Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 16(738): eadk1866, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478630

ABSTRACT

Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as nonalcoholic steatohepatitis (NASH), is an advanced stage of metabolic fatty liver disease. The pathogenic mechanisms of MASH center on hepatocyte injury and the ensuing immune response within the liver microenvironment. Recent work has implicated TREM2+ macrophages in various disease conditions, and substantial induction of TREM2+ NASH-associated macrophages (NAMs) serves as a hallmark of metabolic liver disease. Despite this, the mechanisms through which NAMs contribute to MASH pathogenesis remain poorly understood. Here, we identify membrane-spanning 4-domains a7 (MS4A7) as a NAM-specific pathogenic factor that exacerbates MASH progression in mice. Hepatic MS4A7 expression was strongly induced in mouse and human MASH and associated with the severity of liver injury. Whole-body and myeloid-specific ablation of Ms4a7 alleviated diet-induced MASH pathologies in male mice. We demonstrate that exposure to lipid droplets (LDs), released upon injury of steatotic hepatocytes, triggered NAM induction and exacerbated MASH-associated liver injury in an MS4A7-dependent manner. Mechanistically, MS4A7 drove NLRP3 inflammasome activation via direct physical interaction and shaped disease-associated cell states within the liver microenvironment. This work reveals the LD-MS4A7-NLRP3 inflammasome axis as a pathogenic driver of MASH progression and provides insights into the role of TREM2+ macrophages in disease pathogenesis.


Subject(s)
Inflammasomes , Non-alcoholic Fatty Liver Disease , Animals , Humans , Male , Mice , Inflammasomes/metabolism , Liver/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Immunologic/metabolism
2.
Plants (Basel) ; 13(5)2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38475435

ABSTRACT

Excessive soil salinity is a major stressor inhibiting crops' growth, development, and yield. Seed germination is a critical stage of crop growth and development, as well as one of the most salt-sensitive stages. Salt stress has a significant inhibitory effect on seed germination. Okra is a nutritious vegetable, but its seed germination percentage (GP) is low, whether under salt stress conditions or suitable conditions. In this study, we used 180 okra accessions and conducted a genome-wide association study (GWAS) on the germination percentage using 20,133,859 single nucleotide polymorphic (SNP) markers under 0 (CK, diluted water), 70 (treatment 1, T1), and 140 mmol/L (treatment 2, T2) NaCl conditions. Using the mixed linear model (MLM) in Efficient Mixed-model Association eXpedated (EMMAX) and Genome-wide Efficient Mixed Model Association (GEMMA) software, 511 SNP loci were significantly associated during germination, of which 167 SNP loci were detected simultaneously by both programs. Among the 167 SNPs, SNP2619493 on chromosome 59 and SNP2692266 on chromosome 44 were detected simultaneously under the CK, T1, and T2 conditions, and were key SNP loci regulating the GP of okra seeds. Linkage disequilibrium block analysis revealed that nsSNP2626294 (C/T) in Ae59G004900 was near SNP2619493, and the amino acid changes caused by nsSNP2626294 led to an increase in the phenotypic values in some okra accessions. There was an nsSNP2688406 (A/G) in Ae44G005470 near SNP2692266, and the amino acid change caused by nsSNP2688406 led to a decrease in phenotypic values in some okra accessions. These results indicate that Ae59G004900 and Ae44G005470 regulate the GP of okra seeds under salt and no-salt stresses. The gene expression analysis further demonstrated these results. The SNP markers and genes that were identified in this study will provide reference for further research on the GP of okra, as well as new genetic markers and candidate genes for cultivating new okra varieties with high GPs under salt and no-salt stress conditions.

3.
Hepatology ; 79(2): 409-424, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37505219

ABSTRACT

BACKGROUND AND AIMS: NASH represents a severe stage of fatty liver disease characterized by hepatocyte injury, inflammation, and liver fibrosis. Myeloid-derived innate immune cells, such as macrophages and dendritic cells, play an important role in host defense and disease pathogenesis. Despite this, the nature of transcriptomic reprogramming of myeloid cells in NASH liver and its contribution to disease progression remain incompletely defined. APPROACH AND RESULTS: In this study, we performed bulk and single-cell RNA sequencing (sc-RNA seq) analysis to delineate the landscape of macrophage and dendritic cell transcriptomes in healthy and NASH livers. Our analysis uncovered cell type-specific patterns of transcriptomic reprogramming on diet-induced NASH. We identified brain-abundant membrane-attached signal protein 1 (Basp1) as a myeloid-enriched gene that is markedly induced in mouse and human NASH liver. Myeloid-specific inactivation of Basp1 attenuates the severity of diet-induced NASH pathologies, as shown by reduced hepatocyte injury and liver fibrosis in mice. Mechanistically, cultured macrophages lacking Basp1 exhibited a diminished response to pro-inflammatory stimuli, impaired NLRP3 inflammasome activation, and reduced cytokine secretion. CONCLUSIONS: Together, these findings uncover Basp1 as a critical regulator of myeloid inflammatory signaling that underlies NASH pathogenesis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/pathology , Liver/pathology , Hepatocytes/metabolism , Diet , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Disease Models, Animal
4.
Hepatology ; 78(5): 1478-1491, 2023 11 01.
Article in English | MEDLINE | ID: mdl-35950514

ABSTRACT

BACKGROUND AND AIMS: The mammalian liver harbors heterogeneous cell types that communicate via local paracrine signaling. Recent studies have delineated the transcriptomic landscape of the liver in NASH that provides insights into liver cell heterogeneity, intercellular crosstalk, and disease-associated reprogramming. However, the nature of intrahepatic signaling and its role in NASH progression remain obscure. APPROACH AND RESULTS: Here, we performed transcriptomic analyses and identified cardiotrophin-like cytokine factor 1 (CLCF1), a member of the IL-6 family cytokines, as a cholangiocyte-derived paracrine factor that was elevated in the liver from diet-induced NASH mice and patients with NASH. Adenovirus-associated virus-mediated overexpression of CLCF1 in the liver ameliorated NASH pathologies in two diet-induced NASH models in mice, illustrating that CLCF1 induction may serve an adaptive and protective role during NASH pathogenesis. Unexpectedly, messenger RNA and protein levels of leukemia inhibitory factor receptor (LIFR), a subunit of the receptor complex for CLCF1, were markedly downregulated in NASH liver. Hepatocyte-specific inactivation of LIFR accelerated NASH progression in mice, supporting an important role of intrahepatic cytokine signaling in maintaining tissue homeostasis under metabolic stress conditions. CONCLUSIONS: Together, this study sheds light on the molecular nature of intrahepatic paracrine signaling during NASH pathogenesis and uncovers potential targets for therapeutic intervention.


Subject(s)
Non-alcoholic Fatty Liver Disease , Paracrine Communication , Animals , Humans , Mice , Cytokines/genetics , Cytokines/metabolism , Diet/adverse effects , Disease Models, Animal , Interleukins/metabolism , Liver/metabolism , Mammals , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Paracrine Communication/genetics , Paracrine Communication/physiology
5.
Cell Metab ; 34(9): 1359-1376.e7, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35973424

ABSTRACT

The mammalian liver comprises heterogeneous cell types within its tissue microenvironment that undergo pathophysiological reprogramming in disease states, such as non-alcoholic steatohepatitis (NASH). Patients with NASH are at an increased risk for the development of hepatocellular carcinoma (HCC). However, the molecular and cellular nature of liver microenvironment remodeling that links NASH to liver carcinogenesis remains obscure. Here, we show that diet-induced NASH is characterized by the induction of tumor-associated macrophage (TAM)-like macrophages and exhaustion of cytotoxic CD8+ T cells in the liver. The adipocyte-derived endocrine factor Neuregulin 4 (NRG4) serves as a hormonal checkpoint that restrains this pathological reprogramming during NASH. NRG4 deficiency exacerbated the induction of tumor-prone liver immune microenvironment and NASH-related HCC, whereas transgenic NRG4 overexpression elicited protective effects in mice. In a therapeutic setting, recombinant NRG4-Fc fusion protein exhibited remarkable potency in suppressing HCC and prolonged survival in the treated mice. These findings pave the way for therapeutic intervention of liver cancer by targeting the NRG4 hormonal checkpoint.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Neuregulins/metabolism , Non-alcoholic Fatty Liver Disease , Animals , Carcinoma, Hepatocellular/metabolism , Liver/metabolism , Liver Neoplasms/drug therapy , Mammals/metabolism , Mice , Neuregulins/therapeutic use , Non-alcoholic Fatty Liver Disease/metabolism , Tumor Microenvironment
6.
Food Sci Nutr ; 9(7): 3470-3482, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34262707

