Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Nat Immunol ; 18(5): 519-529, 2017 05.
Article in English | MEDLINE | ID: mdl-28346409

ABSTRACT

Obesity is associated with metabolic inflammation and endoplasmic reticulum (ER) stress, both of which promote metabolic disease progression. Adipose tissue macrophages (ATMs) are key players orchestrating metabolic inflammation, and ER stress enhances macrophage activation. However, whether ER stress pathways underlie ATM regulation of energy homeostasis remains unclear. Here, we identified inositol-requiring enzyme 1α (IRE1α) as a critical switch governing M1-M2 macrophage polarization and energy balance. Myeloid-specific IRE1α abrogation in Ern1f/f; Lyz2-Cre mice largely reversed high-fat diet (HFD)-induced M1-M2 imbalance in white adipose tissue (WAT) and blocked HFD-induced obesity, insulin resistance, hyperlipidemia and hepatic steatosis. Brown adipose tissue (BAT) activity, WAT browning and energy expenditure were significantly higher in Ern1f/f; Lyz2-Cre mice. Furthermore, IRE1α ablation augmented M2 polarization of macrophages in a cell-autonomous manner. Thus, IRE1α senses protein unfolding and metabolic and immunological states, and consequently guides ATM polarization. The macrophage IRE1α pathway drives obesity and metabolic syndrome through impairing BAT activity and WAT browning.


Subject(s)
Adipose Tissue, Brown/physiology , Adipose Tissue, White/pathology , Endoribonucleases/metabolism , Macrophages/physiology , Obesity/immunology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/genetics , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress , Endoribonucleases/genetics , Energy Metabolism/genetics , Humans , Macrophage Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics
2.
Blood ; 139(16): 2547-2552, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34990508

ABSTRACT

Intestinal iron absorption is activated during increased systemic demand for iron. The best-studied example is iron deficiency anemia, which increases intestinal iron absorption. Interestingly, the intestinal response to anemia is very similar to that of iron overload disorders, as both the conditions activate a transcriptional program that leads to a hyperabsorption of iron via the transcription factor hypoxia-inducible factor 2α (HIF2α). However, pathways for selective targeting of intestine-mediated iron overload remain unknown. Nuclear receptor coactivator 4 (NCOA4) is a critical cargo receptor for autophagic breakdown of ferritin and the subsequent release of iron, in a process termed ferritinophagy. Our work demonstrates that NCOA4-mediated intestinal ferritinophagy is integrated into systemic iron demand via HIF2α. To demonstrate the importance of the intestinal HIF2α/ferritinophagy axis in systemic iron homeostasis, whole-body and intestine-specific NCOA4-/- mouse lines were generated and assessed. The analyses revealed that the intestinal and systemic response to iron deficiency was not altered after disruption of intestinal NCOA4. However, in a mouse model of hemochromatosis, ablation of intestinal NCOA4 was protective against iron overload. Therefore, NCOA4 can be selectively targeted for the management of iron overload disorders without disrupting the physiological processes involved in the response to systemic iron deficiency.


Subject(s)
Anemia , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hemochromatosis , Iron Overload , Animals , Enterocytes/metabolism , Hemochromatosis/genetics , Iron/metabolism , Mice , Nuclear Receptor Coactivators/genetics , Transcription Factors/metabolism
3.
Annu Rev Nutr ; 42: 91-113, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35584814

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), a spectrum of metabolic liver disease associated with obesity, ranges from relatively benign hepatic steatosis to nonalcoholic steatohepatitis (NASH). The latter is characterized by persistent liver injury, inflammation, and liver fibrosis, which collectively increase the risk for end-stage liver diseases such as cirrhosis and hepatocellular carcinoma. Recent work has shed new light on the pathophysiology of NAFLD/NASH, particularly the role of genetic, epigenetic, and dietary factors and metabolic dysfunctions in other tissues in driving excess hepatic fat accumulation and liver injury. In parallel, single-cell RNA sequencing studies have revealed unprecedented details of the molecular nature of liver cell heterogeneity, intrahepatic cross talk, and disease-associated reprogramming of the liver immune and stromal vascular microenvironment. This review covers the recent advances in these areas, the emerging concepts of NASH pathogenesis, and potential new therapeutic opportunities.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/pathology , Humans , Liver/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Liver Neoplasms/complications , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Tumor Microenvironment
4.
J Biol Chem ; 296: 100235, 2021.
Article in English | MEDLINE | ID: mdl-33376138

