scispace - formally typeset
Search or ask a question

Showing papers by "Michael Karin published in 1999"


Journal ArticleDOI
22 Nov 1999-Oncogene
TL;DR: This review describes the identification of proteins in the IKK complex, and the regulation and physiological functions of IKK.
Abstract: Rel/NF-kappaB transcription factors are primarily regulated by association with inhibitor IkappaB proteins. Thus, in most cells NF-kappaB exists in the cytoplasm in an inactive complex bound to IkappaB. Most agents that activate NF-kappaB do so through a common pathway based on phosphorylation-induced, proteasome-mediated degradation of IkappaB. The key regulatory step in this pathway involves activation of a high molecular weight IkappaB kinase (IKK) complex, whose catalysis is generally carried out by a heterodimeric kinase consisting of IKKalpha and IKKbeta subunits. This review describes the identification of proteins in the IKK complex, and the regulation and physiological functions of IKK.

1,111 citations


Journal ArticleDOI
TL;DR: IKKβ-deficient mice are defective in activation of IKK and NF-κB in response to either tumor necrosis factor α or interleukin 1, and IKKα is unresponsive to IKK activators.
Abstract: The IκB kinase (IKK) complex is composed of three subunits, IKKα, IKKβ, and IKKγ (NEMO). While IKKα and IKKβ are highly similar catalytic subunits, both capable of IκB phosphorylation in vitro, IKKγ is a regulatory subunit. Previous biochemical and genetic analyses have indicated that despite their similar structures and in vitro kinase activities, IKKα and IKKβ have distinct functions. Surprisingly, disruption of the Ikkα locus did not abolish activation of IKK by proinflammatory stimuli and resulted in only a small decrease in nuclear factor (NF)-κB activation. Now we describe the pathophysiological consequence of disruption of the Ikkβ locus. IKKβ-deficient mice die at mid-gestation from uncontrolled liver apoptosis, a phenotype that is remarkably similar to that of mice deficient in both the RelA (p65) and NF-κB1 (p50/p105) subunits of NF-κB. Accordingly, IKKβ-deficient cells are defective in activation of IKK and NF-κB in response to either tumor necrosis factor α or interleukin 1. Thus IKKβ, but not IKKα, plays the major role in IKK activation and induction of NF-κB activity. In the absence of IKKβ, IKKα is unresponsive to IKK activators.

918 citations


Journal ArticleDOI
09 Apr 1999-Science
TL;DR: IkappaB [inhibitor of nuclear factor kappaB] kinase (IKK) phosphorylates IkappaB inhibitory proteins, causing their degradation and activation of transcription factor NF-, a master activator of inflammatory responses.
Abstract: IkappaB [inhibitor of nuclear factor kappaB (NF-kappaB)] kinase (IKK) phosphorylates IkappaB inhibitory proteins, causing their degradation and activation of transcription factor NF-kappaB, a master activator of inflammatory responses. IKK is composed of three subunits-IKKalpha and IKKbeta, which are highly similar protein kinases, and IKKgamma, a regulatory subunit. In mammalian cells, phosphorylation of two sites at the activation loop of IKKbeta was essential for activation of IKK by tumor necrosis factor and interleukin-1. Elimination of equivalent sites in IKKalpha, however, did not interfere with IKK activation. Thus, IKKbeta, not IKKalpha, is the target for proinflammatory stimuli. Once activated, IKKbeta autophosphorylated at a carboxyl-terminal serine cluster. Such phosphorylation decreased IKK activity and may prevent prolonged activation of the inflammatory response.

837 citations


Journal ArticleDOI
09 Apr 1999-Science
TL;DR: Loss of IKKα interfered with multiple morphogenetic events, including limb and skeletal patterning and proliferation and differentiation of epidermal keratinocytes, and phosphorylation of the IκB inhibitors in response to proinflammatory stimuli.
Abstract: The oligomeric IκB kinase (IKK) is composed of three polypeptides: IKKα and IKKβ, the catalytic subunits, and IKKγ, a regulatory subunit. IKKα and IKKβ are similar in structure and thought to have similar function—phosphorylation of the IκB inhibitors in response to proinflammatory stimuli. Such phosphorylation leads to degradation of IκB and activation of nuclear factor κB transcription factors. The physiological function of these protein kinases was explored by analysis of IKKα-deficient mice. IKKα was not required for activation of IKK and degradation of IκB by proinflammatory stimuli. Instead, loss of IKKα interfered with multiple morphogenetic events, including limb and skeletal patterning and proliferation and differentiation of epidermal keratinocytes.