ABSTRACT

Dietary fiber is regarded to improve host metabolic disorders through modulating gut microbiota. The study was to investigate the effects of inulin with different degree of polymerization (DP) on adiposity, related metabolic syndrome, and the possible mechanisms from the points of gut microbiota and metabolite changes. C57Bl/6J male mice were randomly allocated to normal diet (ND) group, high-fat diet (HFD) group, two HFD groups with short-chain inulin (HFD-S) and medium and long-chain inulin (HFD-ML) for 8 weeks. Compared with HFD treatment, ML-inulin supplementation significantly decreased weight gain, hepatic steatosis, chronic inflammation, and increased insulin sensitivity, energy expenditure and thermogenesis. This could be mimicked by S-inulin supplementation to some degree although it is not as effective as ML inulin. Also, mice treated with S and ML inulin had a remarkable alternation in the composition of gut microbiota and increased the production of short-chain fatty acids (SCFAs). However, reduced serum levels of essential fatty acids, vitamins B1 and B3 by HFD were further decreased by both inulin supplementations. ML inulin can prevent HFD-induced obesity and the associated metabolic disorders, and may be used as novel gut microbiota modulator to prevent HFD-induced gut dysbiosis and metabolic disorders.

7.
Sci Adv ; 6(20): eaay6191, 2020 05.
Article in English | MEDLINE | ID: mdl-32426492

ABSTRACT

Depletion of fat-resident regulatory T cells (Tregs) and group 2 innate lymphoid cells (ILC2s) has been causally linked to obesity-associated insulin resistance. However, the molecular nature of the pathogenic signals suppress adipose Tregs and ILC2s in obesity remains unknown. Here, we identified the soluble isoform of interleukin (IL)-33 receptor ST2 (sST2) as an obesity-induced adipokine that attenuates IL-33 signaling and disrupts Treg/ILC2 homeostasis in adipose tissue, thereby exacerbates obesity-associated insulin resistance in mice. We demonstrated sST2 is a target of TNFα signaling in adipocytes that is countered by Zbtb7b. Fat-specific ablation of Zbtb7b augments adipose sST2 gene expression, leading to diminished fat-resident Tregs/ILC2s, more pronounced adipose tissue inflammation and fibrosis, and impaired glucose homeostasis in mice. Mechanistically, Zbtb7b suppresses NF-κB activation in response to TNFα through destabilizing IκBα. These findings uncover an adipokine-immune signaling pathway that is engaged in obesity to drive the pathological changes of the immunometabolic landscape.


Subject(s)
Insulin Resistance , Adipokines/metabolism , Adipose Tissue/metabolism , Animals , DNA-Binding Proteins/metabolism , Immunity, Innate , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Cell Biol ; 219(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31653673

ABSTRACT

Lipid droplets (LDs) are evolutionarily conserved organelles that play important roles in cellular metabolism. Each LD is enclosed by a monolayer of phospholipids, distinct from bilayer membranes. During LD biogenesis and growth, this monolayer of lipids expands by acquiring phospholipids from the endoplasmic reticulum (ER) through nonvesicular mechanisms. Here, in a mini-screen, we find that ORP5, an integral membrane protein of the ER, can localize to ER-LD contact sites upon oleate loading. ORP5 interacts with LDs through its ligand-binding domain, and ORP5 deficiency enhances neutral lipid synthesis and increases the size of LDs. Importantly, there is significantly more phosphatidylinositol-4-phosphate (PI(4)P) and less phosphatidylserine (PS) on LDs in ORP5-deficient cells than in normal cells. The increased presence of PI(4)P on LDs in ORP5-deficient cells requires phosphatidylinositol 4-kinase 2-α. Our results thus demonstrate the existence of PI(4)P on LDs and suggest that LD-associated PI(4)P may be primarily used by ORP5 to deliver PS to LDs.