ABSTRACT

Epigenetic mechanisms that alter heritable gene expression and chromatin structure play an essential role in many biological processes, including liver function. Human MOF (males absent on the first) is a histone acetyltransferase that is globally downregulated in human steatohepatitis. However, the function of MOF in the liver remains unclear. Here, we report that MOF plays an essential role in adult liver. Genetic deletion of Mof by Mx1-Cre in the liver leads to acute liver injury, with increase of lipid deposition and fibrosis akin to human steatohepatitis. Surprisingly, hepatocyte-specific Mof deletion had no overt liver abnormality. Using the in vitro coculturing experiment, we show that Mof deletion-induced liver injury requires coordinated changes and reciprocal signaling between hepatocytes and Kupffer cells, which enables feedforward regulation to augment inflammation and apoptotic responses. At the molecular level, Mof deletion induced characteristic changes in metabolic gene programs, which bore noticeable similarity to the molecular signature of human steatohepatitis. Simultaneous deletion of Mof in both hepatocytes and macrophages results in enhanced expression of inflammatory genes and NO signaling in vitro. These changes, in turn, lead to apoptosis of hepatocytes and lipotoxicity. Our work highlights the importance of histone acetyltransferase MOF in maintaining metabolic liver homeostasis and sheds light on the epigenetic dysregulation in liver pathogenesis.


Subject(s)
Histone Acetyltransferases/genetics , Inflammation/metabolism , Liver Diseases/genetics , Liver/injuries , Nitric Oxide/genetics , Apoptosis/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Histone Acetyltransferases/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Lipids/adverse effects , Lipids/genetics , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Macrophages/metabolism , Macrophages/pathology , Nitric Oxide/metabolism , Signal Transduction/genetics
5.
Proc Natl Acad Sci U S A ; 116(38): 19090-19097, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31481626

ABSTRACT

Aberrant T cell development is a pivotal risk factor for autoimmune disease; however, the underlying molecular mechanism of T cell overactivation is poorly understood. Here, we identified NF-κB-inducing kinase (NIK) and IkB kinase α (IKKα) in thymic epithelial cells (TECs) as essential regulators of T cell development. Mouse TEC-specific ablation of either NIK or IKKα resulted in severe T cell-mediated inflammation, injury, and fibrosis in the liver and lung, leading to premature death within 18 d of age. NIK or IKKα deficiency abrogated medullary TEC development, and led to breakdown of central tolerance, production of autoreactive T cells, and fatal autoimmune destruction in the liver and lung. TEC-specific ablation of NIK or IKKα also impaired thymic T cell development from the double-negative through the double-positive stages and inhibited peripheral B cell development. These results unravel a hitherto unrecognized essential role of TEC-intrinsic NIK and IKKα pathways in autoimmunity and T cell-instigated chronic liver and lung diseases.


Subject(s)
Autoimmunity/immunology , I-kappa B Kinase/physiology , Inflammation/immunology , Liver/immunology , Lung/immunology , Protein Serine-Threonine Kinases/physiology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Homeostasis , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Thymus Gland/metabolism , Thymus Gland/pathology , NF-kappaB-Inducing Kinase
6.
Am J Respir Cell Mol Biol ; 65(2): 214-221, 2021 08.
Article in English | MEDLINE | ID: mdl-33891828

