Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Clin Res Hepatol Gastroenterol ; 47(10): 102230, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37931846

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD), which represents the most common cause of liver disease, is emerging as a major health problem around the world. However, the molecular events that underline the pathogenesis and the progression of MASLD remain to be fully elucidated. Advanced stages of MASLD is strongly associated with liver-related outcomes and overall mortality. Despite this, highly accurate, sensitive, and non-invasive diagnostic tools are currently not aviailable, yet no FDA approved drugs for MASLD. The advance of proteomics has enable the study of protein expression, post-translational modifications (PTMs), subcellular distribution, and interactions. In this review, we discuss insights gained from the recent proteomics studies that shed new light on the pathogenesis, diagnosis and potential theraputic targets of MASLD.


Subject(s)
Fatty Liver , Metabolic Diseases , Humans , Proteomics , Biomarkers
4.
Cell Metab ; 35(5): 742-757.e10, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37040763

ABSTRACT

Nonalcoholic steatohepatitis (NASH) prevalence is rising with no pharmacotherapy approved. A major hurdle in NASH drug development is the poor translatability of preclinical studies to safe/effective clinical outcomes, and recent failures highlight a need to identify new targetable pathways. Dysregulated glycine metabolism has emerged as a causative factor and therapeutic target in NASH. Here, we report that the tripeptide DT-109 (Gly-Gly-Leu) dose-dependently attenuates steatohepatitis and fibrosis in mice. To enhance the probability of successful translation, we developed a nonhuman primate model that histologically and transcriptionally mimics human NASH. Applying a multiomics approach combining transcriptomics, proteomics, metabolomics, and metagenomics, we found that DT-109 reverses hepatic steatosis and prevents fibrosis progression in nonhuman primates, not only by stimulating fatty acid degradation and glutathione formation, as found in mice, but also by modulating microbial bile acid metabolism. Our studies describe a highly translatable NASH model and highlight the need for clinical evaluation of DT-109.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Liver/metabolism , Fibrosis , Lipid Metabolism , Primates
5.
Hepatology ; 78(6): 1763-1776, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-36939197

ABSTRACT

BACKGROUND AND AIMS: Parathyroid hormone receptor-1 (PTH1R) is a class B G protein-coupled receptor central to skeletal development, bone turnover, and calcium homeostasis. However, the role of PTH1R signaling in liver fibrosis is largely unknown. Here, the role of PTH1R signaling in the activation of HSCs and hepatic fibrosis was examined. APPROACH AND RESULTS: PTH1R was highly expressed in activated HSCs and fibrotic liver by using human liver specimens or carbon tetrachloride (CCl 4 )-treated or methionine and choline-deficient diet (MCD)-fed C57/BL6 mice. The mRNA level of hepatic PTH1R was positively correlated to α-smooth muscle actin in patients with liver cirrhosis. Mice with HSCs-specific PTH1R deletion were protected from CCl 4 , MCD, or western diet, plus low-dose CCl 4 -induced liver fibrosis. Conversely, parathyroid hormone (PTH) aggravated liver fibrosis in CCl 4 -treated mice. Mouse primary HSCs and LX2 cell lines were used for in vitro experiments. Molecular analyses by luciferase reporter assays and chromatin immunoprecipitation assays in combination with mRNA sequencing in HSCs revealed that cAMP response element-binding protein-like 2 (Crebl2), a novel regulator in HSCs treated by PTH that interacted with mothers against decapentaplegic homolog 3 (SMAD3) and increased the transcription of TGFß in activating HSCs and collagen deposition. In agreement, HSCs-specific Crebl2 deletion ameliorated PTH-induced liver fibrosis in CCl 4 -treated mice. CONCLUSIONS: In both mouse and human models, we found that PTH1R was highly expressed in activated HSCs and fibrotic liver. PTH1R signaling regulated collagen production in the HSCs through Crebl2/SMAD3/TGFß regulatory circuits. Blockade of PTH1R signaling in HSCs might help mitigate the development of liver fibrosis.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Receptor, Parathyroid Hormone, Type 1 , Humans , Mice , Animals , Receptor, Parathyroid Hormone, Type 1/metabolism , Liver Cirrhosis/metabolism , Collagen , Transforming Growth Factor beta , RNA, Messenger
6.
Front Endocrinol (Lausanne) ; 14: 1161402, 2023.
Article in English | MEDLINE | ID: mdl-36967758