Subject(s)
Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Minor Histocompatibility Antigens/metabolism , Phosphatidylinositol Phosphates/metabolism , Phospholipid Transfer Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Steroid/metabolism , HEK293 Cells , Humans , Lipid Metabolism
9.
Cell Rep ; 28(13): 3406-3422.e7, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31553910

ABSTRACT

Insulin-stimulated hepatic glycogen synthesis is central to glucose homeostasis. Here, we show that PPP1R3G, a regulatory subunit of protein phosphatase 1 (PP1), is directly phosphorylated by AKT. PPP1R3G phosphorylation fluctuates with fasting-refeeding cycle and is required for insulin-stimulated dephosphorylation, i.e., activation of glycogen synthase (GS) in hepatocytes. In this study, we demonstrate that knockdown of PPP1R3G significantly inhibits insulin response. The introduction of wild-type PPP1R3G, and not phosphorylation-defective mutants, increases hepatic glycogen deposition, blood glucose clearance, and insulin sensitivity in vivo. Mechanistically, phosphorylated PPP1R3G displays increased binding for, and promotes dephosphorylation of, phospho-GS. Furthermore, PPP1R3B, another regulatory subunit of PP1, binds to the dephosphorylated GS, thereby relaying insulin stimulation to hepatic glycogen deposition. Importantly, this PP1-mediated signaling cascade is independent of GSK3. Therefore, we reveal a regulatory axis consisting of insulin/AKT/PPP1R3G/PPP1R3B that operates in parallel to the GSK3-dependent pathway, controlling glycogen synthesis and glucose homeostasis in insulin signaling.


Subject(s)
Insulin/metabolism , Liver Glycogen/metabolism , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Humans , Signal Transduction
10.
Mol Cell ; 75(3): 644-660.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398325

ABSTRACT

Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.


Subject(s)
Cell Communication/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Sequence Analysis, RNA , Animals , Cellular Reprogramming/genetics , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligands , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/genetics , Single-Cell Analysis
11.
EMBO J ; 38(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30858281

ABSTRACT

SREBPs are master regulators of lipid homeostasis and undergo sterol-regulated export from ER to Golgi apparatus for processing and activation via COPII-coated vesicles. While COPII recognizes SREBP through its escort protein SCAP, factor(s) specifically promoting SREBP/SCAP loading to the COPII machinery remains unknown. Here, we show that the ER/lipid droplet-associated protein Cideb selectively promotes the loading of SREBP/SCAP into COPII vesicles. Sterol deprivation releases SCAP from Insig and enhances ER export of SREBP/SCAP by inducing SCAP-Cideb interaction, thereby modulating sterol sensitivity. Moreover, Cideb binds to the guanine nucleotide exchange factor Sec12 to enrich SCAP/SREBP at ER exit sites, where assembling of COPII complex initiates. Loss of Cideb inhibits the cargo loading of SREBP/SCAP, reduces SREBP activation, and alleviates diet-induced hepatic steatosis. Our data point to a linchpin role of Cideb in regulated ER export of SREBP and lipid homeostasis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/physiology , Endoplasmic Reticulum/physiology , Golgi Apparatus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterols/pharmacology , Animals , Apoptosis Regulatory Proteins/genetics , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/physiology , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , HEK293 Cells , Hep G2 Cells , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Transport , Sterol Regulatory Element Binding Protein 1/genetics
12.
PLoS Biol ; 17(1): e2006571, 2019 01.
Article in English | MEDLINE | ID: mdl-30653498