ABSTRACT

Obesity elevates the plasma level of leptin, which has been associated with hypertension. Our recent studies in mice demonstrated that leptin increases blood pressure by activating the carotid sinus nerve, which transmits the chemosensory input from carotid bodies (CBs) to the medullary centers, and that the effect of leptin is mediated via Trpm7 (TRP [transient receptor potential] melastatin 7) channels in CB glomus cells. We also found that Trpm7 overexpression and Trpm7 promoter demethylation in CBs correlate positively with the hyperleptinemia and leptin receptor overexpression in CBs. Hence, we postulated that leptin epigenetically regulates Trpm7 expression in CBs. We addressed our hypothesis by using rat adrenal pheochromocytoma (PC12) cells as a model of CB glomus cells. PC12 cells expressing LEPRb (long, active form of leptin receptor) showed dramatic induction of the promoter activity and expression of Trpm7 upon leptin treatment. The increased Trpm7 expression coincided with the reduction of CpG site-specific methylation and trimethylation of H3K27 (H3 [histone 3] K27 [lysine 27]) and the increase of acetylation of H3K27 and trimethylation of H3K4 (H3 lysine 4) at the Trpm7 promoter. The inhibitor of STAT3 (signal transducer and activator of transcription 3) signaling, SD1008, reversed the leptin-induced Trpm7 promoter activity via modulations of the binding of pSTAT3 (phosphorylated STAT3) and DNMT3B (DNA methyltransferase 3B) and modifications of H3K27 and H3K4 at the Trpm7 promoter. Our results suggest that leptin-activated pSTAT3 epigenetically regulates the transcription of Trpm7 through DNA methylation and histone modifications. Because epigenetic changes are reversible, targeting epigenetic modifications of Trpm7 may serve as a new therapeutic approach for the treatment of hypertension in obesity.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Leptin/pharmacology , Neoplasm Proteins/biosynthesis , Pheochromocytoma/metabolism , TRPM Cation Channels/biosynthesis , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Animals , Neoplasm Proteins/genetics , PC12 Cells , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Rats , TRPM Cation Channels/genetics
7.
FASEB J ; 34(11): 14863-14877, 2020 11.
Article in English | MEDLINE | ID: mdl-32918517

ABSTRACT

Appropriate control of hepatic gluconeogenesis is essential for the organismal survival upon prolonged fasting and maintaining systemic homeostasis under metabolic stress. Here, we show protein arginine methyltransferase 1 (PRMT1), a key enzyme that catalyzes the protein arginine methylation process, particularly the isoform encoded by Prmt1 variant 2 (PRMT1V2), is critical in regulating gluconeogenesis in the liver. Liver-specific deletion of Prmt1 reduced gluconeogenic capacity in cultured hepatocytes and in the liver. Prmt1v2 was expressed at a higher level compared to Prmt1v1 in hepatic tissue and cells. Gain-of-function of PRMT1V2 clearly activated the gluconeogenic program in hepatocytes via interactions with PGC1α, a key transcriptional coactivator regulating gluconeogenesis, enhancing its activity via arginine methylation, while no effects of PRMT1V1 were observed. Similar stimulatory effects of PRMT1V2 in controlling gluconeogenesis were observed in human HepG2 cells. PRMT1, specifically PRMT1V2, was stabilized in fasted liver and hepatocytes treated with glucagon, in a PGC1α-dependent manner. PRMT1, particularly Prmt1v2, was significantly induced in the liver of streptozocin-induced type 1 diabetes and high fat diet-induced type 2 diabetes mouse models and liver-specific Prmt1 deficiency drastically ameliorated diabetic hyperglycemia. These findings reveal that PRMT1 modulates gluconeogenesis and mediates glucose homeostasis under physiological and pathological conditions, suggesting that deeper understanding how PRMT1 contributes to the coordinated efforts in glycemic control may ultimately present novel therapeutic strategies that counteracts hyperglycemia in disease settings.


Subject(s)
Gluconeogenesis , Hepatocytes/metabolism , Hyperglycemia/genetics , Protein-Arginine N-Methyltransferases/metabolism , Animals , Cells, Cultured , Gain of Function Mutation , Glucagon/metabolism , Glucose/metabolism , Hep G2 Cells , Humans , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein-Arginine N-Methyltransferases/genetics
8.
Hepatology ; 68(2): 533-546, 2018 08.
Article in English | MEDLINE | ID: mdl-29506314