ABSTRACT

Osteoporosis is a common complication of many types of chronic liver diseases (CLDs), such as cholestatic liver disease, viral hepatitis, and alcoholic liver disease. Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent metabolic liver disease, affecting almost one third of adults around the world, and is emerging as the dominant cause of CLDs. Liver serves as a hub for nutrient and energy metabolism in the body, and its crosstalk with other tissues, such as adipose tissue, heart, and brain, has been well recognized. However, much less is known about the crosstalk that occurs between the liver and bone. Moreover, the mechanisms by which CLDs increase the risk for osteoporosis remain unclear. This review summarizes the latest research on the liver-bone axis and discusses the relationship between NAFLD and osteoporosis. We cover key signaling molecules secreted by liver, such as insulin-like growth factor-1 (IGF-1), fibroblast growth factor 21 (FGF21), insulin-like growth factor binding protein 1 (IGFBP1), fetuin-A, tumor necrosis factor-alpha (TNF-α), and osteopontin (OPN), and their relevance to the homeostasis of bone metabolism. Finally, we consider the disordered liver metabolism that occurs in patients with NAFLD and how this disrupts signaling to the bone, thereby perturbing the balance between osteoclasts and osteoblasts and leading to osteoporosis or hepatic osteodystrophy (HOD).


Subject(s)
Bone Diseases, Metabolic , Non-alcoholic Fatty Liver Disease , Osteoporosis , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Bone and Bones/metabolism , Osteoporosis/metabolism
7.
EMBO Mol Med ; 15(4): e17450, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36847712

ABSTRACT

Premature ovarian insufficiency (POI) is a disease featured by early menopause before 40 years of age, accompanied by an elevation of follicle-stimulating hormone. Though POI affects many aspects of women's health, its major causes remain unknown. Many clinical studies have shown that POI patients are generally underweight, indicating a potential correlation between POI and metabolic disorders. To understand the pathogenesis of POI, we performed metabolomics analysis on serum and identified branch-chain amino acid (BCAA) insufficiency-related metabolic disorders in two independent cohorts from two clinics. A low BCAA diet phenotypically reproduced the metabolic, endocrine, ovarian, and reproductive changes of POI in young C57BL/6J mice. A mechanism study revealed that the BCAA insufficiency-induced POI is associated with abnormal activation of the ceramide-reactive oxygen species (ROS) axis and consequent impairment of ovarian granulosa cell function. Significantly, the dietary supplement of BCAA prevented the development of ROS-induced POI in female mice. The results of this pathogenic study will lead to the development of specific therapies for POI.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Humans , Female , Mice , Animals , Reactive Oxygen Species , Amino Acids , Mice, Inbred C57BL , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/therapy
9.
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
10.
Front Genet ; 13: 872518, 2022.
Article in English | MEDLINE | ID: mdl-35559030

ABSTRACT

Background and aims: As a major cause of liver disease worldwide, non-alcoholic fatty liver disease (NAFLD) comprises non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH). Due to the high prevalence and poor prognosis of NASH, it is critical to understand its mechanisms. However, the etiology and mechanisms remain largely unknown. In addition, the gold standard for the diagnosis of NASH is liver biopsy, which is an invasive procedure. Therefore, there is a pressing need to develop noninvasive tests for NASH diagnosis. The goal of the study is to discover key genes involved in NASH development and investigate their value as noninvasive biomarkers. Methods: The Gene Expression Omnibus (GEO) database was used to obtain two datasets encompassing NASH patients and healthy controls. We used weighted gene co-expression network analysis (WGCNA) and differential expression analysis in order to investigate the association between gene sets and clinical features, as well as to discover co-expression modules. A protein-protein interaction (PPI) network was created to extract hub genes. The results were validated using another publicly available dataset and mice treated with a high-fat diet (HFD) and carbon tetrachloride (CCl4). Results: A total of 24 differentially co-expressed genes were selected by WGCNA and differential expression analysis. KEGG analysis indicated most of them were enriched in the focal adhesion pathway. GO analysis showed these genes were mainly enriched in circadian rhythm, aging, angiogenesis and response to drug (biological process), endoplasmic reticulum lumen (cellular component), and protein binding (molecular function). As a result, eight genes (JUN, SERPINE1, GINS2, TYMS, HMMR, IGFBP2, BIRC3, TNFRSF12A) were identified as hub genes. Finally, three genes were found significantly changed in both the validation dataset and the mouse model. Conclusion: Our research discovered genes that have the potential to mediate the process of NASH and might be useful diagnostic biomarkers for the disorder.

11.
ACS Appl Mater Interfaces ; 13(15): 17998-18009, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33821608

ABSTRACT

The separation of radioactive noble gases, such as Xe and Kr, has attracted special attention in the context of used nuclear fuel (UNF). In this study, 180 metal-organic frameworks (MOFs) formally used for selective adsorptions of ethane and ethylene, with a similar kinetic diameter to Kr and Xe, were initially screened for the Kr/Xe separation using the grand canonical Monte Carlo (GCMC) method. Then, the structure-adsorption property relationships were generalized, that is, the MOFs of higher Kr/Xe selectivity are with the porosity at 0.2-0.4 and the ratio of the largest cavity diameter/pore limiting diameter at 1.0-2.4. Based on the relationships, six reported MOFs with large Kr uptakes and Kr/Xe selectivities were experimentally screened out and validated by GCMC simulations within the CoRE-MOF database, which are higher than most reported MOFs under conditions pertinent to nuclear fuel reprocessing of an 80/20 v/v mixture of Kr/Xe at normal temperature and pressure. Further simulations reveal that higher Kr uptakes and Kr/Xe selectivities of six MOFs result from the confinement effect of the pores. Molecular dynamic simulations showed that the six MOFs are ideal membrane separation materials of Kr from Xe, which are driven by adsorption and diffusion. Analyses of electronic structure-based density functional theory calculations showed that the main interaction between Kr and the six MOFs is van der Waals force dominated by dispersion and induction interactions. Therefore, the generalized structure-adsorption property relationships may assist the screening of MOFs for the separation and production of Kr/Xe from UNF industrially.

12.
J Hepatol ; 75(1): 150-162, 2021 07.
Article in English | MEDLINE | ID: mdl-33548387

ABSTRACT

BACKGROUND & AIMS: Chronic endoplasmic reticulum (ER) stress in the liver has been shown to play a causative role in non-alcoholic fatty liver disease (NAFLD) progression, yet the underlying molecular mechanisms remain to be elucidated. Forkhead box A3 (FOXA3), a member of the FOX family, plays critical roles in metabolic homeostasis, although its possible functions in ER stress and fatty liver progression are unknown. METHODS: Adenoviral delivery, siRNA delivery, and genetic knockout mice were used to crease FOXA3 gain- or loss-of-function models. Tunicamycin (TM) and a high-fat diet (HFD) were used to induce acute or chronic ER stress in mice. Chromatin immunoprecipiation (ChIP)-seq, luciferase assay, and adenoviral-mediated downstream gene manipulations were performed to reveal the transcriptional axis involved. Key axis protein levels in livers from healthy donors and patients with NAFLD were assessed via immunohistochemical staining. RESULTS: FOXA3 transcription is specifically induced by XBP1s upon ER stress. FOXA3 exacerbates the excessive lipid accumulation caused by the acute ER-inducer TM, whereas FOXA3 deficiency in hepatocytes and mice alleviates it. Importantly, FOXA3 deficiency in mice reduced diet-induced chronic ER stress, fatty liver, and insulin resistance. In addition, FOXA3 suppression via siRNA or adeno-associated virus delivery ameliorated the fatty liver phenotype in HFD-fed and db/db mice. Mechanistically, ChIP-Seq analysis revealed that FOXA3 directly regulates Period1 (Per1) transcription, which in turn promotes the expression of lipogenic genes, including Srebp1c, thus enhancing lipid synthesis. Of pathophysiological significance, FOXA3, PER1, and SREBP1c levels were increased in livers of obese mice and patients with NAFLD. CONCLUSION: The present study identified FOXA3 as the bridging molecule that links ER stress and NAFLD progression. Our results highlighted the role of the XBP1s-FOXA3-PER1/Srebp1c transcriptional axis in the development of NAFLD and identified FOXA3 as a potential therapeutic target for fatty liver disease. LAY SUMMARY: The molecular mechanisms linking endoplasmic reticulum stress to non-alcoholic fatty liver disease (NAFLD) progression remain undefined. Herein, via in vitro and in vivo analysis, we identified Forkhead box A3 (FOXA3) as a key bridging molecule. Of pathophysiological significance, FOXA3 protein levels were increased in livers of obese mice and patients with NAFLD, indicating that FOXA3 could be a potential therapeutic target in fatty liver disease.


Subject(s)
Endoplasmic Reticulum Stress , Hepatocyte Nuclear Factor 3-gamma/metabolism , Animals , Drug Discovery , Hepatocytes/metabolism , Humans , Lipogenesis/genetics , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Period Circadian Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , X-Box Binding Protein 1/metabolism
13.
J Clin Hypertens (Greenwich) ; 22(8): 1389-1395, 2020 08.
Article in English | MEDLINE | ID: mdl-32687255

ABSTRACT

Although many studies explored the association between helicobacter pylori (H pylori) infection and hypertension, there is no consensus. This study is to investigate the association between H pylori infection and the prevalence of hypertension among a middle- and old-age Chinese population. A cross-sectional study including 17,100 participants from the Dongfeng-Tongji cohort study was performed. All participants underwent a 14 C-urea breath test and a routine health check-up. Logistics and linear regression with multivariable adjustment were used to quest the association between H pylori infection and hypertension. The individuals with H pylori infection had a higher prevalence of hypertension (57.5% vs 55.1%, P = .002), and infection rate of H pylori in patients with hypertension is higher than that in non-hypertensive individuals (48.8% vs 46.4%, P = .002). After adjustment for potential confounders, H pylori infection increased the prevalence of hypertension (odds ratio, 1.117, 95% confidence interval (CI), 1.029-1.213, P = .008). Moreover, compared with participants without H pylori infection, individuals infected had an increase of 0.905 mm Hg (95% CI, 0.025-1.785, P = .044) for diastolic blood pressure. However, there was no interaction between H pylori infection and traditional risk factors on hypertension. These findings suggested that H pylori infection was positively associated with the prevalence of hypertension.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Hypertension , Adult , China/epidemiology , Cohort Studies , Cross-Sectional Studies , Helicobacter Infections/epidemiology , Humans , Hypertension/epidemiology , Prevalence , Risk Factors
14.
J Clin Invest ; 130(7): 3791-3804, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32510471

ABSTRACT

Type 2 diabetes mellitus (T2DM) has become an expanding global public health problem. Although the glucocorticoid receptor (GR) is an important regulator of glucose metabolism, the relationship between circulating glucocorticoids (GCs) and the features of T2DM remains controversial. Here, we show that 17-hydroxyprogesterone (17-OHP), an intermediate steroid in the biosynthetic pathway that converts cholesterol to cortisol, binds to and stimulates the transcriptional activity of GR. Hepatic 17-OHP concentrations are increased in diabetic mice and patients due to aberrantly increased expression of Cyp17A1. Systemic administration of 17-OHP or overexpression of Cyp17A1 in the livers of lean mice promoted the pathogenesis of hyperglycemia and insulin resistance, whereas knockdown of Cyp17A1 abrogated metabolic disorders in obese mice. Therefore, our results identify a Cyp17A1/17-OHP/GR-dependent pathway in the liver that mediates obesity-induced hyperglycemia, suggesting that selectively targeting hepatic Cyp17A1 may provide a therapeutic avenue for treating T2DM.


