Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Biol Chem ; 300(6): 107319, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677512

ABSTRACT

Lipid metabolism is important for the maintenance of physiological homeostasis. Several members of the small ubiquitin-like modifier (SUMO)-specific protease (SENP) family have been reported as the regulators of lipid homeostasis. However, the function of Senp7 in lipid metabolism remains unclear. In this study, we generated both conventional and adipocyte-specific Senp7 KO mice to characterize the role of Senp7 in lipid metabolism homeostasis. Both Senp7-deficient mice displayed reduced white adipose tissue mass and decreased size of adipocytes. By analyzing the lipid droplet morphology, we demonstrated that the lipid droplet size was significantly smaller in Senp7-deficient adipocytes. Mechanistically, Senp7 could deSUMOylate the perilipin family protein Plin4 to promote the lipid droplet localization of Plin4. Our results reveal an important role of Senp7 in the maturation of lipid droplets via Plin4 deSUMOylation.


Subject(s)
Adipose Tissue, White , Lipid Droplets , Mice, Knockout , Perilipin-4 , Animals , Mice , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Lipid Droplets/metabolism , Lipid Metabolism , Perilipin-4/metabolism , Perilipin-4/genetics , Sumoylation
2.
Am J Pathol ; 189(5): 1041-1052, 2019 05.
Article in English | MEDLINE | ID: mdl-30790561

ABSTRACT

Missense mutations in the gasdermin-A3 (Gsdma3) gene are associated with skin inflammation and hair loss in mice. However, the physiological function of Gsdma3 remains unclear. Herein, we reported that mice carrying the Gsdma3 Y344H mutation that encodes a presumptive activated form of Gsdma3 show increased heat production along with lower body fat percentages. Detailed analysis indicated that this metabolic phenotype is mediated by serum IL-6-induced up-regulation of thermogenesis in brown adipose tissue. The mutant form of Gsdma3 promotes the expression of IL-6 in the epidermis in a c-Jun N-terminal kinase (JNK) signaling-dependent manner. The higher whole-body heat production in alopecia and excoriation mice could be suppressed by an IL-6 receptor/GP130 inhibitor. Our results uncovered Gsdma3/IL-6-dependent cross talk between the skin and brown adipose tissue.


Subject(s)
Adipose Tissue, Brown/physiopathology , Alopecia/physiopathology , Interleukin-6/metabolism , Proteins/metabolism , STAT3 Transcription Factor/metabolism , Skin Diseases/physiopathology , Thermogenesis , Animals , Body Temperature Regulation , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Proteins/genetics , STAT3 Transcription Factor/genetics , Signal Transduction
3.
Biochem J ; 468(2): 325-36, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25825937

ABSTRACT

Gasdermin A3 (Gsdma3) was originally identified in association with hair-loss phenotype in mouse mutants. Our previous study found that AE mutant mice, with a Y344H substitution at the C-terminal domain of Gsdma3, display inflammation-dependent alopecia and excoriation [Zhou et al. (2012) Am. J. Pathol. 180, 763-774]. Interestingly, we found that the newly-generated null mutant of Gsdma3 mice did not display the skin dysmorphology, indicating that Gsdma3 is not essential for differentiation of epidermal cells and maintenance of the hair cycle in normal physiological conditions. Consistently, human embryonic kidney (HEK)293 and HaCaT cells transfected with wild-type (WT) Gsdma3 did not show abnormal morphology. However, Gsdma3 Y344H mutation induced autophagy. Gsdma3 N-terminal domain, but not the C-terminal domain, also displayed the similar pro-autophagic activity. The Gsdma3 Y344H mutant protein and N-terminal domain-induced autophagy was associated with mitochondria and ROS generation. Co-expression of C-terminal domain reversed the cell autophagy induced by N-terminal domain. Moreover, C-terminal domain could be co-precipitated with N-terminal domain. These data indicated that the potential pro-autophagic activity of WT Gsdma3 protein is suppressed through an intramolecular inhibition mechanism. Studies on other members of the GSDM family suggested this mechanism is conserved in several sub-families.


Subject(s)
Autophagy , Cell Death , Mutation/genetics , Proteins/physiology , Animals , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria , Phenotype , Reactive Oxygen Species
4.
Cell Death Differ ; 27(2): 466-481, 2020 02.
Article in English | MEDLINE | ID: mdl-31209359