ABSTRACT

Obesity is associated with both endoplasmic reticulum (ER) stress and chronic metabolic inflammation. ER stress activates the unfolded protein response (UPR) and has been implicated in a variety of cancers, including hepatocellular carcinoma (HCC). It is unclear whether individual UPR pathways are mechanistically linked to HCC development, however. Here we report a dual role for inositol-requiring enzyme 1α (IRE1α), the ER-localized UPR signal transducer, in obesity-promoted HCC development. We found that genetic ablation of IRE1α in hepatocytes not only markedly reduced the occurrence of diethylnitrosamine (DEN)-induced HCC in liver-specific IRE1α knockout (LKO) mice when fed a normal chow (NC) diet, but also protected against the acceleration of HCC progression during high-fat diet (HFD) feeding. Irrespective of their adiposity states, LKO mice showed decreased hepatocyte proliferation and signal transducer and activator of transcription 3 (STAT3) activation, even in the face of increased hepatic apoptosis. Furthermore, IRE1α abrogation blunted obesity-associated activation of hepatic inhibitor of nuclear factor kappa B kinase subunit beta (IKKß)-nuclear factor kappa B (NF-κB) pathway, leading to reduced production of the tumor-promoting inflammatory cytokines tumor necrosis factor (TNF) and interleukin 6 (IL-6). Importantly, higher IRE1α expression along with elevated STAT3 phosphorylation was also observed in the tumor tissues from human HCC patients, correlating with their poorer survival rate. CONCLUSION: IRE1α acts in a feed-forward loop during obesity-induced metabolic inflammation to promote HCC development through STAT3-mediated hepatocyte proliferation. (Hepatology 2018).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Endoribonucleases/metabolism , Liver Neoplasms/metabolism , Obesity/complications , Protein Serine-Threonine Kinases/metabolism , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Cytokines/metabolism , Diet, High-Fat , Diethylnitrosamine/pharmacology , Hepatocytes/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/veterinary , STAT3 Transcription Factor/metabolism , Signal Transduction
9.
FASEB J ; 32(4): 1830-1840, 2018 04.
Article in English | MEDLINE | ID: mdl-29180441

ABSTRACT

Psychiatric disorders are associated with aberrant brain development and/or aggressive behavior and are influenced by genetic factors; however, genes that affect brain aggression circuits remain elusive. Here, we show that neuronal Src-homology-2 (SH2)B adaptor protein-1 ( Sh2b1) is indispensable for both brain growth and protection against aggression. Global and brain-specific deletion of Sh2b1 decreased brain weight and increased aggressive behavior. Global and brain-specific Sh2b1 knockout (KO) mice exhibited fatal, intermale aggression. In a resident-intruder paradigm, latency to attack was markedly reduced, whereas the number and the duration of attacks was significantly increased in global and brain-specific Sh2b1 KO mice compared with wild-type littermates. Consistently, core aggression circuits were activated to a higher level in global and brain-specific Sh2b1 KO males, based on c-fos immunoreactivity in the amygdala and periaqueductal gray. Brain-specific restoration of Sh2b1 normalized brain size and reversed pathologic aggression and aberrant activation of core aggression circuits in Sh2b1 KO males. SH2B1 mutations in humans were linked to aberrant brain development and behavior. At the molecular level, Sh2b1 enhanced neurotrophin-stimulated neuronal differentiation and protected against oxidative stress-induced neuronal death. Our data suggest that neuronal Sh2b1 promotes brain development and the integrity of core aggression circuits, likely through enhancing neurotrophin signaling.-Jiang, L., Su, H., Keogh, J. M., Chen, Z., Henning, E., Wilkinson, P., Goodyer, I., Farooqi, I. S., Rui, L. Neural deletion of Sh2b1 results in brain growth retardation and reactive aggression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Aggression , Brain/growth & development , Adult , Animals , Brain/metabolism , Brain/physiology , Child , Female , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mutation , PC12 Cells , Rats
10.
FASEB J ; 31(2): 711-718, 2017 02.
Article in English | MEDLINE | ID: mdl-27871061

ABSTRACT

Potent and selective chemical probes are valuable tools for discovery of novel treatments for human diseases. NF-κB-inducing kinase (NIK) is a key trigger in the development of liver injury and fibrosis. Whether inhibition of NIK activity by chemical probes ameliorates liver inflammation and injury is largely unknown. In this study, a small-molecule inhibitor of NIK, B022, was found to be a potent and selective chemical probe for liver inflammation and injury. B022 inhibited the NIK signaling pathway, including NIK-induced p100-to-p52 processing and inflammatory gene expression, both in vitro and in vivo Furthermore, in vivo administration of B022 protected against not only NIK but also CCl4-induced liver inflammation and injury. Our data suggest that inhibition of NIK is a novel strategy for treatment of liver inflammation, oxidative stress, and injury.-Ren, X., Li, X., Jia, L., Chen, D., Hou, H., Rui, L., Zhao, Y., Chen, Z. A small-molecule inhibitor of NF-κB-inducing kinase (NIK) protects liver from toxin-induced inflammation, oxidative stress, and injury.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Carbon Tetrachloride Poisoning , Cell Line , Humans , Male , Mice , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , RNA, Messenger , Reactive Oxygen Species , NF-kappaB-Inducing Kinase
11.
J Hepatol ; 67(1): 100-109, 2017 07.
Article in English | MEDLINE | ID: mdl-28267623