ABSTRACT

Beiging of white adipose tissue (WAT) is a particularly appealing target for therapeutics in the treatment of metabolic diseases through norepinephrine (NE)-mediated signaling pathways. Although previous studies report NE clearance mechanisms via SLC6A2 on sympathetic neurons or proinflammatory macrophages in adipose tissues (ATs), the low catecholamine clearance capacity of SLC6A2 may limit the cleaning efficiency. Here, we report that mouse organic cation transporter 3 (Oct3; Slc22a3) is highly expressed in WAT and displays the greatest uptake rate of NE as a selective non-neural route of NE clearance in white adipocytes, which differs from other known routes such as adjacent neurons or macrophages. We further show that adipocytes express high levels of NE degradation enzymes Maoa, Maob, and Comt, providing the molecular basis on NE clearance by adipocytes together with its reuptake transporter Oct3. Under NE administration, ablation of Oct3 induces higher body temperature, thermogenesis, and lipolysis compared with littermate controls. After prolonged cold challenge, inguinal WAT (ingWAT) in adipose-specific Oct3-deficient mice shows much stronger browning characteristics and significantly elevated expression of thermogenic and mitochondrial biogenesis genes than in littermate controls, and this response involves enhanced ß-adrenergic receptor (ß-AR)/protein kinase A (PKA)/cyclic adenosine monophosphate (cAMP)-responsive element binding protein (Creb) pathway activation. Glycolytic genes are reprogrammed to significantly higher levels to compensate for the loss of ATP production in adipose-specific Oct3 knockout (KO) mice, indicating the fundamental role of glucose metabolism during beiging. Inhibition of ß-AR largely abolishes the higher lipolytic and thermogenic activities in Oct3-deficient ingWAT, indicating the NE overload in the vicinity of adipocytes in Oct3 KO adipocytes. Of note, reduced functional alleles in human OCT3 are also identified to be associated with increased basal metabolic rate (BMR). Collectively, our results demonstrate that Oct3 governs ß-AR activity as a NE recycling transporter in white adipocytes, offering potential therapeutic applications for metabolic disorders.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Catecholamines/metabolism , Octamer Transcription Factor-3/metabolism , Organic Cation Transport Proteins/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Metabolism , HEK293 Cells , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Obesity/metabolism , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Organic Cation Transport Proteins/biosynthesis , Organic Cation Transport Proteins/genetics , Signal Transduction , Thermogenesis/physiology
13.
Diabetes ; 67(10): 1935-1948, 2018 10.
Article in English | MEDLINE | ID: mdl-29986925

ABSTRACT

Metabolic homeostasis is maintained by an interplay among tissues, organs, intracellular organelles, and molecules. Cidea and Cidec are lipid droplet (LD)-associated proteins that promote lipid storage in brown adipose tissue (BAT) and white adipose tissue (WAT). Using ob/ob/Cidea-/- , ob/ob/Cidec-/- , and ob/ob/Cidea-/-/Cidec-/- mouse models and CIDE-deficient cells, we studied metabolic regulation during severe obesity to identify ways to maintain metabolic homeostasis and promote antiobesity effects. The phenotype of ob/ob/Cidea-/- mice was similar to that of ob/ob mice in terms of serum parameters, adipose tissues, lipid storage, and gene expression. Typical lipodystrophy accompanied by insulin resistance occurred in ob/ob/Cidec-/- mice, with ectopic storage of lipids in the BAT and liver. Interestingly, double deficiency of Cidea and Cidec activated both WAT and BAT to consume more energy and to increase insulin sensitivity compared with their behavior in the other three mouse models. Increased lipolysis, which occurred on the LD surfaces and released fatty acids, led to activated ß-oxidation and oxidative phosphorylation in peroxisomes and mitochondria in CIDE-deficient adipocytes. The coordination among LDs, peroxisomes, and mitochondria was regulated by adipocyte triglyceride lipase (ATGL)-peroxisome proliferator-activated receptor α (PPARα). Double deficiency of Cidea and Cidec activated energy consumption in both WAT and BAT, which provided new insights into therapeutic approaches for obesity and diabetes.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Lipid Droplets/chemistry , PPAR alpha/metabolism , Peroxisomes/metabolism , Proteins/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Energy Metabolism/physiology , Lipase/genetics , Lipase/metabolism , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidative Phosphorylation , PPAR alpha/genetics , Proteins/genetics
14.
Nat Commun ; 9(1): 1916, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29765047

ABSTRACT

Obesity is characterized by excessive fatty acid conversion to triacylglycerols (TAGs) in adipose tissues. However, how signaling networks sense fatty acids and connect to the stimulation of lipid synthesis remains elusive. Here, we show that homozygous knock-in mice carrying a point mutation at the Ser86 phosphorylation site of acetyltransferase Tip60 (Tip60 SA/SA ) display remarkably reduced body fat mass, and Tip60 SA/SA females fail to nurture pups to adulthood due to severely reduced milk TAGs. Mechanistically, fatty acids stimulate Tip60-dependent acetylation and endoplasmic reticulum translocation of phosphatidic acid phosphatase lipin 1 to generate diacylglycerol for TAG synthesis, which is repressed by deacetylase Sirt1. Inhibition of Tip60 activity strongly blocks fatty acid-induced TAG synthesis while Sirt1 suppression leads to increased adiposity. Genetic analysis of loss-of-function mutants in Saccharomyces cerevisiae reveals a requirement of ESA1, yeast ortholog of Tip60, in TAG accumulation. These findings uncover a conserved mechanism linking fatty acid sensing to fat synthesis.


