Thr183) and 185 (Tyr185) within their activation loop. This is catalyzed by two primary MAPK kinases (MAP2Ks): MAPK kinase 4 (MKK4, also known as SEK1) and MKK7. MKK4 and MKK7 exhibit complementary roles, with MKK4 preferentially phosphorylating the tyrosine residue and MKK7 the residue, MKK7 being essential for JNK activation in response to proinflammatory cytokines, while their combined action ensures efficient dual across JNK isoforms. Upstream of the MAP2Ks, multiple MAPK kinase kinases (MAP3Ks) initiate the cascade in response to diverse environmental stressors. Key MAP3Ks include , which is activated by through dissociation from ; mitogen-activated protein kinase/ERK kinase kinase 1 (MEKK1), which is regulated by Lys63-linked ubiquitination; mixed lineage kinase 3 (MLK3), activated downstream of Rho ; and transforming growth factor-β-activated kinase 1 (TAK1), which operates within a complex involving TAB1, TAB2, and TAB3. These MAP3Ks are triggered by stimuli such as (UV) radiation, , and cytokines including tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1), leading to sequential of MKK4 and MKK7. Scaffold proteins enhance the specificity and efficiency of the JNK by assembling MAP3Ks, MAP2Ks, and JNKs into multiprotein complexes. The JNK-interacting proteins (JIPs), including JIP1, JIP2, JIP3, and JIP4, bind JNK and MKK7 to facilitate , with JIP1 particularly promoting JNK activation in neuronal contexts. Additional adaptors such as partner of SH3 domain () and β-arrestin 2 organize the ; recruits MLKs and MKK4 to activate JNK1, while β-arrestin 2 scaffolds ASK1, MKK4, and JNK3 in response to stimulation. In addition to the canonical stress-induced pathway, JNKs can be activated non-canonically through receptor tyrosine kinases such as the epidermal growth factor receptor (EGFR) or G protein-coupled receptors, often in contexts promoting cell growth or motility, via adaptor-mediated recruitment of upstream kinases like MEKK1.

Inhibitory and Modulatory Mechanisms

c-Jun N-terminal kinases (JNKs) are subject to tight control through various inhibitory and modulatory mechanisms that prevent aberrant signaling. A primary mode of inactivation involves dephosphorylation of the activation loop residues Thr183 and Tyr185 by dual-specificity phosphatases (DUSPs), also known as mitogen-activated protein kinase phosphatases (MKPs). For instance, DUSP1 (MKP-1) and DUSP16 (MKP-7) specifically target these sites, attenuating JNK activity in response to stress signals; DUSP1 acts primarily in the nucleus, while DUSP16 functions in the cytoplasm. Additionally, serine/threonine phosphatases such as protein phosphatase 2A (PP2A) and protein phosphatase 2C (PP2C, including PPM1J) contribute to JNK suppression by dephosphorylating upstream components or directly interacting with JNK via docking motifs. Negative feedback loops further refine JNK signaling duration and amplitude. JNK activation induces the transcription of DUSPs like DUSP1 and DUSP5 through AP-1-dependent mechanisms, creating an autoregulatory circuit that limits prolonged JNK activity. Scaffold proteins such as JNK-interacting protein 4 (JIP4) sequester JNK in inactive complexes, preventing its interaction with substrates and upstream activators; phosphorylation of JIP4 by JNK itself can modulate this sequestration, forming context-dependent feedback. Cross-talk with other kinase pathways provides additional modulation. Phosphorylation of JNK by (PKA) or (PKC) on specific serine residues inhibits its activity, integrating or diacylglycerol signals to dampen JNK responses. Similarly, ubiquitin-mediated proteasomal degradation regulates JNK turnover, with E3 ligases like promoting the ubiquitination and degradation of JNK-phosphorylated substrates, indirectly modulating pathway output; JNK itself activates via phosphorylation, linking signaling to protein stability control. Scaffold-mediated sequestration also inactivates JNK through binding interactions. The 14-3-3 proteins bind to phosphorylated motifs on JNK or its partners, sequestering them in the and preventing translocation or substrate access, thereby inhibiting pro-apoptotic signaling. These mechanisms collectively ensure precise spatiotemporal control of JNK, balancing its roles in cellular .

Core Biological Functions

Transcriptional Regulation via AP-1

c-Jun N-terminal kinases (JNKs) play a central role in by phosphorylating the c-Jun at serine residues 63 and 73 (Ser63/Ser73) within its . This event, first identified as a key mechanism of JNK action, enhances c-Jun's transcriptional activity by promoting its dimerization with Fos family proteins to form the AP-1 complex and increasing the complex's affinity for TPA-responsive elements (TREs) in promoter regions. The modification stabilizes the AP-1 dimer and facilitates its binding to consensus DNA sequences (5'-TGACTCA-3'), thereby driving the expression of target genes involved in cellular responses to stress and growth signals. Beyond c-Jun, JNKs phosphorylate other transcription factors that contribute to AP-1-mediated regulation, including ATF2 at residues 69 and 71 (Thr69/Thr71), JunB, and Elk-1. of ATF2 by JNK augments its potential, often in heterodimeric complexes with c-Jun to modulate AP-1 activity. These modifications collectively lead to the upregulation of genes associated with , such as cyclin D1, and , including IL-6 and TNF-α, where AP-1 binding sites in their promoters are critical for JNK-dependent induction. For instance, JNK-activated AP-1 drives cyclin D1 expression to promote cell cycle progression in response to mitogenic stimuli. The transcriptional outcomes of JNK-AP-1 signaling exhibit context-specificity, varying with cellular conditions. Under , JNK phosphorylation enhances AP-1 activation of pro-apoptotic genes like FasL and Bim, which contain functional TREs and contribute to pathways. In developmental or proliferative contexts, the same pathway upregulates growth-promoting genes, supporting tissue differentiation and expansion. This versatility arises from the combinatorial nature of AP-1 dimers and their integration with other signaling pathways, including crosstalk with and . For example, JNK-AP-1 can synergize with to amplify inflammatory , while interactions with p53 modulate stress-induced transcription in a cooperative manner.

Mediation of Apoptosis and Cell Survival

c-Jun N-terminal kinases (JNKs) play a pivotal dual role in the regulation of and cell survival, exerting pro-apoptotic effects through mitochondrial pathways while also promoting survival under certain stress conditions. This duality arises from JNK's ability to phosphorylate key regulators of the intrinsic apoptotic pathway and to translocate to mitochondria, influencing the balance between and persistence. In pro-apoptotic contexts, JNK promotes the activation of proteins to facilitate mitochondrial outer membrane permeabilization (MOMP). Specifically, JNK phosphorylates the BH3-only protein BIM, enhancing its binding to anti-apoptotic members like and thereby potentiating BAX oligomerization and activation. This phosphorylation event sensitizes neurons to apoptosis by amplifying BAX-dependent release from mitochondria. Additionally, JNK induces the translocation of BAX to mitochondria by phosphorylating 14-3-3 proteins, which normally sequester BAX in the ; disruption of this interaction allows BAX to integrate into the mitochondrial membrane, triggering MOMP and subsequent efflux. JNK itself translocates to the mitochondria upon activation, where it directly contributes to release, as demonstrated in models of where JNK2 mitochondrial localization correlates with apoptotic commitment. JNK's mitochondrial actions are further amplified through scaffold-dependent mechanisms involving the protein SAB (SH3BP5), a JNK-interacting partner localized to the outer mitochondrial membrane. Upon stress-induced activation, JNK docks to SAB, forming a feed-forward loop that sustains JNK phosphorylation and elevates (ROS) production; this ROS amplification exacerbates mitochondrial dysfunction and promotes . The JNK-SAB interaction is critical for this process, as disrupting SAB-mediated docking prevents JNK's pro-apoptotic signaling at the mitochondria without affecting its nuclear functions. Conversely, JNK can exert anti-apoptotic effects by phosphorylating the pro-apoptotic member BAD at Thr201, which inhibits BAD's ability to heterodimerize with anti-apoptotic proteins like and thereby blocks its death-promoting activity. This phosphorylation occurs in cytokine-dependent cells, where JNK suppresses and supports signaling. The temporal dynamics of JNK are crucial in determining cell fate: transient JNK signaling generally promotes by integrating protective responses, whereas prolonged shifts toward through non-transcriptional mitochondrial disruption and induction of pro-death factors. Sustained JNK activity, often triggered by persistent stressors like () stress, upregulates CHOP (C/EBP homologous protein), which in turn transcriptionally induces the BH3-only protein to amplify BAX/BAK and cytochrome c release. This prolonged phase overrides signals, committing cells to programmed death via the intrinsic pathway.

Control of Proliferation and Differentiation

c-Jun N-terminal kinases (JNKs), particularly JNK1 and JNK2 isoforms, play a pivotal role in promoting by facilitating the G1/S phase transition through c-Jun-mediated upregulation of expression. In hepatocytes, JNK activation drives transcription via c-Jun phosphorylation, enabling quiescent cells to enter the following stimuli such as partial . Similarly, JNK2 contributes to G1/S progression in various cell types by enhancing c-Jun activity, which in turn boosts levels and supports proliferative responses. In contrast, JNK3 exerts inhibitory effects on specifically in neurons, where its activation promotes stress responses that limit to favor and maintenance of neuronal identity. Isoform-specific functions of JNKs further highlight their nuanced control over proliferation and differentiation in distinct cellular contexts. JNK1, acting downstream of (EGFR) signaling, supports keratinocyte proliferation by integrating mitogenic cues that drive epidermal renewal. Meanwhile, JNK2 is essential for T-cell differentiation, particularly in the commitment to T helper 1 (Th1) lineages, where it modulates production and effector functions through c-Jun-dependent pathways. These differential roles underscore how JNK isoforms fine-tune lineage-specific responses without uniform effects across cell types. In embryonic development, JNK signaling regulates key morphogenetic processes such as and by integrating with the planar cell polarity () pathway. JNK activation downstream of non-canonical Wnt/PCP cues promotes convergent extension movements during , ensuring proper tissue elongation and axis formation. In , JNK coordinates polarized cell behaviors, including apical constriction and directed migration, to prevent defects like . This involvement in PCP-mediated polarity is conserved across species, as evidenced by JNK's role in , where it balances and cell fate decisions. JNK also drives by enhancing re-epithelialization and migration, critical for tissue repair. In , JNK promotes migratory responses to cover wound beds, facilitated by growth factor-induced activation that aligns with upstream kinase cascades. Concurrently, JNK signaling in fibroblasts stimulates motility via pathways involving (bFGF) and Rac1, contributing to formation without overlapping stress-induced arrest mechanisms.

Specialized Physiological Roles

Response to DNA Damage and Repair

c-Jun N-terminal kinases (JNKs) play a critical role in the cellular response to DNA damage by integrating signals from DNA lesion-sensing kinases to coordinate repair mechanisms and cell cycle checkpoints. Upon exposure to DNA-damaging agents such as ultraviolet (UV) radiation or ionizing radiation (IR), ATM and ATR kinases detect double-strand breaks (DSBs) or replication stress, respectively, leading to activation of the upstream kinase ASK1, which in turn phosphorylates and activates MKK4 and MKK7. These MAP2Ks subsequently phosphorylate JNKs at Thr183 and Tyr185, initiating the JNK signaling cascade to propagate the DNA damage response. JNKs contribute to DNA repair by modulating key effectors in multiple pathways. In nucleotide excision repair (NER), JNKs enhance the efficiency of transcription-coupled NER (TC-NER) by phosphorylating the microprocessor component DGCR8 at Ser153 following UV-induced stalling of RNA polymerase II, thereby facilitating the recruitment of repair factors to remove bulky lesions like cyclobutane pyrimidine dimers. Additionally, JNKs support homologous recombination (HR) for DSB repair through phosphorylation of SIRT6, which recruits PARP1 to damage sites and promotes end resection and repair fidelity under oxidative stress. JNKs also stabilize p53 by direct phosphorylation at Ser20, disrupting its interaction with MDM2 and enabling p53-dependent transcription of repair genes. To enforce cell cycle checkpoints during repair, JNKs mediate G2/M arrest by phosphorylating Cdc25C , inhibiting its activity and preventing premature activation of CDK1-cyclin B1 complexes; this process is complemented by JNK-induced stabilization of , which transcriptionally upregulates 14-3-3σ to sequester CDK1 in the . If DNA lesions persist unrepaired, sustained JNK activation shifts toward promoting through p53-dependent mechanisms, ensuring elimination of potentially mutagenic cells. Isoform-specific contributions further refine JNK functions in . JNKs support DSB repair by phosphorylating SIRT6 to enhance recruitment and pathway engagement following . In contrast, JNKs are implicated in (BER), where they coordinate the removal of oxidative base lesions like those induced by , maintaining genomic stability in stressed cells such as pancreatic β-cells.

Contribution to Aging and

c-Jun N-terminal kinases (JNKs) contribute to cellular aging by promoting telomere attrition through inhibition of activity. JNK activation leads to repression of the human telomerase reverse transcriptase (hTERT) gene via the complex, which binds to the hTERT promoter and suppresses its expression, thereby reducing telomerase levels and accelerating shortening. This mechanism is evident in various cell types, where sustained JNK signaling exacerbates replicative by limiting maintenance. Additionally, JNK disrupts the function of complexes, the protective protein assemblies at , leading to increased telomere instability and dysfunction that further drives age-related cellular decline. Persistent JNK activation plays a central role in aspects of , including the (SASP), through enhancement of pro-inflammatory cytokines such as IL-6 and IL-8, which amplify SASP components and reinforce a senescent state in neighboring cells via . This persistent signaling creates a loop that sustains arrest and contributes to the chronic observed in aging tissues. However, JNK often antagonizes p16INK4a expression and senescence induction in stress contexts, such as doxorubicin-treated cells, highlighting its context-dependent effects. In model organisms, JNK exhibits context-dependent effects on lifespan regulation. In Caenorhabditis elegans and Drosophila melanogaster, JNK signaling extends longevity by interacting with FOXO transcription factors, promoting nuclear localization of FOXO and activation of stress resistance genes that delay aging. In contrast, mammalian studies reveal a more complex role, where knockout of JNK2 isoform delays age-related phenotypes and extends healthspan by reducing senescence accumulation in tissues like the liver and brain. Recent investigations highlight JNK's integration with circadian rhythms and mitochondrial dynamics in aging. JNK phosphorylates the period 2 (PER2) protein, disrupting oscillations and linking disrupted rhythms to accelerated , as observed in aged tissues. Furthermore, mitochondrial (ROS) activate JNK in a feedback loop, where JNK in turn amplifies ROS production, hastening in stem cells and contributing to organismal aging.

Involvement in Inflammation and Immunity

c-Jun N-terminal kinases (JNKs) play a pivotal role in innate immune signaling, particularly through activation by Toll-like receptors (TLRs) and the interleukin-1 receptor (IL-1R). Upon ligand binding, these receptors recruit the adaptor protein TRAF6, which ubiquitinates and activates the upstream kinase TAK1, leading to JNK phosphorylation and downstream signaling. This cascade is essential for propagating inflammatory signals in immune cells, including the production of pro-inflammatory mediators. In adaptive immunity, activated JNK phosphorylates the c-Jun, a component of the AP-1 complex, thereby promoting the expression of key cytokines such as interleukin-2 (IL-2) and interferon-gamma (IFN-γ) in T cells. JNK2, in particular, is required for IFN-γ production by CD4+ T cells, supporting Th1 differentiation and effector functions during immune responses. This mechanism enhances T cell activation and proliferation in response to antigenic stimulation. JNK isoforms exhibit distinct contributions to macrophage activation, a cornerstone of innate immunity. JNK2 drives (pro-inflammatory) macrophage polarization in response to stimuli like (LPS), promoting the production of (ROS) and cytokines that amplify defenses. In contrast, JNK1 facilitates by regulating cytoskeletal rearrangements and engulfment of pathogens or debris, ensuring efficient clearance in inflammatory environments. These isoform-specific roles underscore JNK's versatility in modulating macrophage phenotypes and functions. In autoimmune diseases, dysregulated JNK signaling contributes to pathogenesis, as seen in (RA) where hyperactive JNK forms a loop with tumor necrosis factor-alpha (TNF-α). TNF-α activates JNK via its receptor, leading to sustained production of matrix metalloproteinases and that perpetuate synovial inflammation and joint destruction. JNK inhibitors have shown promise in preclinical models by disrupting this loop and reducing RA severity. JNK also exerts regulatory effects on Th17 cell differentiation, balancing pro-inflammatory responses. While JNK activation via the MyD88 pathway promotes pathogenic Th17 cells in conditions like experimental autoimmune encephalomyelitis, it suppresses IL-17 expression through c-Jun-mediated inhibition in certain contexts, thereby limiting excessive Th17-driven . This dual role highlights JNK's importance in fine-tuning adaptive immune homeostasis. In antiviral immunity, JNK enhances type I (IFN) production by cooperating with at a point in the signaling . The TAK1-JNK pathway phosphorylates on serine 173, facilitating its activation, dimerization, and nuclear translocation to induce IFN-β transcription in response to viral infections. This mechanism amplifies innate antiviral defenses, including the expression of interferon-stimulated genes that restrict .

Pathophysiological Implications

Role in Cancer Development

c-Jun N-terminal kinases (JNKs) play a dual, context-dependent role in cancer development, exerting both oncogenic and tumor-suppressive effects across various malignancies. Sustained JNK activation promotes tumorigenesis in Ras-driven cancers, such as , where oncogenic K-Ras mutations trigger JNK signaling to enhance AP-1-dependent survival pathways, thereby supporting tumor and to . In , the JNK2 isoform specifically amplifies metastatic potential by regulating multiple receptor tyrosine kinases, including and c-Met, which facilitate tumor cell invasion and dissemination to distant sites like the lungs. These pro-oncogenic functions highlight JNK's contribution to malignant progression through sustained signaling that overrides stress-induced cell death. Conversely, JNKs mediate tumor suppression by inducing anoikis in detached epithelial cells, a process essential for preventing metastatic spread; ablation of JNK1 and JNK2 impairs this anoikis response, allowing survival of non-adherent cells and potentially fostering tumor dissemination. In , loss of JNK1/2 function in epithelial cells triggers biliary hyperproliferation and cholangiocarcinoma-like features, underscoring their role in restraining aberrant proliferation and maintaining hepatic homeostasis. Isoform-specific differences further modulate these effects: JNK1 promotes cell survival in by modulating arrest and apoptotic thresholds, enabling tumor persistence under stress. Recent studies emphasize isoform biases and therapeutic vulnerabilities. For instance, JNK inhibition exhibits anti-proliferative activity in cells, where it disrupts stem-like properties and reduces tumor initiation capacity, contrasting with JNK1's pro-survival role in other contexts. In KRAS-mutant , JNK inhibitors like SP600125 sensitize cells to such as by enhancing apoptotic pathways. Additionally, epigenetic silencing of JNK signaling through BRG1 suppression drives stemness and expansion, as BRG1 loss activates JNK-dependent pathways that sustain tumor-initiating cells.

Impact on Neurodegenerative Disorders

c-Jun N-terminal kinases (JNKs), particularly the neuron-specific isoform JNK3, play a pivotal role in axon degeneration following injury by activating the dual leucine zipper kinase (DLK)-MKK4/7 signaling cascade, which drives Wallerian degeneration. In this process, axonal injury triggers DLK activation, leading to phosphorylation and activation of MKK4 and MKK7, which in turn phosphorylate and activate JNK3 to promote pro-degenerative transcription factors such as c-Jun, resulting in the dismantling of the axonal cytoskeleton. This pathway is essential for the timed degeneration of severed axons, and its disruption, such as through DLK or JNK3 inhibition, delays Wallerian degeneration in various neuronal models. In (AD), JNKs contribute to pathology by phosphorylating at serine 422 (Ser422), a modification that promotes tau aggregation and formation. This phosphorylation event is upregulated in AD brains and is induced by amyloid-β (Aβ) oligomers, which activate JNK signaling to enhance tau pathology and neuronal , linking JNK to Aβ-mediated toxicity. Additionally, JNK2 isoform activation in exacerbates in AD by promoting the release of pro-inflammatory cytokines in response to Aβ plaques, thereby amplifying neuronal damage. In (), JNK1 and JNK2 isoforms mediate the toxicity of (MPTP) and α-synuclein through pathways involving mitochondrial dysfunction. MPTP exposure activates JNK1/2, leading to neuron loss via induction of (COX-2) and subsequent in mitochondria. Similarly, mutant α-synuclein triggers JNK activation, impairing mitochondrial protein import and dynamics, which contributes to energy failure and neuronal death in models. Studies up to 2023 highlight the therapeutic potential of JNK inhibitors in (ALS) models, where they protect cortical neurons from degeneration linked to TDP-43 aggregation. In human iPSC-derived motor neurons harboring ALS mutations, JNK inhibition reduces TDP-43 phosphorylation and aggregation, preserving neuronal viability and mitigating formation. These findings build on JNK's role in , suggesting isoform-specific inhibitors could attenuate TDP-43-induced toxicity in ALS.

Therapeutic Targeting and Inhibitors

Small-molecule inhibitors represent a primary strategy for therapeutically targeting c-Jun N-terminal kinases (JNKs), with efforts focused on modulating their activity to treat diseases involving dysregulated JNK signaling, such as cancer and neurodegeneration. Early inhibitors like SP600125, an anthrapyrazolone compound, act as broad ATP-competitive antagonists of JNK1, JNK2, and JNK3, exhibiting IC50 values in the low micromolar range and demonstrating selectivity over other MAPKs while blocking JNK-mediated of substrates like c-Jun. Substrate-competitive inhibitors, such as BI-78D3, disrupt JNK interactions with docking proteins like JIP1, achieving potent inhibition (IC50 = 280 nM) with over 100-fold selectivity against p38α and no activity at or PI3K, thereby offering an alternative to ATP-site binding that may reduce off-target effects. For isoform selectivity, covalent inhibitors like JNK-IN-8 target a conserved residue (Cys116) in the ATP-binding pocket, providing irreversible inhibition of JNK1 (IC50 = 4.7 nM), JNK2 (18.7 nM), and JNK3 (1 nM) with greater than 10-fold selectivity over other kinases, enabling more precise modulation of specific JNK isoforms implicated in . Clinical translation of JNK inhibitors has faced hurdles, with several candidates advancing to trials for inflammatory and fibrotic conditions but yielding mixed results. For instance, CC-90001, a selective JNK1 , was evaluated in a phase II trial for (a condition linked to JNK-driven ), where 200 mg and 400 mg daily doses showed numerical improvements in percent predicted forced (ppFVC) over 24 weeks compared to , though it failed to meet the primary of significant FVC decline reduction, leading to discontinuation of development. In , where JNK contributes to synovial inflammation, preclinical models demonstrate efficacy of inhibitors like SP600125 in reducing swelling and degradation, but no JNK-specific agents have progressed beyond early-phase testing in humans due to toxicity concerns. For neurodegeneration, such as (ALS), the brain-penetrant SR-3306 has shown preclinical promise by completely preventing death induced by ALS in mouse models, highlighting potential for isoform-selective (JNK3-focused) approaches in neural protection. Key challenges in JNK inhibitor development include achieving isoform specificity, as JNK1/2/3 share over 90% sequence identity in their domains, complicating selective targeting and risking from broad inhibition—such as JNK1's role in or JNK3's in neuronal survival. Combination therapies address these limitations by leveraging JNK inhibition to sensitize cells to other agents; for example, JNK-IN-8 enhances chemotherapy efficacy in pancreatic ductal models by promoting biogenesis and , reducing tumor growth without standalone . Recent advances emphasize novel modalities beyond traditional small molecules. Proteolysis-targeting chimeras (PROTACs) designed against JNK1, such as compound PA2, induce ubiquitin-mediated degradation of JNK1, achieving potent degradation (DC50 = 10 ). Additionally, allosteric modulators targeting the JIP1-JNK interaction, exemplified by BI-78D3's disruption of scaffold assembly, have evolved with 2024 developments in reversible covalent inhibitors that bind non-ATP sites, offering enhanced selectivity for therapeutic applications in and .

References

  1. [1]
    The c-Jun Kinase/Stress-activated Pathway: Regulation, Function ...
    c-Jun N-terminal kinases (JNKs), also referred to as stress-activated kinases (SAPKs), were initially characterized by their activation in response to cell ...
  2. [2]
    Uses for JNK: the Many and Varied Substrates of the c-Jun N ...
    The c-Jun N-terminal kinases (JNKs) are members of a larger group of serine/threonine (Ser/Thr) protein kinases from the mitogen-activated protein kinase family ...
  3. [3]
    c-Jun N-terminal Kinase (JNK) Signaling as a Therapeutic Target for ...
    Jan 12, 2016 · c-Jun N-terminal kinases (JNKs) are a family of protein kinases that play a central role in stress signaling pathways implicated in gene ...
  4. [4]
    MAPK8 mitogen-activated protein kinase 8 [ (human)] - NCBI
    Sep 14, 2025 · The protein encoded by this gene is a member of the MAP kinase family. MAP kinases act as an integration point for multiple biochemical ...
  5. [5]
    MAPK9 mitogen-activated protein kinase 9 [ (human)] - NCBI
    Sep 5, 2025 · This study was designed to investigate the effect of c-Jun N-terminal kinase (JNK) 1 and 2 on TGF-beta(1)-regulated CTGF gene expression and ...Missing: MAPK10 | Show results with:MAPK10
  6. [6]
    MAPK10 mitogen-activated protein kinase 10 [ (human)] - NCBI
    Sep 5, 2025 · This kinase is specifically expressed in a subset of neurons in the nervous system, and is activated by threonine and tyrosine phosphorylation.
  7. [7]
    Selective interaction of JNK protein kinase isoforms with ... - NIH
    Ten JNK isoforms were identified in human brain by molecular cloning. These protein kinases correspond to alternatively spliced isoforms derived from the JNK1, ...Missing: review | Show results with:review
  8. [8]
    JNK Signaling: Regulation and Functions Based on Complex ...
    For JNK3, there are 8 possible isoforms (including the longer [L] and shorter [S] N-terminal extensions), but only 3 isoforms have been characterized to date.
  9. [9]
    Differential activation of JNK1 isoforms by TRAIL receptors modulate ...
    JNK1α1, JNK1β1, JNK2α1 and JNK2β1 isoforms all have a molecular weight of 46 kDa, whereas JNK1α2, JNK1β2, JNK2α2 and JNK2β2 isoforms are larger due to an ...
  10. [10]
    The Role of the Dysregulated JNK Signaling Pathway in the ... - MDPI
    The molecular weights of the three JNK isoforms (JNK1, JNK2, and JNK3) are either 46 kDa or 55 kDa. There is a COOH-terminal extension in some JNK isoforms [40] ...
  11. [11]
    JNK3 as Therapeutic Target and Biomarker in Neurodegenerative ...
    The c-Jun N-terminal kinase 3 (JNK3) is the JNK isoform mainly expressed in the brain. It is the most responsive to many stress stimuli in the central nervous ...
  12. [12]
    The crystal structure of JNK from Drosophila melanogaster reveals ...
    Sep 16, 2015 · Similar to mammalian cells, the JNK signaling pathway is also conserved in Drosophila. The Drosophila JNK pathway consists of Drosophila JNK or ...
  13. [13]
    Effects of Dual-Specificity Phosphatases (DUSPs) on the JNK Pathway
    Dec 6, 2019 · In this review, we will discuss the dynamics of phosphorylation/dephosphorylation, the mechanism of JNK pathway regulation by DUSPs, and the new ...
  14. [14]
    JNK Signaling: Regulation and Functions Based on Complex ...
    Jul 27, 2016 · Several cytoskeletal proteins show conserved motifs surrounding their JNK phosphorylation sites. In most cases, it is unclear whether the ...
  15. [15]
    Phosphorylation Dynamics of JNK Signaling: Effects of Dual ... - NIH
    Dual specificity phosphatases (DUSPs) regulate the magnitude and duration of signal transduction of the JNK pathway by dephosphorylating their substrates.
  16. [16]
    Cyclic AMP inhibits JNK activation by CREB-mediated induction of c ...
    Jun 20, 2008 · The inhibition of JNK activity and phosphorylation by cAMP was not restricted to UV. Immunoblotting analysis and immune complex kinase assays ...
  17. [17]
    JNK antagonizes Akt-mediated survival signals by phosphorylating ...
    14-3-3 promotes cell survival by sequestering and inactivating several proapoptotic proteins, including Bad and FOXO3a, after their phosphorylation by survival- ...
  18. [18]
    Kinase-Independent Feedback of the TAK1/TAB1 Complex on ... - NIH
    Thus, by directly promoting BCL10 degradation, TAK1 counterbalances NF-κB and JNK signals essential for the activation and survival of lymphocytes and CARMA1- ...
  19. [19]
    and UV-responsive protein kinase that binds and potentiates the c ...
    Phosphorylation results in dissociation of the c-Jun-JNK complex. Mutations that disrupt the kinase-binding site attenuate the response of c-Jun to Ha-Ras and ...
  20. [20]
    Uses for JNK: the Many and Varied Substrates of the c-Jun N ...
    Dec 1, 2006 · The alternative names for these JNK isoforms are provided, alongside information on their splice forms and positions on the mouse chromosomes.
  21. [21]
    Transcriptional regulation of the cyclin D1 gene at a glance - NIH
    The human cyclin D1 promoter contains a consensus AP-1 site, at −903 bp, which is regulated by Fos and Jun (Albanese et al., 1995; Shen et al., 2007). Jun ...
  22. [22]
    AP-1 subunits: quarrel and harmony among siblings
    Dec 1, 2004 · Among the pro-apoptotic targets of Jun are the genes that encode FasL and TNF-α, which both contain AP-1-binding sites. How and when the JNK ...
  23. [23]
    AP-1 Transcription Factors as Regulators of Immune Responses in ...
    Importantly, JNK-mediated phosphorylation of c-Jun is required for c-Jun and FOXP3 interaction. In addition, FOXP3 also interacts with c-Fos, but not JunB and ...
  24. [24]
    Signaling to NF-κB - Genes & Development
    NF-κB induction can lead to the synthesis of certain effectors that inhibit the JNK pathway, thereby limiting JNK/AP1 activation. One such JNK-inhibiting factor ...
  25. [25]
    Crosstalk in NF-κB signaling pathways | Nature Immunology
    Jul 19, 2011 · Activation of AP-1 is promoted by the mitogen-activated protein kinases (MAPKs) Jnk, Erk1-Erk2 and p38, and activated AP-1 affects cell survival ...
  26. [26]
    Role of JNK activation in apoptosis: A double-edged sword - Nature
    Jan 1, 2005 · Recent studies reveal that JNK can suppress apoptosis in IL-3-dependent hematopoietic cells via phosphorylation of the proapoptotic Bcl-2 family protein BAD.
  27. [27]
    Mitochondrial P-JNK target, SAB (SH3BP5), in regulation of cell death
    Mar 15, 2024 · The JNK-SAB-ROS feed forward activation loop sustains the JNK activation and incrementally increases ROS production, leading to apoptosis via ...
  28. [28]
    CHOP and AP-1 cooperatively mediate PUMA expression during ...
    These data suggest that CHOP and AP-1 cooperatively mediate PUMA induction during hepatocyte lipoapoptosis.
  29. [29]
    Neuronal Apoptosis Induced by Endoplasmic Reticulum Stress Is ...
    Dec 15, 2010 · Specifically, we demonstrate that CHOP binds to the PUMA promoter during ER stress and that CHOP knockdown attenuates PUMA induction and ...
  30. [30]
    Activation of the JNKs/ATM-p53 axis is indispensable for ... - Nature
    Jul 25, 2022 · JNKs are implicated in the activation of the DNA damage response of dermal fibroblasts after UVB treatment. We demonstrated that UVB radiation ...
  31. [31]
    DGCR8 mediates repair of UV-induced DNA damage independently ...
    When DNA is damaged by UV, RNAPII stalls and triggers phosphorylation of S153 on DGCR8, which facilitates removal of UV-induced DNA lesions by TC-NER and ...
  32. [32]
    DNA damage and oxidant stress activate p53 through differential ...
    Aug 20, 2021 · JNK phosphorylates p53 on Ser20 ... Chehab, et al. Phosphorylation of Ser-20 mediates stabilization of human p53 in response to DNA damage.
  33. [33]
    JNK-mediated Phosphorylation of Cdc25C Regulates Cell Cycle ...
    These findings indicate that G2/M arrest (delay in mitosis progression), which is central in the cellular response to DNA damage, is mediated in part by JNK- ...Missing: 3σ | Show results with:3σ
  34. [34]
    Repair of Nitric Oxide-damaged DNA in β-Cells Requires JNK ...
    Previous reports have shown that DNA damage induced by oxidizing agents, such as nitric oxide, is repaired through the base excision repair pathway (23), but ...Missing: JNK1 double JNK2
  35. [35]
    Jnk2 Effects on Tumor Development, Genetic Instability and ...
    May 3, 2010 · Together, these data support that JNK2 prevents replicative stress by coordinating cell cycle progression and DNA damage repair mechanisms.
  36. [36]
  37. [37]
  38. [38]
  39. [39]
  40. [40]
    Proteostasis failure and mitochondrial dysfunction leads to ...
    We uncovered a role of c-Jun N-terminal kinase (JNK) in driving senescence as a consequence of dysfunctional mitochondria and ROS.
  41. [41]
    Interleukin-1 and TRAF6-dependent activation of TAK1 in the ...
    We conclude that IL-1β can induce the activation of TAK1 in two ways, only one of which requires the binding of K63-Ub chains to TAB2/3.
  42. [42]
    The TAK1-JNK cascade is required for IRF3 function in the innate ...
    Jan 20, 2009 · This study demonstrates that the TAK1-JNK cascade is required for IRF3 function, in addition to TBK1/IKKɛ, uncovering a new mechanism for mitogen-activated ...Tak1, Jnk1 And Jnk2 Can... · Jnk Can Directly... · Jnk Regulates Irf3 Activity...
  43. [43]
    TAK1 targeting by glucocorticoids determines JNK and IκB ... - NIH
    These findings identify TAK1 as a novel target for glucocorticoids that integrates their anti-inflammatory action in innate immunity signaling pathways.
  44. [44]
    c-Jun NH2-Terminal Kinase (JNK)1 and JNK2 Have Distinct Roles ...
    These results indicate that JNK1 and JNK2 have distinct functions in CD4+ T cell differentiation. In this study we show that Jnk2 − / − CD8+ T cells ...
  45. [45]
    The Effects of TLR Activation on T-Cell Development and ...
    Activated Th1 cell can produce IFN-γ and IL-2 to help CD8+ effector T-cell functioning. ... The TAK1/TABs complex also phosphorylates and activates c-Jun N- ...
  46. [46]
    Macrophage polarization and its role in the pathogenesis of acute ...
    Jul 10, 2020 · JNK is reportedly required for M1-related inflammation and fibrosis, insulin resistance, macrophage infiltration, M1 macrophage polarization, ...
  47. [47]
    TLR2 mediates phagocytosis and autophagy through JNK signaling ...
    Collectively, our results indicate that TLR2 may be critical for phagocytosis and autophagy through JNK signaling pathway, and provide an underlying mechanistic ...Missing: JNK1 | Show results with:JNK1
  48. [48]
    miR-185-5p Regulates Inflammation and Phagocytosis through ...
    Mar 7, 2022 · Silencing of JNK could dramatically inhibit macrophage phagocytosis [11,12]. Targeting JNK pathway would be a potential therapeutic approach in ...
  49. [49]
    The Macrophage Switch in Obesity Development - Frontiers