ABSTRACT

BACKGROUND & AIMS: The liver is an immunologically-privileged organ. Breakdown of liver immune privilege has been reported in chronic liver disease; however, the role of adaptive immunity in liver injury is poorly defined. Nuclear factor-κB-inducing kinase (NIK) is known to regulate immune tissue development, but its role in maintaining liver homeostasis remains unknown. This study aimed to assess the role of NIK, particularly thymic NIK, in regulating liver adaptive immunity. METHODS: NIK was deleted systemically or conditionally using the Cre/loxp system. Cluster of differentiation [CD]4+ or CD8+ T cells were depleted using anti-CD4 or anti-CD8 antibody. Donor bone marrows or thymi were transferred into recipient mice. Immune cells were assessed by immunohistochemistry and flow cytometry. RESULTS: Global, but not liver-specific or hematopoietic lineage cell-specific, deletion of NIK induced fatal liver injury, inflammation, and fibrosis. Likewise, adoptive transfer of NIK-null, but not wild-type, thymi into immune-deficient mice induced liver inflammation, injury, and fibrosis in recipients. Liver inflammation was characterized by a massive expansion of T cells, particularly the CD4+ T cell subpopulation. Depletion of CD4+, but not CD8+, T cells fully protected against liver injury, inflammation, and fibrosis in NIK-null mice. NIK deficiency also resulted in inflammation in the lung, kidney, and pancreas, but to a lesser degree relative to the liver. CONCLUSIONS: Thymic NIK suppresses development of autoreactive T cells against liver antigens, and NIK deficiency in the thymus results in CD4+ T cell-orchestrated autoimmune hepatitis and liver fibrosis. Thus, thymic NIK is essential for the maintenance of liver immune privilege and liver homeostasis. LAY SUMMARY: We found that global or thymus-specific ablation of the NIK gene results in fatal autoimmune liver disease in mice. NIK-deficient mice develop liver inflammation, injury, and fibrosis. Our findings indicate that thymic NIK is essential for the maintenance of liver integrity and homeostasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis, Autoimmune/etiology , Liver Cirrhosis, Experimental/etiology , Liver/immunology , Protein Serine-Threonine Kinases/physiology , Thymus Gland/physiology , Adaptive Immunity , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappaB-Inducing Kinase
12.
J Lipid Res ; 57(7): 1219-30, 2016 07.
Article in English | MEDLINE | ID: mdl-27252523

ABSTRACT

Upon food intake, insulin stimulates de novo lipogenesis (DNL) in hepatocytes via the AKT-mTORC1-sterol regulatory element-binding protein (SREBP)-1c pathway. How insulin maintains the maximal SREBP-1c activities during the entire feeding state remains elusive. We previously reported that insulin induced b-ZIP transcription factor, E4-binding protein 4 (E4BP4), in hepatocytes. In the current study, we show that insulin injection increases hepatic E4bp4 expression by activating the AKT-mTORC1-SREBP-1c pathway in hepatocytes. E4bp4-deficient hepatocytes not only fail to maintain robust DNL but also become resistant to SREBP-1c-induced lipogenesis. In vivo, acute depletion of E4bp4 in the liver by adenoviral shRNA reduces the expression of lipogenic enzymes and results in reduced levels of serum triglycerides and cholesterol during the postprandial phase. In hepatocytes, E4BP4 interacts with nuclear SREBP-1c to preserve its acetylation, and subsequently protects it from ubiquitination-dependent degradation. In conclusion, the current studies uncover a novel positive feedback pathway mediated by E4BP4 to augment SREBP-1c-mediated DNL in the liver during the fed state.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Fatty Liver/metabolism , Insulin/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Transcription, Genetic , Animals , Appetite Regulation/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Fatty Acids/biosynthesis , Fatty Acids/genetics , Fatty Liver/pathology , Gene Expression Regulation , Hepatocytes/metabolism , Humans , Insulin/genetics , Lipogenesis/genetics , Liver/metabolism , Mice , Postprandial Period , Proteolysis , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/biosynthesis
13.
J Biol Chem ; 290(18): 11704-14, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25792747

ABSTRACT

Ser/Thr kinase NIK (NF-κB-inducing kinase) mediates the activation of the noncanonical NF-κB2 pathway, and it plays an important role in regulating immune cell development and liver homeostasis. NIK levels are extremely low in quiescent cells due to ubiquitin/proteasome-mediated degradation, and cytokines stimulate NIK activation through increasing NIK stability; however, regulation of NIK stability is not fully understood. Here we identified CHIP (carboxyl terminus of HSC70-interacting protein) as a new negative regulator of NIK. CHIP contains three N-terminal tetratricopeptide repeats (TPRs), a middle dimerization domain, and a C-terminal U-box. The U-box domain contains ubiquitin E3 ligase activity that promotes ubiquitination of CHIP-bound partners. We observed that CHIP bound to NIK via its TPR domain. In both HEK293 and primary hepatocytes, overexpression of CHIP markedly decreased NIK levels at least in part through increasing ubiquitination and degradation of NIK. Accordingly, CHIP suppressed NIK-induced activation of the noncanonical NF-κB2 pathway. CHIP also bound to TRAF3, and CHIP and TRAF3 acted coordinately to efficiently promote NIK degradation. The TPR but not the U-box domain was required for CHIP to promote NIK degradation. In mice, hepatocyte-specific overexpression of NIK resulted in liver inflammation and injury, leading to death, and liver-specific expression of CHIP reversed the detrimental effects of hepatic NIK. Our data suggest that CHIP/TRAF3/NIK interactions recruit NIK to E3 ligase complexes for ubiquitination and degradation, thus maintaining NIK at low levels. Defects in CHIP regulation of NIK may result in aberrant NIK activation in the liver, contributing to live injury, inflammation, and disease.


Subject(s)
Down-Regulation , Liver/injuries , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Enzyme Activation , HEK293 Cells , Hepatocytes/metabolism , Humans , Liver/cytology , Male , Mice , NF-kappa B p52 Subunit/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proteolysis , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/chemistry , NF-kappaB-Inducing Kinase
14.
J Biol Chem ; 289(37): 25925-35, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25063808

ABSTRACT

The clock protein BMAL1 (brain and muscle Arnt-like protein 1) participates in circadian regulation of lipid metabolism, but its contribution to insulin AKT-regulated hepatic lipid synthesis is unclear. Here we used both Bmal1(-/-) and acute liver-specific Bmal1-depleted mice to study the role of BMAL1 in refeeding-induced de novo lipogenesis in the liver. Both global deficiency and acute hepatic depletion of Bmal1 reduced lipogenic gene expression in the liver upon refeeding. Conversely, Bmal1 overexpression in mouse liver by adenovirus was sufficient to elevate the levels of mRNA of lipogenic enzymes. Bmal1(-/-) primary mouse hepatocytes displayed decreased levels of de novo lipogenesis and lipogenic enzymes, supporting the notion that BMAL1 regulates lipid synthesis in hepatocytes in a cell-autonomous manner. Both refed mouse liver and insulin-treated primary mouse hepatocytes showed impaired AKT activation in the case of either Bmal1 deficiency or Bmal1 depletion by adenoviral shRNA. Restoring AKT activity by a constitutively active mutant of AKT nearly normalized de novo lipogenesis in Bmal1(-/-) hepatocytes. Finally, Bmal1 deficiency or knockdown decreased the protein abundance of RICTOR, the key component of the mTORC2 complex, without affecting the gene expression of key factors of insulin signaling. Thus, our study uncovered a novel metabolic function of hepatic BMAL1 that promotes de novo lipogenesis via the insulin-mTORC2-AKT signaling during refeeding.