Subject(s)
Endoplasmic Reticulum/enzymology , Lysine Acetyltransferase 5/metabolism , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Trans-Activators/metabolism , Triglycerides/biosynthesis , Acetylation , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Fatty Acids/metabolism , Female , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Kinetics , Lysine Acetyltransferase 5/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Phosphatidate Phosphatase/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Trans-Activators/genetics , Triglycerides/chemistry
15.
Lipids Health Dis ; 17(1): 34, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29482628

ABSTRACT

BACKGROUND: Several mutations in leucine rich repeat kinase 2 (LRRK2) gene have been associated with pathogenesis of Parkinson's disease (PD), a neurodegenerative disorder marked by resting tremors, and rigidity, leading to Postural instability. It has been revealed that mutations that lead to an increase of kinase activity of LRRK2 protein are significantly associated with PD pathogenesis. Recent studies have shown that some Rab GTPases, especially Rab8, serve as substrates of LRRK2 and undergo phosphorylation in its switch II domain upon interaction. Current study was performed in order to find out the effects of the phosphorylation of Rab8 and its mutants on lipid metabolism and lipid droplets growth. METHODS: The phosphorylation status of Rab8a was checked by phos-tag gel. Point mutant construct were generated to investigate the function of Rab8a. 3T3L1 cells were transfected with indicated plasmids and the lipid droplets were stained with Bodipy. Fluorescent microscopy experiments were performed to examine the sizes of lipid droplets. The interactions between Rab8a and Optineurin were determined by immunoprecipitation and western blot. RESULTS: Our assays demonstrated that Rab8a was phosphorylated by mutated LRRK2 that exhibits high kinase activity. Phosphorylation of Rab8a on amino acid residue T72 promoted the formation of large lipid droplets. T72D mutant of Rab8a had higher activity to promote the formation of large lipid droplets compared with wild type Rab8a, with increase in average diameter of lipid droplets from 2.10 µm to 2.46 µm. Moreover, phosphorylation of Rab8a weakened the interaction with its effector Optineurin. CONCLUSIONS: Y1699C mutated LRRK2 was able to phosphorylate Rab8a and phosphorylation of Rab8a on site 72 plays important role in the fusion and enlargement of lipid droplets. Taken together, our study suggests an indirect relationship between enhanced lipid storage capacity and PD pathogenesis.


Subject(s)
Lipid Metabolism , Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism , 3T3-L1 Cells , Animals , Cell Cycle Proteins , Humans , Lipid Droplets/metabolism , Membrane Proteins/genetics , Membrane Transport Proteins , Mice , Mutation , Parkinson Disease/metabolism , Transcription Factor TFIIIA/metabolism , Tyrosine/genetics , Tyrosine/metabolism , rab GTP-Binding Proteins/genetics
16.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(10 Pt B): 1197-1204, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28648584

ABSTRACT

Cell death-inducing DFF45-like effector (CIDE) family proteins including Cidea, Cideb and Cidec/Fsp27 are expressed in many different tissues and are known as lipid droplet (LD)-and ER-associated proteins. Systematic analyses using genetically modified animal models have demonstrated that CIDE proteins play important roles in regulating various aspects of lipid homeostasis, including lipid storage, lipolysis and lipid secretion. Recent research in ours and other laboratories has revealed that CIDE proteins are crucial regulators of LD fusion and growth in the adipose tissue, liver, skin and mammary glands. CIDE-mediated LD fusion and growth is different from other membrane fusions in that it requires CIDE proteins to be enriched and clustered at the LD-LD contact sites (LDCS). The enriched CIDE proteins then allow the recruitment of other proteins to the LDCS and the formation of potential fusion pores. Neutral lipids in the smaller LDs of the contacted pair are transferred to the larger LDs, owing to the internal pressure difference, thus resulting in the fusion and growth of the LDs. This review summarizes the physiological roles of CIDE proteins in controlling lipid homeostasis, insulin sensitivity and the development of metabolic diseases including obesity, diabetes and fatty liver, with a particular focus on the role of CIDE proteins in controlling LD fusion and growth. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Membrane Fusion , Animals , Apoptosis Regulatory Proteins/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Insulin Resistance , Lipid Droplets/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology
17.
J Clin Invest ; 127(4): 1353-1369, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28287402