801 citations


Journal ArticleDOI
TL;DR: Recent studies have advanced the understanding of the signal transduction pathway leading to NF‐κB activation by cytokines and will provide insights for the mechanism by which NF-κB is regulated by oxidative stress.
Abstract: NF-kappaB is a dimeric transcription factor that is involved in the regulation of a large number of genes that control various aspects of the immune and inflammatory response. It is activated by a variety of stimuli ranging from cytokines, to various forms of radiation, to oxidative stress (such as exposure to H2O2). Recent studies have advanced our understanding of the signal transduction pathway leading to NF-kappaB activation by cytokines and will provide insights for the mechanism by which NF-kappaB is regulated by oxidative stress. An important question that is yet to be answered is whether reactive oxygen species play a physiological role in NF-kappaB activation.

796 citations


Journal ArticleDOI
TL;DR: Three-dimensional structures of NF-kBzIkB ternary complexes were solved, and fascinating structures indicate that the ankyrin repeats of IkBa form a slightly bent cylinder through a stacked arrangement of a-helices that compose their ankyrIn repeats.

643 citations


Journal ArticleDOI
TL;DR: It is demonstrated that fibroblasts derived from c-jun-/- mouse fetuses exhibit a severe proliferation defect and undergo a prolonged crisis before spontaneous immortalization, and a mechanistic link between c-Jun-dependent mitogenic signaling and cell-cycle regulation is established.
Abstract: The c-jun proto-oncogene encodes a component of the mitogen-inducible immediate-early transcription factor AP-1 and has been implicated as a positive regulator of cell proliferation and G1-to-S-phase progression. Here we report that fibroblasts derived from c-jun-/- mouse fetuses exhibit a severe proliferation defect and undergo a prolonged crisis before spontaneous immortalization. The cyclin D1- and cyclin E-dependent kinases (CDKs) and transcription factor E2F are poorly activated, resulting in inefficient G1-to-S-phase progression. Furthermore, the absence of c-Jun results in elevated expression of the tumor suppressor gene p53 and its target gene, the CDK inhibitor p21, whereas overexpression of c-Jun represses p53 and p21 expression and accelerates cell proliferation. Surprisingly, protein stabilization, the common mechanism of p53 regulation, is not involved in up-regulation of p53 in c-jun-/- fibroblasts. Rather, c-Jun regulates transcription of p53 negatively by direct binding to a variant AP-1 site in the p53 promoter. Importantly, deletion of p53 abrogates all defects of cells lacking c-Jun in cell cycle progression, proliferation, immortalization, and activation of G1 CDKs and E2F. These results demonstrate that an essential, rate-limiting function of c-Jun in fibroblast proliferation is negative regulation of p53 expression, and establish a mechanistic link between c-Jun-dependent mitogenic signaling and cell-cycle regulation.

561 citations


Journal ArticleDOI
TL;DR: Exposure of cells to various extracellular stimuli leads to the rapid phosphorylation, ubiquitinylation, and ultimately proteolytic degradation of IκB, which frees NF-κB to translocate to the nucleus, where it regulates gene transcription.
Abstract: Nuclear factor kappa B (NF-kappaB)/Rel proteins are dimeric, sequence-specific transcription factors involved in the activation of an exceptionally large number of genes in response to inflammation, viral and bacterial infections, and other stressful situations requiring rapid reprogramming of gene expression. In unstimulated cells, NF-kappaB is sequestered in an inactive form in the cytoplasm bound to inhibitory IkappaB proteins. Stimulation leads to the rapid phosphorylation, ubiquitinylation, and ultimately proteolytic degradation of IkappaB, which frees NF-kappaB to translocate to the nucleus and activate the transcription of its target genes. The multisubunit IkappaB kinase (IKK) responsible for the inducible phosphorylation of IkappaB appears to be the initial point of convergence for most stimuli that activate NF-kappaB. IKK contains two catalytic subunits, IKKalpha and IKKbeta, both of which phosphorylate IkappaB at sites phosphorylated in vivo. Gene knockout studies indicate that IKKbeta is primarily responsible for the activation of NF-kappaB in response to proinflammatory stimuli, whereas IKKalpha is essential for keratinocyte differentiation. The activity of IKK is regulated by phosphorylation. IKK contains a regulatory subunit, IKKgamma, which is critical for activation of IKK and is postulated to serve as a recognition site for upstream activators. When phosphorylated, the IKK recognition site on IkappaBalpha serves as a specific recognition site for the kappa-TrCP-like component of a Skp1-Cullin-F-box-type E3 ubiquitin-protein ligase. A variety of other signaling events, including phosphorylation of NF-kappaB, phosphorylation of IKK, new synthesis of IkappaBs, and the processing of NF-kappaB precursors provide mechanisms of modulating the amount and duration of NF-kappaB activity.

