Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Cell Sci ; 136(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36912451

ABSTRACT

Neuroinflammation is causally associated with Alzheimer's disease (AD) pathology. Reactive glia cells secrete various neurotoxic factors that impair neuronal homeostasis eventually leading to neuronal loss. Although the glial activation mechanism in AD has been relatively well studied, how it perturbs intraneuronal signaling, which ultimately leads to neuronal cell death, remains poorly understood. Here, we report that compound stimulation with the neurotoxic factors TNF and glutamate aberrantly activates neuronal TAK1 (also known as MAP3K7), which promotes the pathogenesis of AD in mouse models. Glutamate-induced Ca2+ influx shifts TNF signaling to hyper-activate TAK1 enzymatic activity through Ca2+/calmodulin-dependent protein kinase II, which leads to necroptotic cellular damage. Genetic ablation and pharmacological inhibition of TAK1 ameliorated AD-associated neuronal loss and cognitive impairment in the AD model mice. Our findings provide a molecular mechanism linking cytokines, Ca2+ signaling and neuronal necroptosis in AD.


Subject(s)
Alzheimer Disease , Animals , Mice , Alzheimer Disease/genetics , Calcium , Cytokines/metabolism , Neuroinflammatory Diseases , Signal Transduction/physiology
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161265

ABSTRACT

Mitogen-activated protein kinase kinase kinase 7 (MAP3K7), known as TAK1, is an intracellular signaling intermediate of inflammatory responses. However, a series of mouse Tak1 gene deletion analyses have revealed that ablation of TAK1 does not prevent but rather elicits inflammation, which is accompanied by elevation of reactive oxygen species (ROS). This has been considered a consequence of impaired TAK1-dependent maintenance of tissue integrity. Contrary to this view, here we propose that TAK1 inhibition-induced ROS are an active cellular process that targets intracellular bacteria. Intracellular bacterial effector proteins such as Yersinia's outer membrane protein YopJ are known to inhibit TAK1 to circumvent the inflammatory host responses. We found that such TAK1 inhibition induces mitochondrial-derived ROS, which effectively destroys intracellular bacteria. Two cell death-signaling molecules, caspase 8 and RIPK3, cooperatively participate in TAK1 inhibition-induced ROS and blockade of intracellular bacterial growth. Our results reveal a previously unrecognized host defense mechanism, which is initiated by host recognition of pathogen-induced impairment in a host protein, TAK1, but not directly of pathogens.


Subject(s)
Bacteria/growth & development , Intracellular Space/microbiology , MAP Kinase Kinase Kinases/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Caspase 3/metabolism , Colony Count, Microbial , Hydrogen Sulfide/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Salmonella/drug effects , Salmonella/growth & development , Yersinia/drug effects
3.
J Cell Biol ; 218(6): 1994-2005, 2019 06 03.
Article in English | MEDLINE | ID: mdl-30975711

ABSTRACT

RIPK3, a key mediator of necroptosis, has been implicated in the host defense against viral infection primary in immune cells. However, gene expression analysis revealed that RIPK3 is abundantly expressed not only in immune organs but also in the gastrointestinal tract, particularly in the small intestine. We found that orally inoculated Listeria monocytogenes, a bacterial foodborne pathogen, efficiently spread and caused systemic infection in Ripk3-deficient mice while almost no dissemination was observed in wild-type mice. Listeria infection activated the RIPK3-MLKL pathway in cultured cells, which resulted in suppression of intracellular replication of Listeria Surprisingly, Listeria infection-induced phosphorylation of MLKL did not result in host cell killing. We found that MLKL directly binds to Listeria and inhibits their replication in the cytosol. Our findings have revealed a novel functional role of the RIPK3-MLKL pathway in nonimmune cell-derived host defense against Listeria invasion, which is mediated through cell death-independent mechanisms.