ABSTRACT

Obesity is characterized by aberrant fat accumulation. However, the intracellular signaling pathway that senses dietary fat and leads to fat storage remains elusive. Here, we have observed that the levels of histone deacetylase 6 (HDAC6) and the related family member HDAC10 are markedly reduced in adipose tissues of obese animals and humans. Mice with adipocyte-specific depletion of Hdac6 exhibited increased fat accumulation and reduced insulin sensitivity. In normal adipocytes, we found that reversal of P300/CBP-associated factor-induced (PCAF-induced) acetylation at K56 on cell death-inducing DFFA-like effector C (CIDEC, also known as FSP27) critically regulated lipid droplet fusion and lipid storage. Importantly, HDAC6 deacetylates CIDEC, leading to destabilization and reduced lipid droplet fusion. Accordingly, we observed elevated levels of CIDEC and its acetylated form in HDAC-deficient adipocytes as well as the adipose tissue of obese animals and humans. Fatty acids (FAs) prevented CIDEC deacetylation by promoting the dissociation of CIDEC from HDAC6, which resulted in increased association of CIDEC with PCAF on the endoplasmic reticulum. Control of CIDEC acetylation required the conversion of FAs to triacylglycerols. Thus, we have revealed a signaling axis that is involved in the coordination of nutrient availability, protein acetylation, and cellular lipid metabolic responses.


Subject(s)
Histone Deacetylases/physiology , Lipid Metabolism , Protein Processing, Post-Translational , Proteins/metabolism , 3T3-L1 Cells , Acetylation , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Endoplasmic Reticulum/metabolism , Fatty Acids/physiology , HEK293 Cells , Histone Deacetylase 6 , Humans , Lipid Droplets/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/pathology , Protein Stability , Triglycerides/biosynthesis , p300-CBP Transcription Factors/metabolism
18.
Sci Rep ; 6: 36083, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786292

ABSTRACT

The fibroblast cell line of 3T3-L1 was used as a cell model for screening and evaluating the feasibility of probiotic components in improving animal lipid metabolisms. The extracts from 12 Lactobacillus strains caused significantly reduced triacylglycerol (TAG) accumulation but with severe inflammation induction in 3T3-L1 adipocytes. Interestingly, exopolysaccharides (EPS) from LGG (Lactobacillus rhamnosus GG) significantly decreased the TAG accumulation without any inflammation. The anti-obesity effect of EPS was confirmed in high-fat-diets feeding mice. Fat pads of mice injected with EPS (50 mg/kg) every two days for two weeks were significantly reduced with much smaller adipocytes, compared with the counterparts. The levels of TAG and cholesterol ester in liver, as well as serum TAG, were decreased in EPS injected mice. In addition, down-regulated inflammation was observed in adipose tissue and liver. Interestingly, the expression of TLR2 in adipose tissue and 3T3-L1 cells was significantly increased by EPS addition. Moreover, the reverse of TAG accumulation in TLR2 knockdown 3T3-L1 in the presence of EPS confirmed that the inhibition effect of EPS on adipogenesis was mediated by TLR2. EPS from LGG has the potential for therapeutic development to intervene lipid metabolic disorders in mammals.