    There is a crosstalk between the two isoforms of JNK (JNK1 and JNK2) ... Molecular mechanisms that influence the macrophage m1-m2 polarization balance.
  50. [50]
    c-Jun N-Terminal Kinase in Inflammation and Rheumatic Diseases
    Taken together, these data suggest that the role of JNK in T cell immune responses is to reduce proliferative responses of the activated THz cells and to ...
  51. [51]
    The JNK pathway represents a novel target in the treatment of ... - NIH
    The pathophysiology of rheumatoid arthritis (RA) is characterized by excess production of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), ...
  52. [52]
    Activation of c-Jun N-Terminal Kinase, a Potential Therapeutic ... - NIH
    Nov 12, 2020 · JNK is one of the crucial downstream signaling molecules of various immune triggers, mainly proinflammatory cytokines, in autoimmune arthritic conditions.
  53. [53]
    Activation of the MyD88-JNK pathway promotes pathogenetic Th17 ...
    Jun 13, 2025 · Activation of the MyD88-JNK pathway by IL-1β and IL-23 promotes activin-A production in pathogenic Th17 cells and exacerbates EAE.
  54. [54]
    The JNK Signaling Pathway in Inflammatory Skin Disorders ... - MDPI
    The c-Jun N-terminal kinases (JNKs), with its members JNK1, JNK2, and JNK3, is a subfamily of (MAPK) mitogen-activated protein kinases.
  55. [55]
    MALT1 regulates Th2 and Th17 differentiation via NF-κB and JNK ...
    Jul 27, 2022 · MALT1 regulates Th2 and Th17 differentiation via NF-κB and JNK pathways, as well as correlates with disease activity and treatment outcome in ...Abstract · Methods · Results · Discussion
  56. [56]
    Adenovirus Induction of IRF3 Occurs through a Binary Trigger ...
    We demonstrate that maximal JNK/TBK1/IRF3 stimulation by rAdV depends on an intact type I interferon (IFN) signaling cascade.
  57. [57]
    Therapeutic effect of c‐Jun N‐terminal kinase inhibition on ... - NIH
    We next investigated the role of Ras in JNK activation in pancreatic cancer because oncogenic K‐ras mutation is known to be highly prevalent in pancreatic ...
  58. [58]
    c-Jun N-terminal Kinase 2 Regulates Multiple Receptor Tyrosine ...
    These studies identify JNK2 as a tumor and host target to inhibit breast cancer growth and metastasis.
  59. [59]
    JNK promotes epithelial cell anoikis by transcriptional and post ...
    Developmental morphogenesis, tissue injury, and oncogenic transformation can cause the detachment of epithelial cells. These cells are eliminated by a ...Missing: protects | Show results with:protects
  60. [60]
    Loss of c‐Jun N‐terminal Kinase 1 and 2 Function in Liver Epithelial ...
    Apr 16, 2020 · Loss of c‐Jun N‐terminal Kinase 1 and 2 Function in Liver Epithelial Cells Triggers Biliary Hyperproliferation Resembling Cholangiocarcinoma
  61. [61]
    JNK supports survival in melanoma cells by controlling ... - PubMed
    JNK supports survival in melanoma cells by controlling cell cycle arrest and apoptosis. Pigment Cell Melanoma Res. 2008 Aug;21(4):429-38. doi: 10.1111/j.1755 ...Missing: pro- | Show results with:pro-
  62. [62]
    Targeting JNK for therapeutic depletion of stem-like glioblastoma cells
    Jul 19, 2012 · Here we show that targeting JNK in vivo, the activity of which is required for the maintenance of stem-like glioblastoma cells, via transient, systemic ...Missing: JNK3 | Show results with:JNK3
  63. [63]
    The Role and Efficacy of JNK Inhibition in Inducing Lung Cancer ...
    Jun 18, 2024 · It is known that A549 is a RAS-mutated cell line. The same as in A549 dependence of the role of JNK on cisplatin concentration was found with ...<|control11|><|separator|>
  64. [64]
    JNK pathway plays a critical role for expansion of human colorectal ...
    Therefore, the JNK pathway plays a critical role for expansion and stemness of human CRC cells in the context of BRG1 suppression, and thus a combined blockade ...
  65. [65]
    Palmitoylation couples the kinases DLK and JNK3 to facilitate ...
    Mar 29, 2022 · Here, we identified two ways through which DLK is coupled to the neural-specific isoform JNK3 to control prodegenerative signaling.
  66. [66]
    β-Amyloid Oligomers Induce Phosphorylation of Tau and ...
    Jul 15, 2009 · We report Aβ oligomers significantly increased active JNK and phosphorylation of IRS-1 (Ser616) and tau (Ser422) in cultured hippocampal neurons.
  67. [67]
    JNK & Tau Phosphorylation in Alzheimer's Disease
    Our findings support the fundamental role of JNK in the regulation of tau hyperphosphorylation and subsequently in AD pathogenesis.
  68. [68]
    JNK-mediated induction of cyclooxygenase 2 is required for ...
    Examination of various JNK-deficient mice shows that both JNK2 and JNK3, but not JNK1, are required for MPTP-induced c-Jun activation and dopaminergic cell ...
  69. [69]
    Mutant α-synuclein causes death of human cortical neurons ... - NIH
    Mar 5, 2024 · Mutant α-synuclein causes death of human cortical neurons via ERK1/2 and JNK activation. Hidefumi Suzuki.
  70. [70]
    Screens in aging-relevant human ALS-motor neurons identify ...
    Jan 4, 2024 · We identify a neuroprotective compound and show that MAP4Ks may serve as therapeutic targets for treating ALS.
  71. [71]
    The JNK/c-Jun signaling axis contributes to the TDP-43-induced cell ...
    Dysregulation of transactive response DNA-binding protein-43 (TDP-43) is closely linked to the pathogenesis of amyotrophic lateral sclerosis (ALS) and ...Missing: cortical 2024 2025