Subject(s)
ARNTL Transcription Factors/genetics , Insulin/metabolism , Lipogenesis , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , ARNTL Transcription Factors/antagonists & inhibitors , Animals , Eating/genetics , Gene Expression Regulation , Hepatocytes/metabolism , Insulin Resistance/genetics , Lipid Metabolism/genetics , Liver/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , Signal Transduction
15.
Am J Physiol Endocrinol Metab ; 308(6): E460-9, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25628422

ABSTRACT

Myeloid cells, particularly macrophages, mediate metabolic inflammation, thus promoting insulin resistance and metabolic disease progression in obesity. Numerous cytokines, toxic metabolites, damage-associated molecular patterns, and pathogen-associated molecular patterns are involved in activating macrophages via their cognate receptors in obesity. TRAF3 (TNF receptor-associated factor 3) is a common signaling molecule for these ligands/receptors and negatively regulates the proinflammatory NF-κB and MAPK pathways, but its metabolic activity is unknown. We here show that myeloid cell TRAF3 is required for metabolic inflammation and metabolic disease progression in obesity. Myeloid cell-specific deletion of TRAF3 significantly attenuated insulin resistance, hyperglycemia, hyperinsulinemia, glucose intolerance, and hepatic steatosis in mice with either genetic (ob/ob) or high-fat diet (HFD)-induced obesity. Myeloid cell-specific deletion of TRAF3 had the opposite effects on metabolic inflammation between obese and lean mice. It decreased the expression of proinflammatory cytokines in the liver and adipose tissue of obese mice and largely prevented HFD-induced inflammation in these metabolic tissues; by contrast, in lean mice, it increased the expression of proinflammatory cytokines in the liver and adipose tissue. These data suggest that, in obesity progression, myeloid TRAF3 functionally switches its activity from anti-inflammatory to proinflammatory modes, thereby coupling overnutrition to metabolic inflammation, insulin resistance, and metabolic disease.


Subject(s)
Fatty Liver/genetics , Inflammation/genetics , Insulin Resistance/genetics , Obesity/complications , Obesity/genetics , TNF Receptor-Associated Factor 3/physiology , Animals , Cells, Cultured , Fatty Liver/immunology , Fatty Liver/metabolism , Gene Deletion , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Myeloid Cells/metabolism , Obesity/immunology , Obesity/metabolism
16.
J Hepatol ; 62(3): 590-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25457211

ABSTRACT

BACKGROUND & AIMS: As the main detoxifying organ of the body, the liver possesses a remarkable ability to regenerate after toxic injury, tissue resection or viral infection. A growing number of cellular signaling pathways have been implicated in orchestrating the process of liver regeneration. Here we investigated the role of inositol-requiring enzyme-1α (IRE1α), a key signal transducer of the unfolded protein response (UPR), in liver regeneration. METHODS: Using mice with hepatocyte-specific deletion of IRE1α, we examined the role of IRE1α in liver regeneration after challenges with carbon tetrachloride (CCl4) or hepatic surgery. We also investigated if IRE1α deficiency could affect the activation state of signal transducer and activator of transcription 3 (STAT3) in hepatocytes. Using co-immunoprecipitation and glutathione S-transferase (GST) pull-down assays, we analyzed whether IRE1α could interact with STAT3 to regulate its phosphorylation. RESULTS: We found that in response to CCl4-induced liver damage or after two-thirds partial hepatectomy (PH), abrogation of IRE1α caused marked exacerbation of liver injury and impairment in regenerative proliferation of hepatocytes in mice. Furthermore, IRE1α deficiency resulted in dampened STAT3 activation, and restoration of IRE1α expression led to sustained phosphorylation of STAT3 in IRE1α-null hepatocytes. Additionally, IRE1α could directly and constitutively associate with STAT3, leading to elevated phosphorylation when stimulated by IL-6. CONCLUSIONS: These results suggest that IRE1α may promote liver regeneration through acting as a signaling platform to regulate the STAT3 pathway.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Endoribonucleases/physiology , Liver Regeneration/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Carbon Tetrachloride/toxicity , Cell Proliferation/physiology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , DNA-Binding Proteins/metabolism , Endoribonucleases/deficiency , Endoribonucleases/genetics , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Regeneration/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Regulatory Factor X Transcription Factors , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/metabolism
17.
Hepatology ; 60(6): 2065-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25088600