Subject(s)
Adipogenesis/drug effects , Fibroblasts/drug effects , Lacticaseibacillus rhamnosus/chemistry , Obesity/prevention & control , Polysaccharides, Bacterial/administration & dosage , Probiotics/administration & dosage , Toll-Like Receptor 2/metabolism , Animals , Cells, Cultured , Cholesterol/analysis , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Fibroblasts/metabolism , Gene Knockdown Techniques , Inflammation/pathology , Liver/pathology , Mice , Polysaccharides, Bacterial/isolation & purification , Polysaccharides, Bacterial/metabolism , Probiotics/isolation & purification , Probiotics/metabolism , Toll-Like Receptor 2/deficiency , Triglycerides/analysis
19.
IUBMB Life ; 68(11): 847-853, 2016 11.
Article in English | MEDLINE | ID: mdl-27650434

ABSTRACT

Metabolism refers to a chain of chemical reactions converting food/fuel into energy to conduct cellular processes, including the synthesis of the building blocks of the body, such as proteins, lipids, nucleic acids, and carbohydrates, and the elimination of nitrogenous wastes. Metabolic chain reactions are catalyzed by various enzymes that are orchestrated in specific pathways. Metabolic pathways are important for organisms to grow and reproduce, maintain their structures, and respond to their environments. The coordinated regulation of metabolic pathways is important for maintaining metabolic homeostasis. The key steps and crucial enzymes in these pathways have been well investigated. However, the crucial regulatory factors and feedback (or feedforward) mechanisms of nutrients and intermediate metabolites of these biochemical processes remain to be fully elucidated. In addition, the roles of these enzymes and regulatory factors in controlling metabolism under physiological and pathological conditions are largely unknown. In particular, metabolic dysregulation is closely linked to the development of many diseases, including obesity, fatty liver, diabetes, cancer, cardiovascular, cerebrovascular, and neurodegenerative diseases. Therefore, metabolism, an old area of biochemistry, has attracted much attention in the last decade. With substantially increased government funding, the involvement of talented researchers, an improved infrastructure and scientific environment over the last ten years, the basic research in the field of metabolism in China has dramatically advanced. Here, we have summarized the major discoveries of scientists in China in the last decade in the area of metabolism. Due to the vast amount of information, we focused this review on specific aspects of metabolism, particularly metabolic regulation and lipid metabolism in vertebrates. © 2016 IUBMB Life, 68(11):847-853, 2016.


Subject(s)
Biomedical Research/standards , Metabolic Diseases/metabolism , Animals , China , Humans , Lipid Metabolism , Metabolic Diseases/therapy , Metabolic Networks and Pathways , Quality Improvement
20.
J Biol Chem ; 291(9): 4282-93, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26733203

ABSTRACT

Lipid droplets (LDs) are dynamic subcellular organelles whose growth is closely linked to obesity and hepatic steatosis. Cell death-inducing DNA fragmentation factor-α-like effector (CIDE) proteins, including Cidea, Cideb, and Cidec (also called Fsp27), play important roles in lipid metabolism. Cidea and Cidec are LD-associated proteins that promote atypical LD fusion in adipocytes. Here, we find that CIDE proteins are all localized to LD-LD contact sites (LDCSs) and promote lipid transfer, LD fusion, and growth in hepatocytes. We have identified two types of hepatocytes, one with small LDs (small LD-containing hepatocytes, SLHs) and one with large LDs (large LD-containing hepatocytes, LLHs) in the liver. Cideb is localized to LDCSs and promotes lipid exchange and LD fusion in both SLHs and LLHs, whereas Cidea and Cidec are specifically localized to the LDCSs and promote lipid exchange and LD fusion in LLHs. Cideb-deficient SLHs have reduced LD sizes and lower lipid exchange activities. Fasting dramatically induces the expression of Cidea/Cidec and increases the percentage of LLHs in the liver. The majority of the hepatocytes from the liver of obese mice are Cidea/Cidec-positive LLHs. Knocking down Cidea or Cidec significantly reduced lipid storage in the livers of obese animals. Our data reveal that CIDE proteins play differential roles in promoting LD fusion and lipid storage; Cideb promotes lipid storage under normal diet conditions, whereas Cidea and Cidec are responsible for liver steatosis under fasting and obese conditions.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Fatty Liver/etiology , Hepatocytes/metabolism , Lipid Droplets/pathology , Obesity/pathology , Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Cell Line , Cells, Cultured , Food Deprivation , Hepatocytes/cytology , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Lipid Droplets/ultrastructure , Membrane Fusion , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Mice, Obese , Obesity/metabolism , Obesity/physiopathology , Organelle Biogenesis , Organelle Size , Perilipin-2 , Protein Transport , Proteins/antagonists & inhibitors , Proteins/genetics , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...