Subject(s)
Listeria/growth & development , Listeriosis/prevention & control , Necroptosis , Protein Kinases/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Female , Humans , Listeria/immunology , Listeria/metabolism , Listeriosis/metabolism , Listeriosis/microbiology , Listeriosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
4.
Stem Cells ; 37(6): 766-778, 2019 06.
Article in English | MEDLINE | ID: mdl-30786091

ABSTRACT

Aberrant wound healing presents as inappropriate or insufficient tissue formation. Using a model of musculoskeletal injury, we demonstrate that loss of transforming growth factor-ß activated kinase 1 (TAK1) signaling reduces inappropriate tissue formation (heterotopic ossification) through reduced cellular differentiation. Upon identifying increased proliferation with loss of TAK1 signaling, we considered a regenerative approach to address insufficient tissue production through coordinated inactivation of TAK1 to promote cellular proliferation, followed by reactivation to elicit differentiation and extracellular matrix production. Although the current regenerative medicine paradigm is centered on the effects of drug treatment ("drug on"), the impact of drug withdrawal ("drug off") implicit in these regimens is unknown. Because current TAK1 inhibitors are unable to phenocopy genetic Tak1 loss, we introduce the dual-inducible COmbinational Sequential Inversion ENgineering (COSIEN) mouse model. The COSIEN mouse model, which allows us to study the response to targeted drug treatment ("drug on") and subsequent withdrawal ("drug off") through genetic modification, was used here to inactivate and reactivate Tak1 with the purpose of augmenting tissue regeneration in a calvarial defect model. Our study reveals the importance of both the "drug on" (Cre-mediated inactivation) and "drug off" (Flp-mediated reactivation) states during regenerative therapy using a mouse model with broad utility to study targeted therapies for disease. Stem Cells 2019;37:766-778.


Subject(s)
Bone Regeneration/drug effects , Fractures, Bone/genetics , MAP Kinase Kinase Kinases/genetics , Mesenchymal Stem Cells/enzymology , Osteoblasts/enzymology , Wound Healing/genetics , Animals , Bone Regeneration/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Female , Founder Effect , Fractures, Bone/drug therapy , Fractures, Bone/enzymology , Fractures, Bone/pathology , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/deficiency , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Skull/drug effects , Skull/injuries , Skull/metabolism , Wound Healing/drug effects
5.
Genesis ; 56(3): e23093, 2018 03.
Article in English | MEDLINE | ID: mdl-29411501

ABSTRACT

BMP signaling plays a critical role in craniofacial development. Augmentation of BMPR1A signaling through neural crest-specific expression of constitutively active Bmpr1a (caBmpr1a) results in craniofacial deformities in mice. To investigate whether deletion of Tak1 may rescue the craniofacial deformities caused by enhanced Smad-dependent signaling through caBMPR1A, we generated embryos to activate transcription of caBmpr1a transgene and ablate Tak1 in neural crest derivatives at the same time. We found that deformities of the double mutant mice showed more severe than those with each single mutation, including median facial cleft and cleft palate. We found higher levels of cell death in the medial nasal and the lateral nasal processes at E10.5 in association with higher levels of p53 in the double mutant embryos. We also found higher levels of pSmad1/5/9 in the lateral nasal processes at E10.5 in the double mutant embryos. Western analyses revealed that double mutant embryos showed similar degrees of upregulation of pSmad1/5/9 with caBmpr1a or Tak1-cKO embryos while the double mutant embryos showed higher levels of phospho-p38 than caBmpr1a or Tak1-cKO embryos at E17.5, but not at E10.5. It suggested that deletion of Tak1 aggravates the craniofacial deformities of the caBmpr1a mutants by increasing p53 and phospho-p38 at different stage of embryogenesis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Cell Death/genetics , Craniofacial Abnormalities/genetics , Genetic Association Studies , Genotype , MAP Kinase Kinase Kinases/genetics , Mutation , Animals , Apoptosis/genetics , Biomarkers , Bone Morphogenetic Protein Receptors, Type I/metabolism , Craniofacial Abnormalities/diagnosis , Gestational Age , Immunohistochemistry , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Transgenic , Phenotype , Signal Transduction , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Sci Rep ; 7(1): 10695, 2017 09 06.
Article in English | MEDLINE | ID: mdl-28878387