ABSTRACT

Hypomagnesemia is a significant risk factor for critically ill patients to develop sepsis, a life-threatening disease with a mortality rate over 25%. Our clinic data analysis showed that hypomagnesemia is associated with a decreased monocyte count in septic patients. At the cellular level, we found that Mg2+ inhibits pyroptosis. Specifically, Mg2+ limits the oligomerization and membrane localization of gasdermin D N-terminal (GSDMD-NT) upon the activation of either the canonical or noncanonical pyroptotic pathway. Mechanistically, we demonstrated that Ca2+ influx is a prerequisite for the function of GSDMD-NT. Mg2+ blocks Ca2+ influx by inhibiting the ATP-gated Ca2+ channel P2X7, thereby impeding the function of GSDMD-NT and inhibiting lipopolysaccharide (LPS)-induced noncanonical pyroptosis. Furthermore, Mg2+ administration protects mice from LPS-induced lethal septic shock. Together, our data reveal the underlying mechanism of how Mg2+ inhibits pyroptosis and suggest potential clinic applications of magnesium supplementation for sepsis prevention and treatment.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Magnesium/pharmacology , Phosphate-Binding Proteins/antagonists & inhibitors , Pyroptosis/drug effects , Sepsis/drug therapy , Animals , Cells, Cultured , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Magnesium/blood , Male , Mice , Mice, Inbred C57BL , Phosphate-Binding Proteins/metabolism , Sepsis/metabolism , Sepsis/pathology
5.
EBioMedicine ; 52: 102652, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32058942

ABSTRACT

BACKGROUND: Alteration of commensal bacterial composition is associated with many inflammatory diseases. However, few studies have pinpointed the specific bacterial genes that may suppress host immune responses against microbes and maintain homeostasis in the host intestine. METHODS: High-throughput screening was performed in Caenorhabditis elegans with a single gene knockout ut screening was performed in Caenorhabditis elegans with a single gene knockout Escherichia coli (E. coli) library and identified the immune suppression gene blc. The coding sequences of blc among different kinds of E. coli strains were aligned to identify the single nucleotide polymorphisms (SNPs). Physiological and biochemical experiments were performed in C. elegans and mice to explore the function of the blc variant. FINDINGS: By screening 3983 E. coli mutants, we discovered that 9 bacterial genes, when deleted, activate innate immunity in the host C. elegans. Among these 9 genes, the gene encoding blc showed a distinctive SNP in many clinically pathogenic bacteria. We found that bacteria with this SNP, which converts Blc G84 to Blc E84, are highly enriched in the faeces of patients with inflammatory bowel disease (IBD). Exposure to BlcE84-encoding bacteria resulted in epithelial barrier disruption and immune activation in both worms and mice. Detailed analysis indicated that infection with BlcE84-encoding bacteria causes a significant decrease in LPE levels in the intestine and subsequently disrupts gut epithelial integrity in mice. Consistently, the levels of LPE in patients with IBD are significantly lower than those in healthy people. Finally, supplementation with LPE, which activates LPA1/PLCß/PKC signaling, reversed the defects induced by BlcE84-encoding bacteria. INTERPRETATION: Our results identified a novel bacterial gene, blc, in E. coli that regulates host gut integrity and immunity. FUND: The Ministry of Science and Technology of China; the National Natural Science Foundation of China; and the Natural Science Foundation of Jiangsu Province.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Bacterial Outer Membrane Proteins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lipocalins/genetics , Lysophospholipids/metabolism , Polymorphism, Single Nucleotide , Animals , Base Sequence , Biomarkers , Cell Line , Disease Models, Animal , Disease Susceptibility , Escherichia coli Proteins/genetics , Homeostasis , Host-Pathogen Interactions/immunology , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Lysophospholipids/chemistry , Male , Mice , Mutation , Permeability
6.
J Mol Cell Biol ; 11(12): 1069-1082, 2019 12 19.
Article in English | MEDLINE | ID: mdl-30860577

ABSTRACT

Disrupted mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) generation are often associated with macrophage pyroptosis. It remains unclear how these forms of mitochondrial dysfunction relate to inflammasome activation and gasdermin-D (Gsdmd) cleavage, two central steps of the pyroptotic process. Here, we also found MMP collapse and ROS generation induced by Nlrp3 inflammasome activation as previous studies reported. The elimination of ROS alleviated the cleavage of Gsdmd, suggesting that Gsdmd cleavage occurs downstream of ROS release. Consistent with this result, hydrogen peroxide treatment augmented the cleavage of Gsdmd by caspase-1. Indeed, four amino acid residues of Gsdmd were oxidized under oxidative stress in macrophages. The efficiency of Gsdmd cleavage by inflammatory caspase-1 was dramatically reduced when oxidative modification was blocked by mutation of these amino acid residues. These results demonstrate that Gsdmd oxidation serves as a de novo mechanism by which mitochondrial ROS promote Nlrp3 inflammasome-dependent pyroptotic cell death.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Phosphate-Binding Proteins/metabolism , Pyroptosis , Reactive Oxygen Species/metabolism , Animals , Cell Line , Inflammasomes , Intracellular Signaling Peptides and Proteins/genetics , Mice , Models, Biological , Oxidative Stress , Phosphate-Binding Proteins/genetics , Proteolysis
7.
J Mol Cell Biol ; 11(6): 496-508, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30321352

ABSTRACT

Gasdermin B (GSDMB) has been reported to be associated with immune diseases in humans, but the detailed molecular mechanisms remain unsolved. The N-terminus of GSDMB by itself, unlike other gasdermin family proteins, does not induce cell death. Here, we show that GSDMB is highly expressed in the leukocytes of septic shock patients, which is associated with increased release of the gasdermin D (GSDMD) N-terminus. GSDMB expression and the accumulation of the N-terminal fragment of GSDMD are induced by the activation of the non-canonical pyroptosis pathway in a human monocyte cell line. The downregulation of GSDMB alleviates the cleavage of GSDMD and cell death. Consistently, the overexpression of GSDMB promotes GSDMD cleavage, accompanied by increased LDH release. We further found that GSDMB promotes caspase-4 activity, which is required for the cleavage of GSDMD in non-canonical pyroptosis, by directly binding to the CARD domain of caspase-4. Our study reveals a GSDMB-mediated novel regulatory mechanism for non-canonical pyroptosis and suggests a potential new strategy for the treatment of inflammatory diseases.