ABSTRACT

UNLABELLED: Damaged, necrotic, or apoptotic hepatocytes release damage-associated molecular patterns that initiate sterile inflammation, and liver inflammation drives liver injury and fibrosis. Here we identified hepatic nuclear factor kappa B (NF-κB)-inducing kinase (NIK), a Ser/Thr kinase, as a novel trigger of fatal liver inflammation. NIK is activated by a broad spectrum of stimuli. It was up-regulated in injured livers in both mice and humans. In primary mouse hepatocytes, NIK overexpression stimulated, independently of cell injury and death, release of numerous chemokines and cytokines that activated bone marrow-derived macrophages (BMDMs). BMDMs in turn secreted proapoptotic molecules that stimulated hepatocyte apoptosis. Hepatocyte-specific expression of the NIK transgene triggered massive liver inflammation, oxidative stress, hepatocyte apoptosis, and liver fibrosis, leading to weight loss, hypoglycemia, and death. Depletion of Kupffer cells/macrophages reversed NIK-induced liver destruction and death. CONCLUSION: the hepatocyte NIK-liver immune cell axis promotes liver inflammation, injury, and fibrosis, thus driving liver disease progression.


Subject(s)
Hepatocytes/physiology , Liver Diseases/metabolism , Protein Serine-Threonine Kinases/metabolism , Adult , Animals , Apoptosis , Female , Fibrosis , Humans , Immunity, Innate , Liver/pathology , Liver Diseases/immunology , Liver Diseases/pathology , Macrophages/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Oxidative Stress , Young Adult , NF-kappaB-Inducing Kinase
18.
Proc Natl Acad Sci U S A ; 108(38): 15852-7, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21911379

ABSTRACT

The endoplasmic reticulum (ER)-resident protein kinase/endoribonuclease inositol-requiring enzyme 1 (IRE1) is activated through transautophosphorylation in response to protein folding overload in the ER lumen and maintains ER homeostasis by triggering a key branch of the unfolded protein response. Here we show that mammalian IRE1α in liver cells is also phosphorylated by a kinase other than itself in response to metabolic stimuli. Glucagon-stimulated protein kinase PKA, which in turn phosphorylated IRE1α at Ser(724), a highly conserved site within the kinase activation domain. Blocking Ser(724) phosphorylation impaired the ability of IRE1α to augment the up-regulation by glucagon signaling of the expression of gluconeogenic genes. Moreover, hepatic IRE1α was highly phosphorylated at Ser(724) by PKA in mice with obesity, and silencing hepatic IRE1α markedly reduced hyperglycemia and glucose intolerance. Hence, these results suggest that IRE1α integrates signals from both the ER lumen and the cytoplasm in the liver and is coupled to the glucagon signaling in the regulation of glucose metabolism.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Endoribonucleases/metabolism , Glucagon/pharmacology , Glucose/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Cyclic AMP-Dependent Protein Kinases/genetics , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/genetics , Gene Expression Profiling , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunoblotting , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Obesity/genetics , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Serine/genetics , Serine/metabolism
19.
Cell Rep ; 43(3): 113875, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38451818

ABSTRACT

Liver injury stimulates hepatocyte replication and hepatic stellate cell (HSC) activation, thereby driving liver regeneration. Aberrant HSC activation induces liver fibrosis. However, mechanisms underlying liver regeneration and fibrosis remain poorly understood. Here, we identify hepatic Snai1 and Snai2 as important transcriptional regulators for liver regeneration and fibrosis. Partial hepatectomy or CCl4 treatment increases occupancies of Snai1 and Snai2 on cyclin A2 and D1 promoters in the liver. Snai1 and Snai2 in turn increase promoter H3K27 acetylation and cyclin A2/D1 expressions. Hepatocyte-specific deletion of both Snai1 and Snai2, but not one alone, suppresses liver cyclin A2/D1 expression and regenerative hepatocyte proliferation after hepatectomy or CCl4 treatments but augments CCl4-stimulated HSC activation and liver fibrosis. Conversely, Snai2 overexpression in the liver enhances hepatocyte replication and suppresses liver fibrosis after CCl4 treatment. These results suggest that hepatic Snai1 and Snai2 directly promote, via histone modifications, reparative hepatocyte replication and indirectly inhibit liver fibrosis.


Subject(s)
Cyclin A2 , Liver Regeneration , Animals , Mice , Cyclin A2/metabolism , Hepatectomy , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Regeneration/physiology
20.
Adv Sci (Weinh) ; : e2400437, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885417

ABSTRACT

SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.

SELECTION OF CITATIONS
SEARCH DETAIL