508 citations


Journal ArticleDOI
TL;DR: Oligomerization of the TRAF2 effector domain results in specific binding to MEKK1, a protein kinase capable of JNK, p38, and IKK activation, and induction of TNF-alpha and IL-1 responsive genes.
Abstract: Interleukin-1 (IL-1) and tumor necrosis factor (TNF-alpha) stimulate transcription factors AP-1 and NF-kappaB through activation of the MAP kinases JNK and p38 and the IkappaB kinase (IKK), respectively. The TNF-alpha and IL-1 signals are transduced through TRAF2 and TRAF6, respectively. Overexpressed TRAF2 or TRAF6 activate JNK, p38, or IKK in the absence of extracellular stimulation. By replacing the carboxy-terminal TRAF domain of TRAF2 and TRAF6 with repeats of the immunophilin FKBP12, we demonstrate that their effector domains are composed of their amino-terminal Zn and RING fingers. Oligomerization of the TRAF2 effector domain results in specific binding to MEKK1, a protein kinase capable of JNK, p38, and IKK activation, and induction of TNF-alpha and IL-1 responsive genes. TNF-alpha also enhances the binding of native TRAF2 to MEKK1 and stimulates the kinase activity of the latter. Thus, TNF-alpha and IL-1 signaling is based on oligomerization of TRAF2 and TRAF6 leading to activation of effector kinases.

487 citations


Journal ArticleDOI
TL;DR: The JNK- to-c-Jun-to-FasL pathway is an important mediator of stress-induced neuronal apoptosis and is used to understand how JNK activation leads to apoptosis.
Abstract: The JNK pathway modulates AP-1 activity. While in some cells it may have proliferative and protective roles, in neuronal cells it is involved in apoptosis in response to stress or withdrawal of survival signals. To understand how JNK activation leads to apoptosis, we used PC12 cells and primary neuronal cultures. In PC12 cells, deliberate JNK activation is followed by induction of Fas ligand (FasL) expression and apoptosis. JNK activation detected by c-Jun phosphorylation and FasL induction are also observed after removal of either nerve growth factor from differentiated PC12 cells or KCl from primary cerebellar granule neurons (CGCs). Sequestation of FasL by incubation with a Fas-Fc decoy inhibits apoptosis in all three cases. CGCs derived from gld mice (defective in FasL) are less sensitive to apoptosis caused by KCl removal than wild-type neurons. In PC12 cells, protection is also conferred by a c-Jun mutant lacking JNK phosphoacceptor sites and a small molecule inhibitor of p38 mitogen-activated protein kinase and JNK, which inhibits FasL induction. Hence, the JNK-to-c-Jun-to-FasL pathway is an important mediator of stress-induced neuronal apoptosis.

485 citations


Journal ArticleDOI
01 Dec 1999-Immunity
TL;DR: A general role is established for IKKbeta and JNK2 are essential for efficient induction of type I IFN and other cytokines in response to viral infection or dsRNA and a pathway leading to NF-kappaB is described.

Journal ArticleDOI
TL;DR: These results assign both pro- and anti-apoptotic functions for JNK1 and JNK2 in the development of the fetal brain.

Journal ArticleDOI
TL;DR: The discovery of IKK is summarized and recent knowledge about its composition, regulation, and physiological functions are summarized, indicating that the physiological IκB kinase had to fulfill several criteria.
Abstract: Protein kinases that regulate the activity of specific transcription factors in response to extracellular stimuli not only are the subject of intense research but also are being chased as potential targets for development of new drugs for treatment of various human diseases. One such protein kinase is IKK, the IκB kinase that activates nuclear factor κB (NF-κB) through phosphorylation of IκB inhibitory proteins. In this review, we summarize the discovery of IKK and recent knowledge about its composition, regulation, and physiological functions. NF-κB transcription factors regulate the expression of a large number of genes that are necessary for proper functioning of the immune system and are key mediators of inflammatory responses to pathogens (2, 4, 5). NF-κB is also associated with cellular transformation and oncogenesis, and one of its most important, but lately discovered functions, is the activation of an antiapoptotic gene expression program (6, 29, 36, 38, 39, 42). As a transcription factor that orchestrates the inflammatory response, NF-κB is rapidly activated, independently of new protein synthesis, in response to signals produced during infection (e.g., bacterial endotoxins and viral double-stranded RNA) (for a review, see reference 3). NF-κB activation is also a transient response; this is of importance because many of the genes that are activated by NF-κB encode potentially toxic products such as tumor necrosis factor (TNF). The key to NF-κB regulation is the inhibitory κB (IκB) proteins which retain NF-κB in the cytoplasm (reviewed in reference 37). In response to diverse stimuli, IκBs are rapidly degraded and the freed NF-κB dimers translocate to the nucleus. Several years ago, it was established that the critical event which triggers the polyubiquitination and degradation of IκBs via the 26S proteasome is their stimulus-dependent phosphorylation at two serine residues (residues 32 and 36 in IκBα) that are located within their conserved N-terminal regulatory region (1, 10–12, 18, 20, 34, 40). The protein kinase that phosphorylates these regulatory sites remained elusive, and without detailed knowledge about its molecular identity, there was little progress towards a full understanding of the signaling pathways that control NF-κB activity. The initial hunt for such a protein kinase yielded many false candidates, such as protein kinase C, casein kinase II, and ribosomal S6 kinase (pp90rsk) (reviewed in reference 43). Although most of these kinases phosphorylate IκB proteins in the test tube on different serine, threonine, or tyrosine residues, none of them was found to phosphorylate the two regulatory sites that trigger the degradation of IκBs in a stimulus-dependent manner. A large-molecular-mass (700-kDa) protein kinase activity that phosphorylates IκBα on S32 and S36 in a ubiquitin-dependent manner was also detected in extracts of nonstimulated HeLa cells (13, 25). However, this activity was not reported to be stimulus dependent, and to date, its components and molecular identity are unknown. A careful consideration of IκB phosphorylation indicated that the physiological IκB kinase had to fulfill several criteria. Its activity should be stimulated by inducers of NF-κB with kinetics that are consistent with those of NF-κB activation, and it should phosphorylate both S32 and S36 in the N terminus of IκBα and both S19 and S23 in the N terminus of IκBβ. In addition, since substitution of threonines for these serines results in IκB mutants that are resistant to degradation, the physiological IκB kinase should be serine specific (18).

Journal ArticleDOI
TL;DR: JNK2 functions in a cell-type-specific and stimulus-dependent manner, being required for apoptosis of immature thymocytes induced by anti-CD3 antibody but not for apoptotic induced byAnti-Fas antibody, UVC or dexamethasone, and is not required for activation-induced cell death of mature T cells.

Journal ArticleDOI
TL;DR: Results demonstrated important roles for NF-κB in both inducible cytokine expression and T cell proliferation after TCR engagement and α-CD3–mediated apoptosis of DP thymocytes through a pathway that involves the regulation of the antiapoptotic gene, bcl-xL.
Abstract: To examine the role of nuclear factor (NF)-kappaB in T cell development and activation in vivo, we produced transgenic mice that express a superinhibitory mutant form of inhibitor kappaB-alpha (IkappaB-alphaA32/36) under the control of the T cell-specific CD2 promoter and enhancer (mutant [m]IkappaB-alpha mice). Thymocyte development proceeded normally in the mIkappaB-alpha mice. However, the numbers of peripheral CD8(+) T cells were significantly reduced in these animals. The mIkappaB-alpha thymocytes displayed a marked proliferative defect and significant reductions in interleukin (IL)-2, IL-3, and granulocyte/macrophage colony-stimulating factor production after cross-linking of the T cell antigen receptor. Perhaps more unexpectedly, double positive (CD4(+)CD8(+); DP) thymocytes from the mIkappaB-alpha mice were resistant to alpha-CD3-mediated apoptosis in vivo. In contrast, they remained sensitive to apoptosis induced by gamma-irradiation. Apoptosis of wild-type DP thymocytes after in vivo administration of alpha-CD3 mAb was preceded by a significant reduction in the level of expression of the antiapoptotic gene, bcl-xL. In contrast, the DP mIkappaB-alpha thymocytes maintained high level expression of bcl-xL after alpha-CD3 treatment. Taken together, these results demonstrated important roles for NF-kappaB in both inducible cytokine expression and T cell proliferation after TCR engagement. In addition, NF-kappaB is required for the alpha-CD3-mediated apoptosis of DP thymocytes through a pathway that involves the regulation of the antiapoptotic gene, bcl-xL.

Journal ArticleDOI
TL;DR: Calcineurin was found to specifically down-regulate the transcriptional activity of transcription factor Elk1, following stimulation of this activity by the ERK, Jun N-terminal kinase, or p38 MAP kinase pathways.

Journal ArticleDOI
TL;DR: It is found that JNK activation by UV irradiation and anisomycin treatment precedes the induction of gadd45mRNA by these agents, and stress-induced JNK does not depend on Gadd45 induction, and stresses such as γ irradiation induce Gadd 45 and do not activate JNK in mouse fibroblasts.


Patent
20 Aug 1999
TL;DR: In this paper, a regulatory subunit of the IκB kinase (IKK) complex, IKK-γ, has been identified, which has substantially the same amino acid sequence as SEQ ID NO: 2 shown in FIG. 2.
Abstract: The present invention provides a novel essential regulatory subunit of the IκB kinase (IKK) complex, IKK-γ. The isolated IKK-γ subunit of the invention has substantially the same amino acid sequence as SEQ ID NO: 2 shown in FIG. 2.