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

7.
Sci Rep ; 7(1): 2918, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592892

ABSTRACT

Programmed cell death (PCD) occurs in several forms including apoptosis and necroptosis. Apoptosis is executed by the activation of caspases, while necroptosis is dependent on the receptor interacting protein kinase 3 (RIPK3). Precise control of cell death is crucial for tissue homeostasis. Indeed, necroptosis is triggered by caspase inhibition to ensure cell death. Here we identified a previously uncharacterized cell death pathway regulated by TAK1, which is unexpectedly provoked by inhibition of caspase activity and necroptosis cascades. Ablation of TAK1 triggers spontaneous death in macrophages. Simultaneous inhibition of caspases and RIPK3 did not completely restore cell viability. Previous studies demonstrated that loss of TAK1 in fibroblasts causes TNF-induced apoptosis and that additional inhibition of caspase leads to necroptotic cell death. However, we surprisingly found that caspase and RIPK3 inhibitions do not completely suppress cell death in Tak1-deficient cells. Mechanistically, the execution of the third cell death pathway in Tak1-deficient macrophages and fibroblasts were mediated by RIPK1-dependent rapid accumulation of reactive oxygen species (ROS). Conversely, activation of RIPK1 was sufficient to induce cell death. Therefore, loss of TAK1 elicits noncanonical cell death which is mediated by RIPK1-induced oxidative stress upon caspase and necroptosis inhibition to further ensure induction of cell death.

8.
Cell Death Dis ; 8(2): e2598, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28182011

ABSTRACT

Hematopoietic cell survival and death is critical for development of a functional immune system. Here, we report that a protein kinase, TAK1, is selectively required for resident macrophage integrity during embryogenesis. Hematopoietic lineage-specific deletion of Tak1 gene (Tak1HKO) caused accumulation of cellular debris in the thymus in perinatal mice. Although no overt alteration in thymocytes and blood myeloid populations was observed in Tak1HKO mice, we found that thymic and lung macrophages were diminished. In the in vitro setting, Tak1 deficiency caused profound disruption of lysosomes and killed bone marrow-derived macrophages (BMDMs) without any exogenous stressors. Inhibition of the lysosomal protease, cathepsin B, partially blocked Tak1-deficient BMDM death, suggesting that leakage of the lysosomal contents is in part the cause of cell death. To identify the trigger of this cell death, we examined involvement of TNF and Toll-like receptor pathways. Among them, we found that deletion of Tnfr1 partially rescued cell death. Finally, we show that Tnfr1 deletion partially restored thymic and lung macrophages in vivo. These results suggest that autocrine and potentially paracrine TNF kills Tak1-deficient macrophages during development. Our results reveal that TAK1 signaling maintains proper macrophage populations through protecting lysosomal integrity.


Subject(s)
Lysosomes/metabolism , MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , Protective Agents/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Embryonic Development/physiology , Lung/metabolism , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/physiology , Thymocytes/physiology , Thymus Gland/metabolism , Toll-Like Receptors/metabolism
9.
Antioxid Redox Signal ; 25(17): 953-964, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27245349

ABSTRACT

AIMS: Nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) is the master transcriptional regulator of antioxidant gene expression. On increased oxidative stress, an adaptor for Nrf2 degradation, Kelch-like ECH-associated protein 1 (Keap1), is directly modulated by oxidants in the cytoplasm, which results in stabilization and activation of Nrf2. Nrf2 is also constitutively active, to some extent, in the absence of exogenous oxidative stress. We have previously demonstrated that intestinal epithelium-specific TGF-ß-activated kinase 1 (TAK1) deletion downregulates the level of Nrf2 protein, resulting in an increase of reactive oxygen species (ROS) in a mouse model. We aim at determining the mechanism by which TAK1 modulates the level of Nrf2. RESULTS: We found that TAK1 upregulated serine 351 phosphorylation of an autophagic adaptor protein, p62/Sequestosome-1 (SQSTM1), which facilitates interaction between p62/SQSTM1 and Keap1 and subsequent Keap1 degradation. This, ultimately, causes increased Nrf2. Tak1 deficiency reduced the phosphorylation of p62/SQSTM1, resulting in decreased steady-state levels of Nrf2 along with increased Keap1. We also found that this regulation is independent of the canonical redox-mediated Nrf2 activation mechanism. In Tak1-deficient intestinal epithelium, a synthetic phenolic electrophile, butylated hydroxyanisole still effectively upregulated Nrf2 and reduced ROS. INNOVATION: Our results identify for the first time that TAK1 is a modulator of p62/SQSTM1-dependent Keap1 degradation and maintains the steady state-level of Nrf2. CONCLUSION: TAK1 regulates Nrf2 through modulation of Keap-p62/SQSTM1 interaction. This regulation is important for homeostatic antioxidant protection in the intestinal epithelium. Antioxid. Redox Signal. 25, 953-964.


Subject(s)
Antioxidants/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Intestinal Mucosa/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Models, Biological , NF-E2-Related Factor 2/genetics , Oxidative Stress , Protein Binding , Proteolysis , Reactive Oxygen Species/metabolism
10.
J Cell Sci ; 129(9): 1855-65, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26985063

ABSTRACT

Sustained endoplasmic reticulum (ER) stress disrupts normal cellular homeostasis and leads to the development of many types of human diseases, including metabolic disorders. TAK1 (also known as MAP3K7) is a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family and is activated by a diverse set of inflammatory stimuli. Here, we demonstrate that TAK1 regulates ER stress and metabolic signaling through modulation of lipid biogenesis. We found that deletion of Tak1 increased ER volume and facilitated ER-stress tolerance in cultured cells, which was mediated by upregulation of sterol-regulatory-element-binding protein (SREBP)-dependent lipogenesis. In the in vivo setting, central nervous system (CNS)-specific Tak1 deletion upregulated SREBP-target lipogenic genes and blocked ER stress in the hypothalamus. Furthermore, CNS-specific Tak1 deletion prevented ER-stress-induced hypothalamic leptin resistance and hyperphagic obesity under a high-fat diet (HFD). Thus, TAK1 is a crucial regulator of ER stress in vivo, which could be a target for alleviation of ER stress and its associated disease conditions.


Subject(s)
Endoplasmic Reticulum Stress , Hypothalamus/metabolism , Leptin/metabolism , MAP Kinase Kinase Kinases/metabolism , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Hyperphagia/chemically induced , Hyperphagia/genetics , Hyperphagia/metabolism , Hyperphagia/pathology , Hypothalamus/pathology , Leptin/genetics , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
11.
Dev Biol ; 398(2): 231-41, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25523394

ABSTRACT

Transforming growth factor-beta3 (TGF-ß3) plays a critical role in palatal epithelial cells by inducing palatal epithelial fusion, failure of which results in cleft palate, one of the most common birth defects in humans. Recent studies have shown that Smad-dependent and Smad-independent pathways work redundantly to transduce TGF-ß3 signaling in palatal epithelial cells. However, detailed mechanisms by which this signaling is mediated still remain to be elucidated. Here we show that TGF-ß activated kinase-1 (Tak1) and Smad4 interact genetically in palatal epithelial fusion. While simultaneous abrogation of both Tak1 and Smad4 in palatal epithelial cells resulted in characteristic defects in the anterior and posterior secondary palate, these phenotypes were less severe than those seen in the corresponding Tgfb3 mutants. Moreover, our results demonstrate that Trim33, a novel chromatin reader and regulator of TGF-ß signaling, cooperates with Smad4 during palatogenesis. Unlike the epithelium-specific Smad4 mutants, epithelium-specific Tak1:Smad4- and Trim33:Smad4-double mutants display reduced expression of Mmp13 in palatal medial edge epithelial cells, suggesting that both of these redundant mechanisms are required for appropriate TGF-ß signal transduction. Moreover, we show that inactivation of Tak1 in Trim33:Smad4 double conditional knockouts leads to the palatal phenotypes which are identical to those seen in epithelium-specific Tgfb3 mutants. To conclude, our data reveal added complexity in TGF-ß signaling during palatogenesis and demonstrate that functionally redundant pathways involving Smad4, Tak1 and Trim33 regulate palatal epithelial fusion.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Palate/embryology , Palate/metabolism , Signal Transduction , Smad4 Protein/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta3/metabolism , Animals , Apoptosis/genetics , Cell Fusion , Cell Proliferation , Crosses, Genetic , Embryo, Mammalian/metabolism , Enzyme Activation , Epithelial Cells/metabolism , Epithelium/metabolism , Female , Gene Deletion , Gene Expression Regulation, Developmental , Male , Matrix Metalloproteinase 13/metabolism , Mice, Knockout , Models, Biological , Mutation/genetics , Organ Specificity , Palate/abnormalities , Palate/enzymology
12.
PLoS One ; 9(4): e94982, 2014.
Article in English | MEDLINE | ID: mdl-24736749

ABSTRACT

Macrophages play diverse roles in tissue homeostasis and immunity, and canonically activated macrophages are critically associated with acute inflammatory responses. It is known that activated macrophages undergo cell death after transient activation in some settings, and the viability of macrophages impacts on inflammatory status. Here we report that TGFß- activated kinase (TAK1) activators, TAK1-binding protein 1 (TAB1) and TAK1-binding protein 2 (TAB2), are critical molecules in the regulation of activated macrophage survival. While deletion of Tak1 induced cell death in bone marrow derived macrophages even without activation, Tab1 or Tab2 deletion alone did not profoundly affect survival of naïve macrophages. However, in lipopolysaccharide (LPS)-activated macrophages, even single deletion of Tab1 or Tab2 resulted in macrophage death with both necrotic and apoptotic features. We show that TAB1 and TAB2 were redundantly involved in LPS-induced TAK1 activation in macrophages. These results demonstrate that TAK1 activity is the key to activated macrophage survival. Finally, in an in vivo setting, Tab1 deficiency impaired increase of peritoneal macrophages upon LPS challenge, suggesting that TAK1 complex regulation of macrophages may participate in in vivo macrophage homeostasis. Our results demonstrate that TAB1 and TAB2 are required for activated macrophages, making TAB1 and TAB2 effective targets to control inflammation by modulating macrophage survival.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Survival/genetics , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Deletion , Lipopolysaccharides/immunology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout
13.
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
14.
PLoS One ; 9(2): e88037, 2014.
Article in English | MEDLINE | ID: mdl-24498425

ABSTRACT

The liver is the first line of defense from environmental stressors in that hepatocytes respond to and metabolize them. Hence, hepatocytes can be damaged by stressors. Protection against hepatic cell damage and cell death is important for liver function and homeostasis. TAK1 (MAP3K7) is an intermediate of stressors such as bacterial moieties-induced signal transduction pathways in several cell types. Tak1 deficiency has been reported to induce spontaneous hepatocellular carcinoma. However, the regulatory mechanism of TAK1 activity in liver stress response has not yet been defined. Here we report that activation of TAK1 through TAK1 binding protein 2 (TAB2) is required for liver protection from stressors. We found that a bacterial moiety, lipopolysaccharides (LPS), activated TAK1 in primary hepatocytes, which was diminished by deletion of TAB2. Mice having hepatocyte-specific deletion of the Tab2 gene exhibited only late-onset moderate liver lesions but were hypersensitive to LPS-induced liver injury. Furthermore, we show that a chemical stressor induced greatly exaggerated liver injury in hepatocyte-specific Tab2-deficient mice. These results demonstrate that TAB2 is a sensor of stress conditions in the liver and functions to protect the liver by activating the TAK1 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/prevention & control , Diethylnitrosamine/toxicity , Lipopolysaccharides/toxicity , MAP Kinase Kinase Kinases/physiology , Alkylating Agents/toxicity , Animals , Blotting, Western , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Electrophoretic Mobility Shift Assay , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Integrases/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
J Biol Chem ; 288(19): 13467-80, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23546880

