The JNK signal transduction pathway

https://doi.org/10.1016/j.ceb.2007.02.001Get rights and content

The c-Jun NH2-terminal kinases (JNKs) are an evolutionarily conserved sub-group of mitogen-activated protein (MAP) kinases. Recent studies have improved our understanding of the physiological function of the JNK pathway. Roles of novel molecules that participate in the JNK pathway have been defined and new insight into the role of JNK in survival signaling, cell death, cancer and diabetes has been achieved.

Introduction

The response of a cell to changes in its environment is induced, in part, by a diverse array of intracellular signaling pathways. These pathways serve to relay, amplify and integrate signals from extracellular stimuli, ultimately resulting in a genomic and physiological response. In mammalian systems, these responses include cellular proliferation, differentiation, development, the inflammatory response and apoptosis. Mitogen-activated protein (MAP) kinases are one such family of signaling proteins.

The c-Jun NH2-terminal kinase (JNK) pathway represents one sub-group of MAP kinases that is activated primarily by cytokines and exposure to environmental stress [1, 2]. Specific stimuli trigger the activation of MAP3Ks, which then phosphorylate and activate the MAP2K isoforms MKK4 and MKK7, which in turn phosphorylate and activate JNK [1, 2]. Components of the JNK pathway can be organized into signaling complexes, mediated by one of the protein kinases (e.g. a MAP3K or a MAP2K) or by a scaffold protein, for example a member of the JNK-interacting protein (JIP) family [3]. A major target of the JNK signaling pathway is the activator protein-1 (AP-1) transcription factor, which is activated, in part, by the phosphorylation of c-Jun and related molecules [1, 2]. This review aims to highlight recent progress in JNK-related research; we refer the reader to earlier reviews [1, 2, 3] for references and discussion of previous work.

Section snippets

Novel components of the JNK signaling pathway

Recent research has provided new insights into regulatory components of the JNK signaling pathway. These fall broadly into three categories: upstream regulators (e.g. MAP3Ks); down-stream inhibitors (e.g. phosphatases); and scaffold proteins (e.g. JIPs).

The canonical JNK signaling cascade has been well characterized; however, the specific role of MAP3K in the response to various stimuli remains largely unresolved. Recent studies of the MAP3K isoform transforming growth factor-β activated kinase

JNK in cell death

A role for JNK in apoptosis is well established [1, 2]; however, the mechanism by which this occurs is controversial and appears to be stimulus- and tissue-specific [35]. One explanation for some of the differences observed could be the temporal aspect of JNK activation, and two recent studies have addressed this issue [36]. First, Chang and colleagues have described how sustained, but not transient, JNK activation promotes TNF-α killing via the E3 ubiquitin ligase Itch-mediated degradation of

Mechanism of JNK-induced activation of the mitochondrial apoptotic pathway

Primary fibroblasts prepared from Jnk1−/−Jnk2−/− embryos and from Mkk4−/−Mkk7−/− embryos exhibit marked defects in stress-induced apoptosis [46, 47]. Detailed analysis demonstrated that the apoptosis defect was associated with failure to release mitochondrial pro-apoptotic proteins, including cytochrome c. Indeed, micro-injection experiments demonstrated that the mutant cells did not exhibit defects in apoptosis if cytochrome c was directly injected into the cytoplasm [47]. These studies

JNK in cancer

JNK is implicated in oncogenic transformation; however, its role in tumor development remains controversial [66]. A role for the JNK pathway in tumorigenesis is supported by the high levels of JNK activity found in several cancer cell lines [66]. Indeed, in a recent study using a Drosophila model of tumor formation, oncogenic Raf and JNK were shown to cooperate to induce massive hyperplasia [67]. Studies of JNK signaling in mammals also support a role for JNK in tumor development. Thus, Nateri

JNK in diabetes and metabolism

Biochemical studies have established that JNK phosphorylates the insulin receptor substrate-1 (IRS-1) at the inhibitory site Ser-307 [77, 78]. JNK activation can therefore suppress signal transduction by the insulin receptor. These observations implicate the JNK signaling pathway in insulin resistance, metabolic syndrome and type 2 diabetes. Indeed, JNK is activated in obese mice [79], in part because of lipotoxic stress [80] that may be mediated by a mechanism involving the ataxia

JNK in lifespan

Aging of a eukaryotic organism is affected by its nutritional state and by its ability to repair oxidative damage. Consequently, signal transduction systems that control metabolism and oxidative stress responses influence lifespan. Two recent studies have shown that JNK can control lifespan in Drosophila and C. elegans by promoting phosphorylation of the forkhead protein FOXO [63•, 89•]. Oh and colleagues show that JNK promotes daf-16 (FOXO) activity, which regulates life span and stress

Conclusions

Significant progress towards understanding the function of the JNK signaling pathway has been achieved during the past few years. The determination of atomic structures for components of the JNK signaling pathway and also some complexes formed by these components represents a critical step towards a more complete understanding. Recent studies using the chemical genetic approach [90] to define the function of JNK in vivo using mice with a germ-line point mutation that confers sensitivity to a

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (96)

  • A. Jaeschke et al.

    An essential role of the JIP1 scaffold protein for JNK activation in adipose tissue

    Genes Dev

    (2004)
  • A. Lin

    A five-year itch in TNF-α cytotoxicity: the time factor determines JNK action

    Dev Cell

    (2006)
  • J.J. Ventura et al.

    Chemical genetic analysis of the time course of signal transduction by JNK

    Mol Cell

    (2006)
  • C. Lu et al.

    Cell apoptosis: requirement of H2AX in DNA ladder formation, but not for the activation of caspase-3

    Mol Cell

    (2006)
  • H.K. Sluss et al.

    H2AX is a target of the JNK signaling pathway that is required for apoptotic DNA fragmentation

    Mol Cell

    (2006)
  • F. Adhami et al.

    Cerebral ischemia-hypoxia induces intravascular coagulation and autophagy

    Am J Pathol

    (2006)
  • C.Y. Kuan et al.

    A critical role of neural-specific JNK3 for ischemic apoptosis

    Proc Natl Acad Sci USA

    (2003)
  • C. Tournier et al.

    Requirement of JNK for stress-induced activation of the cytochrome c- mediated death pathway

    Science

    (2000)
  • B.J. Kim et al.

    JNK- and p38 kinase-mediated phosphorylation of Bax leads to its activation and mitochondrial translocation and to apoptosis of human hepatoma HepG2 cells

    J Biol Chem

    (2006)
  • M. Nomura et al.

    14-3-3 Interacts directly with and negatively regulates pro-apoptotic Bax

    J Biol Chem

    (2003)
  • F. Tsuruta et al.

    JNK promotes Bax translocation to mitochondria through phosphorylation of 14-3-3 proteins

    EMBO J

    (2004)
  • J. Sunayama et al.

    JNK antagonizes Akt-mediated survival signals by phosphorylating 14-3-3

    J Cell Biol

    (2005)
  • C. Yu et al.

    JNK suppresses apoptosis via phosphorylation of the proapoptotic Bcl-2 family protein BAD

    Mol Cell

    (2004)
  • J. Whitfield et al.

    Dominant-negative c-Jun promotes neuronal survival by reducing BIM expression and inhibiting mitochondrial cytochrome c release

    Neuron

    (2001)
  • G.V. Putcha et al.

    Induction of BIM, a proapoptotic BH3-only BCL-2 family member, is critical for neuronal apoptosis

    Neuron

    (2001)
  • M. Uhlirova et al.

    Non-cell-autonomous induction of tissue overgrowth by JNK/Ras cooperation in a Drosophila tumor model

    Proc Natl Acad Sci USA

    (2005)
  • A.S. Nateri et al.

    Interaction of phosphorylated c-Jun with TCF4 regulates intestinal cancer development

    Nature

    (2005)
  • T. Sakurai et al.

    Loss of hepatic NF-κ B activity enhances chemical hepatocarcinogenesis through sustained c-Jun N-terminal kinase 1 activation

    Proc Natl Acad Sci USA

    (2006)
  • Whitmarsh AJ, Davis RJ: Role of mitogen-activated protein kinase kinase 4 in cancer. Oncogene 2007, in...
  • T.S. Khatlani et al.

    c-Jun N-terminal kinase is activated in non-small-cell lung cancer and promotes neoplastic transformation in human bronchial epithelial cells

    Oncogene

    (2006)
  • A.M. Stark et al.

    Reduced metastasis-suppressor gene mRNA-expression in breast cancer brain metastases

    J Cancer Res Clin Oncol

    (2005)
  • D.J. Vander Griend et al.

    Suppression of metastatic colonization by the context-dependent activation of the c-Jun NH2-terminal kinase kinases JNKK1/MKK4 and MKK7

    Cancer Res

    (2005)
  • Q.H. Guan et al.

    Neuroprotection against ischemic brain injury by a small peptide inhibitor of c-Jun N-terminal kinase (JNK) via nuclear and non-nuclear pathways

    Neuroscience

    (2006)
  • L. Hirt et al.

    D-JNKI1, a cell-penetrating c-Jun-N-terminal kinase inhibitor, protects against cell death in severe cerebral ischemia

    Stroke

    (2004)
  • D.K. Morrison et al.

    Regulation of MAP kinase signaling modules by scaffold proteins in mammals

    Annu Rev Cell Dev Biol

    (2003)
  • J.H. Shim et al.

    TAK1, but not TAB1 or TAB2, plays an essential role in multiple signaling pathways in vivo

    Genes Dev

    (2005)
  • Y.Y. Wan et al.

    The kinase TAK1 integrates antigen and cytokine receptor signaling for T cell development, survival and function

    Nat Immunol

    (2006)
  • S. Sato et al.

    Essential function for the kinase TAK1 in innate and adaptive immune responses

    Nat Immunol

    (2005)
  • J. Qin et al.

    TLR8-mediated NF-kappaB and JNK activation are TAK1-independent and MEKK3-dependent

    J Biol Chem

    (2006)
  • S. Das et al.

    Tpl2/cot signals activate ERK, JNK, and NF-kappaB in a cell-type and stimulus-specific manner

    J Biol Chem

    (2005)
  • D. Brancho et al.

    Role of MLK3 in the regulation of mitogen-activated protein kinase signaling cascades

    Mol Cell Biol

    (2005)
  • Q. Zhao et al.

    MAP kinase phosphatase 1 controls innate immune responses and suppresses endotoxic shock

    J Exp Med

    (2006)
  • H. Kamata et al.

    Reactive oxygen species promote TNFalpha-induced death and sustained JNK activation by inhibiting MAP kinase phosphatases

    Cell

    (2005)
  • M. Hamdi et al.

    DNA damage in transcribed genes induces apoptosis via the JNK pathway and the JNK-phosphatase MKP-1

    Oncogene

    (2005)
  • D.G. Jeong et al.

    Crystal structure of the catalytic domain of human MAP kinase phosphatase 5: structural insight into constitutively active phosphatase

    J Mol Biol

    (2006)
  • E.A. Willoughby et al.

    Dynamic interaction between the dual specificity phosphatase MKP7 and the JNK3 scaffold protein β-arrestin 2

    J Biol Chem

    (2005)
  • E.A. Willoughby et al.

    The JNK-interacting protein-1 scaffold protein targets MAPK phosphatase-7 to dephosphorylate JNK

    J Biol Chem

    (2003)
  • D. Horiuchi et al.

    APLIP1, a kinesin binding JIP-1/JNK scaffold protein, influences the axonal transport of both vesicles and mitochondria in Drosophila

    Curr Biol

    (2005)
  • Cited by (0)

    View full text