Subject(s)
Caspases, Initiator/metabolism , Monocytes/metabolism , Neoplasm Proteins/metabolism , Pyroptosis , Cell Line , Humans , Protein Domains
8.
Stem Cell Res Ther ; 9(1): 111, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29673401

ABSTRACT

The original article [1] mistakenly omitted a source of funding, and the authors would like to rectify this by acknowledging the additional support of the Natural Science Foundation in Jiangsu Province (BK20150687).

9.
Stem Cell Res Ther ; 9(1): 47, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482657

ABSTRACT

BACKGROUND: Changes in metabolic pathway preferences are key events in the reprogramming process of somatic cells to induced pluripotent stem cells (iPSCs). The optimization of metabolic conditions can enhance reprogramming; however, the detailed underlying mechanisms are largely unclear. By comparing the gene expression profiles of somatic cells, intermediate-phase cells, and iPSCs, we found that carnitine palmitoyltransferase (Cpt)1b, a rate-limiting enzyme in fatty acid oxidation, was significantly upregulated in the early stage of the reprogramming process. METHODS: Mouse embryonic fibroblasts isolated from transgenic mice carrying doxycycline (Dox)-inducible Yamanaka factor constructs were used for reprogramming. Various fatty acid oxidation-related metabolites were added during the reprogramming process. Colony counting and fluorescence-activated cell sorting (FACS) were used to calculate reprogramming efficiency. Fatty acid oxidation-related metabolites were measured by liquid chromatography-mass spectrometry. Seahorse was used to measure the level of oxidative phosphorylation. RESULTS: We found that overexpression of cpt1b enhanced reprogramming efficiency. Furthermore, palmitoylcarnitine or acetyl-CoA, the primary and final products of Cpt1-mediated fatty acid oxidation, also promoted reprogramming. In the early reprogramming process, fatty acid oxidation upregulated oxidative phosphorylation and downregulated protein kinase C activity. Inhibition of protein kinase C also promoted reprogramming. CONCLUSION: We demonstrated that fatty acid oxidation promotes reprogramming by enhancing oxidative phosphorylation and inhibiting protein kinase C activity in the early stage of the reprogramming process. This study reveals that fatty acid oxidation is crucial for the reprogramming efficiency.


Subject(s)
Cellular Reprogramming , Embryo, Mammalian/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Oxidative Phosphorylation , Protein Kinase C/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Mice , Oxidation-Reduction , Protein Kinase C/antagonists & inhibitors
10.
Cell Cycle ; 15(11): 1450-61, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27096707

ABSTRACT

Studies using in vitro cultured oocytes have indicated that the protein phosphatase 2A (PP2A), a major serine/threonine protein phosphatase, participates in multiple steps of meiosis. Details of oocyte maturation regulation by PP2A remain unclear and an in vivo model can provide more convincing information. Here, we inactivated PP2A by mutating genes encoding for its catalytic subunits (PP2Acs) in mouse oocytes. We found that eliminating both PP2Acs caused female infertility. Oocytes lacking PP2Acs failed to complete 1(st) meiotic division due to chromosome misalignment and abnormal spindle assembly. In mitosis, PP2A counteracts Aurora kinase B/C (AurkB/C) to facilitate correct kinetochore-microtubule (KT-MT) attachment. In meiosis I in oocyte, we found that PP2Ac deficiency destabilized KT-MT attachments. Chemical inhibition of AurkB/C in PP2Ac-null oocytes partly restored the formation of lateral/merotelic KT-MT attachments but not correct KT-MT attachments. Taken together, our findings demonstrate that PP2Acs are essential for chromosome alignments and regulate the formation of correct KT-MT attachments in meiosis I in oocytes.


Subject(s)
Kinetochores/metabolism , Meiosis , Microtubules/metabolism , Oocytes/metabolism , Protein Phosphatase 2/genetics , Protein Subunits/genetics , Amino Acid Sequence , Animals , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Aurora Kinase C/genetics , Aurora Kinase C/metabolism , Chromosomes, Mammalian/metabolism , Chromosomes, Mammalian/ultrastructure , Female , Gene Expression Regulation, Developmental , Infertility, Female/genetics , Infertility, Female/metabolism , Kinetochores/ultrastructure , Mice , Mice, Transgenic , Microtubules/ultrastructure , Mitosis , Oocytes/ultrastructure , Primary Cell Culture , Protein Phosphatase 2/metabolism , Protein Subunits/metabolism , Signal Transduction , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL