Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 204(4): 607-23, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24535827

ABSTRACT

TNF activates three distinct intracellular signaling cascades leading to cell survival, caspase-8-mediated apoptosis, or receptor interacting protein kinase 3 (RIPK3)-dependent necrosis, also called necroptosis. Depending on the cellular context, one of these pathways is activated upon TNF challenge. When caspase-8 is activated, it drives the apoptosis cascade and blocks RIPK3-dependent necrosis. Here we report the biological event switching to activate necrosis over apoptosis. TAK1 kinase is normally transiently activated upon TNF stimulation. We found that prolonged and hyperactivation of TAK1 induced phosphorylation and activation of RIPK3, leading to necrosis without caspase activation. In addition, we also demonstrated that activation of RIPK1 and RIPK3 promoted TAK1 activation, suggesting a positive feedforward loop of RIPK1, RIPK3, and TAK1. Conversely, ablation of TAK1 caused caspase-dependent apoptosis, in which Ripk3 deletion did not block cell death either in vivo or in vitro. Our results reveal that TAK1 activation drives RIPK3-dependent necrosis and inhibits apoptosis. TAK1 acts as a switch between apoptosis and necrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/drug effects , MAP Kinase Kinase Kinases/physiology , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Blotting, Western , Cell Cycle , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Flow Cytometry , Humans , Immunoprecipitation , Integrases/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Phosphorylation , RNA, Small Interfering/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction
2.
Proc Natl Acad Sci U S A ; 109(9): 3365-70, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331902

ABSTRACT

Dysregulation in cellular redox systems results in accumulation of reactive oxygen species (ROS), which are causally associated with a number of disease conditions. Transforming growth factor ß-activated kinase 1 (TAK1) is a signaling intermediate of innate immune signaling pathways and is critically involved in the redox regulation in vivo. Ablation of TAK1 causes accumulation of ROS, resulting in epithelial cell death and inflammation. Here we determine the mechanism by which TAK1 kinase is activated in epithelial tissues. TAB1 and TAB2 are structurally unrelated TAK1 binding protein partners. TAB2 is known to mediate polyubiquitin chain-dependent TAK1 activation in innate immune signaling pathways, whereas the role of TAB1 is not defined. We found that epithelial-specific TAB1 and TAB2 double- but not TAB1 or TAB2 single-knockout mice phenocopied epithelial-specific TAK1 knockout mice. We demonstrate that phosphorylation-dependent basal activity of TAK1 is dependent on TAB1. Ablation of both TAB1 and TAB2 diminished the activity of TAK1 in vivo and causes accumulation of ROS in the epithelial tissues. These results demonstrate that epithelial TAK1 activity is regulated through two unique, TAB1-dependent basal and TAB2-mediated stimuli-dependent mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Epithelial Cells/enzymology , MAP Kinase Kinase Kinases/metabolism , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Enzyme Activation , Epidermis/enzymology , Intestinal Mucosa/enzymology , Keratinocytes/enzymology , Mice , Mice, Knockout , Oxidation-Reduction , Oxidative Stress , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction
3.
Cancer Res ; 70(21): 8417-25, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959492

ABSTRACT

TAK1 kinase activates multiple transcription factors and regulates the level of reactive oxygen species (ROS). We have previously reported that ablation of TAK1 in keratinocytes causes hypersensitivity to ROS-induced cell apoptosis. It is known that some tumor cells produce ROS at higher levels compared with normal cells. We used inducible epidermal-specific TAK1 knockout mice and examined whether ablation of TAK1 in preexisting skin tumors could cause an increase in ROS and result in tumor cell death. Deletion of tak1 gene in skin tumors caused the accumulation of ROS and increased apoptosis, and skin tumors totally regressed within 5 to 10 days. Normal skin did not exhibit any significant abnormality on tak1 gene deletion. Thus, TAK1 kinase could be a new and effective molecular target for ROS-based tumor killing.


Subject(s)
Apoptosis , Gene Deletion , MAP Kinase Kinase Kinases/physiology , Reactive Oxygen Species/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Up-Regulation , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Blotting, Western , Carcinogens/toxicity , Cell Proliferation , Integrases/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , Mice, Knockout , Papilloma/chemically induced , Papilloma/metabolism , Papilloma/pathology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Skin/pathology , Skin Neoplasms/chemically induced
4.
J Immunol ; 185(8): 4729-37, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20855879

ABSTRACT

The intestinal epithelium is constantly exposed to inducers of reactive oxygen species (ROS), such as commensal microorganisms. Levels of ROS are normally maintained at nontoxic levels, but dysregulation of ROS is involved in intestinal inflammatory diseases. In this article, we report that TGF-ß-activated kinase 1 (TAK1) is a key regulator of ROS in the intestinal epithelium. tak1 gene deletion in the mouse intestinal epithelium caused tissue damage involving enterocyte apoptosis, disruption of tight junctions, and inflammation. Disruption of TNF signaling, which is a major intestinal damage inducer, rescued the inflammatory conditions but not apoptosis or disruption of tight junctions in the TAK1-deficient intestinal epithelium, suggesting that TNF is not a primary inducer of the damage noted in TAK1-deficient intestinal epithelium. We found that TAK1 deficiency resulted in reduced expression of several antioxidant-responsive genes and reduced the protein level of a key antioxidant transcription factor NF-E2-related factor 2, which resulted in accumulation of ROS. Exogenous antioxidant treatment reduced apoptosis and disruption of tight junctions in the TAK1-deficient intestinal epithelium. Thus, TAK1 signaling regulates ROS through transcription factor NF-E2-related factor 2, which is important for intestinal epithelial integrity.


Subject(s)
Immunity, Mucosal/physiology , Intestinal Mucosa/enzymology , MAP Kinase Kinase Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Blotting, Western , Epithelium/enzymology , Epithelium/immunology , Gene Expression , Gene Expression Regulation/immunology , Immunohistochemistry , Intestinal Mucosa/immunology , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-E2-Related Factor 2/immunology , Oxidative Stress/immunology , Reactive Oxygen Species/immunology , Reverse Transcriptase Polymerase Chain Reaction
5.
PLoS One ; 4(2): e4561, 2009.
Article in English | MEDLINE | ID: mdl-19234607

ABSTRACT

BACKGROUND: We have previously reported that intestinal epithelium-specific TAK1 deleted mice exhibit severe inflammation and mortality at postnatal day 1 due to TNF-induced epithelial cell death. Although deletion of TNF receptor 1 (TNFR1) can largely rescue those neonatal phenotypes, mice harboring double deletion of TNF receptor 1 (TNFR1) and intestinal epithelium-specific deletion of TAK1 (TNFR1KO/TAK1(IE)KO) still occasionally show increased inflammation. This indicates that TAK1 is important for TNF-independent regulation of intestinal integrity. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we investigated the TNF-independent role of TAK1 in the intestinal epithelium. Because the inflammatory conditions were sporadically developed in the double mutant TNFR1KO/TAK1(IE)KO mice, we hypothesize that epithelial TAK1 signaling is important for preventing stress-induced barrier dysfunction. To test this hypothesis, the TNFR1KO/TAK1(IE)KO mice were subjected to acute colitis by administration of dextran sulfate sodium (DSS). We found that loss of TAK1 significantly augments DSS-induced experimental colitis. DSS induced weight loss, intestinal damages and inflammatory markers in TNFR1KO/TAK1(IE)KO mice at higher levels compared to the TNFR1KO control mice. Apoptosis was strongly induced and epithelial cell proliferation was decreased in the TAK1-deficient intestinal epithelium upon DSS exposure. These suggest that epithelial-derived TAK1 signaling is important for cytoprotection and repair against injury. Finally, we showed that TAK1 is essential for interleukin 1- and bacterial components-induced expression of cytoprotective factors such as interleukin 6 and cycloxygenase 2. CONCLUSIONS: Homeostatic cytokines and microbes-induced intestinal epithelial TAK1 signaling regulates cytoprotective factors and cell proliferation, which is pivotal for protecting the intestinal epithelium against injury.


Subject(s)
Colitis/etiology , Intestinal Mucosa/metabolism , MAP Kinase Kinase Kinases/physiology , Signal Transduction , Animals , Apoptosis , Cell Proliferation , Colitis/chemically induced , Cyclooxygenase 2 , Dextran Sulfate , Interleukin-6 , MAP Kinase Kinase Kinases/deficiency , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/deficiency
6.
J Biol Chem ; 283(48): 33080-6, 2008 Nov 28.
Article in English | MEDLINE | ID: mdl-18829460

ABSTRACT

TAK1 kinase is an indispensable intermediate in several cytokine signaling pathways including tumor necrosis factor, interleukin-1, and transforming growth factor-beta signaling pathways. TAK1 also participates in stress-activated intracellular signaling pathways such as osmotic stress signaling pathway. TAK1-binding protein 1 (TAB1) is constitutively associated with TAK1 through its C-terminal region. Although TAB1 is known to augment TAK1 catalytic activity when it is overexpressed, the role of TAB1 under physiological conditions has not yet been identified. In this study, we determined the role of TAB1 in TAK1 signaling by analyzing TAB1-deficient mouse embryonic fibroblasts (MEFs). Tumor necrosis factor- and interleukin-1-induced activation of TAK1 was entirely normal in Tab1-deficient MEFs and could activate both mitogen-activated protein kinases and NF-kappaB. In contrast, we found that osmotic stress-induced activation of TAK1 was largely impaired in Tab1-deficient MEFs. Furthermore, we showed that the C-terminal 68 amino acids of TAB1 were sufficient to mediate osmotic stress-induced TAK1 activation. Finally, we attempted to determine the mechanism by which TAB1 activates TAK1. We found that TAK1 is spontaneously activated when the concentration is increased and that it is totally dependent on TAB1. Cell shrinkage under the osmotic stress condition increases the concentration of TAB1-TAK1 and may oligomerize and activate TAK1 in a TAB1-dependent manner. These results demonstrate that TAB1 mediates TAK1 activation only in a subset of TAK1 pathways that are mediated through spontaneous oligomerization of TAB1-TAK1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytokines/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , MAP Kinase Kinase Kinases/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Embryo, Mammalian/cytology , Enzyme Activation/physiology , Fibroblasts/cytology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Osmotic Pressure/physiology , Protein Structure, Tertiary/physiology
7.
J Biol Chem ; 283(38): 26161-8, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18606807

ABSTRACT

Mice with a keratinocyte-specific deletion of Tak1 exhibit severe skin inflammation due to hypersensitivity to tumor necrosis factor (TNF) killing. Here we have examined the mechanisms underlying this hypersensitivity. We found that TAK1 deficiency up-regulates reactive oxygen species (ROS) resulting in cell death upon TNF or oxidative stress challenge. Because blockade of NF-kappaB did not increase ROS or did not sensitize cells to oxidative stress in keratinocytes TAK1 regulates ROS mainly through the mechanisms other than those mediated by NF-kappaB. We found that c-Jun was decreased in TAK1-deficient keratinocytes and that ectopic expression of c-Jun could partially inhibit TNF-induced increase of ROS and cell death. Finally, we show that, in an in vivo setting, the antioxidant treatment could reduce an inflammatory condition in keratinocyte-specific Tak1 deletion mice. Thus, TAK1 regulates ROS partially through c-Jun, which is important for preventing ROS-induced skin inflammation.


Subject(s)
Gene Expression Regulation , Keratinocytes/metabolism , MAP Kinase Kinase Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species , Skin/metabolism , Animals , Antioxidants/metabolism , Cytochromes c/metabolism , Inflammation , Keratinocytes/cytology , Mice , Mice, Transgenic , Models, Biological , NF-kappa B/metabolism , Oxidative Stress
8.
J Biol Chem ; 283(1): 137-144, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-17965022

ABSTRACT

Muramyl dipeptide (MDP) is a peptidoglycan moiety derived from commensal and pathogenic bacteria, and a ligand of its intracellular sensor NOD2. Mutations in NOD2 are highly associated with Crohn disease, which is characterized by dysregulated inflammation in the intestine. However, the mechanism linking abnormality of NOD2 signaling and inflammation has yet to be elucidated. Here we show that transforming growth factor beta-activated kinase 1 (TAK1) is an essential intermediate of NOD2 signaling. We found that TAK1 deletion completely abolished MDP-NOD2 signaling, activation of NF-kappaB and MAPKs, and subsequent induction of cytokines/chemokines in keratinocytes. NOD2 and its downstream effector RICK associated with and activated TAK1. TAK1 deficiency also abolished MDP-induced NOD2 expression. Because mice with epidermis-specific deletion of TAK1 develop severe inflammatory conditions, we propose that TAK1 and NOD2 signaling are important for maintaining normal homeostasis of the skin, and its ablation may impair the skin barrier function leading to inflammation.


Subject(s)
Keratinocytes/metabolism , MAP Kinase Kinase Kinases/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Electrophoretic Mobility Shift Assay , Humans , Immunity, Innate/drug effects , Immunoblotting , Immunoprecipitation , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Keratinocytes/cytology , Keratinocytes/drug effects , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Mutant Strains , Models, Biological , NF-kappa B/genetics , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Biol Chem ; 282(13): 9475-9481, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17276978

ABSTRACT

Transforming growth factor-beta (TGF-beta) regulates a variety of physiologic processes through essential intracellular mediators Smads. The SnoN oncoprotein is an inhibitor of TGF-beta signaling. SnoN recruits transcriptional repressor complex to block Smad-dependent transcriptional activation of TGF-beta-responsive genes. Following TGF-beta stimulation, SnoN is rapidly degraded, thereby allowing the activation of TGF-beta target genes. Here, we report the role of TAK1 as a SnoN protein kinase. TAK1 interacted with and phosphorylated SnoN, and this phosphorylation regulated the stability of SnoN. Inactivation of TAK1 prevented TGF-beta-induced SnoN degradation and impaired induction of the TGF-beta-responsive genes. These data suggest that TAK1 modulates TGF-beta-dependent cellular responses by targeting SnoN for degradation.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/physiology , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Cell Line, Transformed , HeLa Cells , Humans , Phosphorylation
10.
J Biol Chem ; 281(39): 28802-10, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16893890

ABSTRACT

Osmotic stress activates MAPKs, including JNK and p38, which play important roles in cellular stress responses. Transforming growth factor-beta-activated kinase 1 (TAK1) is a member of the MAPK kinase kinase (MAPKKK) family and can activate JNK and p38. TAK1 can also activate IkappaB kinase (IKK) that leads to degradation of IkappaB and subsequent NF-kappaB activation. We found that TAK1 is essential for osmotic stress-induced activation of JNK but is not an exclusive mediator of p38 activation. Furthermore, we found that although TAK1 was highly activated upon osmotic stress, it could not induce degradation of IkappaB or activation of NF-kappaB. These results suggest that TAK1 activity is somehow modulated to function specifically in osmotic stress signaling, leading to the activation of JNK but not of IKK. To elucidate the mechanism underlying this modulation, we screened for potential TAK1-binding proteins. We found that TAO2 (thousand-and-one amino acid kinase 2) associates with TAK1 and can inhibit TAK1-mediated activation of NF-kappaB but not of JNK. We observed that TAO2 can interfere with the interaction between TAK1 and IKK and thus may regulate TAK1 function. TAK1 is activated by many distinct stimuli, including cytokines and stresses, and regulation by TAO2 may be important to activate specific intracellular signaling pathways that are unique to osmotic stress.


Subject(s)
Gene Expression Regulation, Enzymologic , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Osmosis , Protein Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Cell Line , Enzyme Activation , Humans , Mice , Protein Binding , Protein Kinases/chemistry , RNA, Small Interfering/metabolism
11.
J Biol Chem ; 281(28): 19610-7, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16675448

ABSTRACT

Transforming growth factor beta-activated kinase 1 (TAK1) functions downstream of inflammatory cytokines to activate c-Jun N-terminal kinase (JNK) as well as NF-kappaB in several cell types. However, the functional role of TAK1 in an in vivo setting has not been determined. Here we have demonstrated that TAK1 is the major regulator of skin inflammation as well as keratinocyte death in vivo. Epidermal-specific deletion of TAK1 causes a severe inflammatory skin condition by postnatal day 6-8. The mutant skin also exhibits massive keratinocyte death. Analysis of keratinocytes isolated from the mutant skin revealed that TAK1 deficiency results in a striking increase in apoptosis in response to tumor necrosis factor (TNF). TAK1-deficient keratinocytes cannot activate NF-kappaB or JNK upon TNF treatment. These results suggest that TNF induces TAK1-deficient keratinocyte death because of the lack of NF-kappaB (and possibly JNK)-mediated cell survival signaling. Finally, we have shown that deletion of the TNF receptor can largely rescue keratinocyte death as well as inflammatory skin condition in epidermal-specific TAK1-deficient mice. Our results demonstrate that TAK1 is a master regulator of TNF signaling in skin and regulates skin inflammation and keratinocyte death.


Subject(s)
Apoptosis , Epidermis/metabolism , Inflammation , MAP Kinase Kinase Kinases/physiology , Skin/pathology , Animals , Cytokines/metabolism , Gene Deletion , Homeostasis , Keratinocytes/cytology , Keratinocytes/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...