ABSTRACT

BACKGROUND: The role of Smad-independent TGF-ß signaling in craniofacial development is poorly elucidated. RESULTS: In craniofacial mesenchymal cells, Tak1 regulates both R-Smad C-terminal and linker region phosphorylation in TGF-ß signaling. CONCLUSION: Tak1 plays an irreplaceable role in craniofacial ecto-mesenchyme during embryogenesis. SIGNIFICANCE: Understanding the mechanisms of TGF-ß signaling contributes to knowledge of pathogenetic mechanisms underlying common craniofacial birth defects. Although the importance of TGF-ß superfamily signaling in craniofacial growth and patterning is well established, the precise details of its signaling mechanisms are still poorly understood. This is in part because of the concentration of studies on the role of the Smad-dependent (so-called "canonical") signaling pathways relative to the Smad-independent ones in many biological processes. Here, we have addressed the role of TGF-ß-activated kinase 1 (Tak1, Map3k7), one of the key mediators of Smad-independent (noncanonical) TGF-ß superfamily signaling in craniofacial development, by deleting Tak1 specifically in the neural crest lineage. Tak1-deficient mutants display a round skull, hypoplastic maxilla and mandible, and cleft palate resulting from a failure of palatal shelves to appropriately elevate and fuse. Our studies show that in neural crest-derived craniofacial ecto-mesenchymal cells, Tak1 is not only required for TGF-ß- and bone morphogenetic protein-induced p38 Mapk activation but also plays a role in agonist-induced C-terminal and linker region phosphorylation of the receptor-mediated R-Smads. Specifically, we demonstrate that the agonist-induced linker region phosphorylation of Smad2 at Thr-220, which has been shown to be critical for full transcriptional activity of Smad2, is dependent on Tak1 activity and that in palatal mesenchymal cells TGFßRI and Tak1 kinases mediate both overlapping and distinct TGF-ß2-induced transcriptional responses. To summarize, our results suggest that in neural crest-derived ecto-mesenchymal cells, Tak1 provides a critical point of intersection in a complex dialogue between the canonical and noncanonical arms of TGF-ß superfamily signaling required for normal craniofacial development.


Subject(s)
MAP Kinase Kinase Kinases/physiology , Neural Crest/cytology , Protein Processing, Post-Translational , Smad Proteins/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Cleft Palate/enzymology , Cleft Palate/genetics , Ectoderm/cytology , Female , Gene Expression Regulation, Developmental , Head/embryology , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Kinase Kinases/genetics , Male , Mandible/abnormalities , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , TGF-beta Superfamily Proteins/physiology , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
16.
Mol Cell Biol ; 33(6): 1149-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23297344

ABSTRACT

Antigen receptors activate pathways that control cell survival, proliferation, and differentiation. Two important targets of antigen receptors, NF-κB and Jun N-terminal kinase (JNK), are activated downstream of CARMA1, a scaffolding protein that nucleates a complex including BCL10, MALT1, and other IκB kinase (IKK)-signalosome components. Somatic mutations that constitutively activate CARMA1 occur frequently in diffuse large B cell lymphoma (DLBCL) and mediate essential survival signals. Mechanisms that downregulate this pathway might thus yield important therapeutic targets. Stimulation of antigen receptors induces not only BCL10 activation but also its degradation downstream of CARMA1, thereby ultimately limiting signals to its downstream targets. Here, using lymphocyte cell models, we identify a kinase-independent requirement for TAK1 and its adaptor, TAB1, in antigen receptor-induced BCL10 degradation. We show that TAK1 acts as an adaptor for E3 ubiquitin ligases that target BCL10 for degradation. Functionally, TAK1 overexpression restrains CARMA1-dependent activation of NF-κB by reducing BCL10 levels. TAK1 also promotes counterselection of NF-κB-addicted DLBCL lines by a dual mechanism involving kinase-independent degradation of BCL10 and kinase-dependent activation of JNK. Thus, by directly promoting BCL10 degradation, TAK1 counterbalances NF-κB and JNK signals essential for the activation and survival of lymphocytes and CARMA1-addicted lymphoma types.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CARD Signaling Adaptor Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Chickens , HEK293 Cells , Humans , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Protein Kinase C/metabolism , Protein Kinase C beta , Proteolysis , Receptors, Antigen/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Ubiquitination
17.
PLoS One ; 7(11): e51073, 2012.
Article in English | MEDLINE | ID: mdl-23226465

ABSTRACT

A cytokine/stress signaling kinase Tak1 (Map3k7) deficiency is known to impair hematopoietic progenitor cells. However, the role of TAK1 signaling in the stem cell function of the hematopoietic system is not yet well defined. Here we characterized hematopoietic stem cells (HSCs) harboring deletion of Tak1 and its activators, Tak1 binding proteins 1 and 2 (Tab1 and Tab2) using a competitive transplantation assay in a mouse model. Tak1 single or Tab1/Tab2 double deletions completely eliminated the reconstitution activity of HSCs, whereas Tab1 or Tab2 single deletion did not cause any abnormality. Tak1 single or Tab1/Tab2 double deficient lineage-negative, Sca-1(+), c-Kit(+) (LSK) cells did not proliferate and underwent cell death. We found that Tnfr1 deficiency restored the reconstitution activity of Tak1 deficient bone marrow cells for 6-18 weeks. However, the reconstitution activity of Tak1- and Tnfr1-double deficient bone marrow cells declined over the long term, and the number of phenotypically identified long-term hematopoietic stem cells were diminished. Our results indicate that TAB1- or TAB2-dependent activation of TAK1 is required for maintenance of the hematopoietic system through two mechanisms: one is prevention of TNF-dependent cell death and the other is TNF-independent maintenance of long-term HSC.


Subject(s)
Hematopoietic Stem Cells/enzymology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, Surface/metabolism , Bone Marrow Cells/enzymology , Cell Death , Cell Proliferation , Chimerism , Hematopoietic Stem Cells/cytology , Humans , MAP Kinase Kinase Kinases/deficiency , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/metabolism , Time Factors
18.
Blood ; 120(18): 3846-57, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22972987

ABSTRACT

TGF-ß activated kinase 1 (TAK1) is a mediator of various cytokine signaling pathways. Germline deficiency of Tak1 causes multiple abnormalities, including dilated blood vessels at midgestation. However, the mechanisms by which TAK1 regulates vessel formation have not been elucidated. TAK1 binding proteins 1 and 2 (TAB1 and TAB2) are activators of TAK1, but their roles in embryonic TAK1 signaling have not been determined. In the present study, we characterized mouse embryos harboring endothelial-specific deletions of Tak1, Tab1, or Tab2 and found that endothelial TAK1 and TAB2, but not TAB1, were critically involved in vascular formation. TAK1 deficiency in endothelial cells caused increased cell death and vessel regression at embryonic day 10.5 (E10.5). Deletion of TNF signaling largely rescued endothelial cell death in TAK1-deficient embryos at E10.5. However, embryos deficient in both TAK1 and TNF signaling still exhibited dilated capillary networks at E12.5. TAB2 deficiency caused reduced TAK1 activity, resulting in abnormal capillary blood vessels, similar to the compound deficiency of TAK1 and TNF signaling. Ablation of either TAK1 or TAB2 impaired cell migration and tube formation. Our results show that endothelial TAK1 signaling is important for 2 biologic processes in angiogenesis: inhibiting TNF-dependent endothelial cell death and promoting TNF-independent angiogenic cell migration.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , MAP Kinase Kinase Kinases/metabolism , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Survival , Embryo, Mammalian , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , RNA, Small Interfering , Umbilical Veins
19.
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
20.
EMBO J ; 30(24): 4908-20, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22081109

ABSTRACT

Autophagic responses are coupled to the activation of the inhibitor of NF-κB kinase (IKK). Here, we report that the essential autophagy mediator Beclin 1 and TGFß-activated kinase 1 (TAK1)-binding proteins 2 and 3 (TAB2 and TAB3), two upstream activators of the TAK1-IKK signalling axis, constitutively interact with each other via their coiled-coil domains (CCDs). Upon autophagy induction, TAB2 and TAB3 dissociate from Beclin 1 and bind TAK1. Moreover, overexpression of TAB2 and TAB3 suppresses, while their depletion triggers, autophagy. The expression of the C-terminal domain of TAB2 or TAB3 or that of the CCD of Beclin 1 competitively disrupts the interaction between endogenous Beclin 1, TAB2 and TAB3, hence stimulating autophagy through a pathway that requires endogenous Beclin 1, TAK1 and IKK to be optimally efficient. These results point to the existence of an autophagy-stimulatory 'switch' whereby TAB2 and TAB3 abandon inhibitory interactions with Beclin 1 to engage in a stimulatory liaison with TAK1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Beclin-1 , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Protein Structure, Tertiary , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...