Subject(s)
17-alpha-Hydroxyprogesterone/blood , Hyperglycemia/blood , Liver/metabolism , Obesity/blood , Receptors, Glucocorticoid/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Female , Humans , Hyperglycemia/drug therapy , Male , Mice , Steroid 17-alpha-Hydroxylase/metabolism
15.
Gastroenterology ; 158(8): 2266-2281.e27, 2020 06.
Article in English | MEDLINE | ID: mdl-32105727

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic fatty liver disease is characterized by excessive hepatic accumulation of triglycerides. We aimed to identify metabolites that differ in plasma of patients with liver steatosis vs healthy individuals (controls) and investigate the mechanisms by which these might contribute to fatty liver in mice. METHODS: We obtained blood samples from 15 patients with liver steatosis and 15 controls from a single center in China (discovery cohort). We performed untargeted liquid chromatography with mass spectrometry analysis of plasma to identify analytes associated with liver steatosis. We then performed targeted metabolomic analysis of blood samples from 2 independent cohorts of individuals who underwent annual health examinations in China (1157 subjects with or without diabetes and 767 subjects with or without liver steatosis; replication cohorts). We performed mass spectrometry analysis of plasma from C57BL/6J mice, germ-free, and mice given antibiotics. C57BL/6J mice were given 0.325% (m/v) N,N,N-trimethyl-5-aminovaleric acid (TMAVA) in their drinking water and placed on a 45% high-fat diet (HFD) for 2 months. Plasma, liver tissues, and fecal samples were collected; fecal samples were analyzed by 16S ribosomal RNA gene sequencing. C57BL/6J mice with CRISPR-mediated disruption of the gene encoding γ-butyrobetaine hydroxylase (BBOX-knockout mice) were also placed on a 45% HFD for 2 months. Hepatic fatty acid oxidation (FAO) in liver tissues was determined by measuring liberation of 3H2O from [3H] palmitic acid. Liver tissues were analyzed by electron microscopy, to view mitochondria, and proteomic analyses. We used surface plasmon resonance analysis to quantify the affinity of TMAVA for BBOX. RESULTS: Levels of TMAVA, believed to be a metabolite of intestinal microbes, were increased in plasma from subjects with liver steatosis compared with controls, in the discovery and replication cohorts. In 1 replication cohort, the odds ratio for fatty liver in subjects with increased liver plasma levels of TMAVA was 1.82 (95% confidence interval [CI], 1.14-2.90; P = .012). Plasma from mice given antibiotics or germ-free mice had significant reductions in TMAVA compared with control mice. We found the intestinal bacteria Enterococcus faecalis and Pseudomonas aeruginosa to metabolize trimethyllysine to TMAVA; levels of trimethyllysine were significantly higher in plasma from patients with steatosis than controls. We found TMAVA to bind and inhibit BBOX, reducing synthesis of carnitine. Mice given TMAVA had alterations in their fecal microbiomes and reduced cold tolerance; their plasma and liver tissue had significant reductions in levels of carnitine and acyl-carnitine and their hepatocytes had reduced mitochondrial FAO compared with mice given only an HFD. Mice given TMAVA on an HFD developed liver steatosis, which was reduced by carnitine supplementation. BBOX-knockout mice had carnitine deficiency and decreased FAO, increasing uptake and liver accumulation of free fatty acids and exacerbating HFD-induced fatty liver. CONCLUSIONS: Levels of TMAVA are increased in plasma from subjects with liver steatosis. In mice, intestinal microbes metabolize trimethyllysine to TMAVA, which reduces carnitine synthesis and FAO to promote steatosis.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Valerates/metabolism , gamma-Butyrobetaine Dioxygenase/antagonists & inhibitors , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Diet, High-Fat , Dysbiosis , Fatty Acids, Nonesterified/metabolism , Feces/microbiology , Female , Humans , Lipolysis/drug effects , Liver/enzymology , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Oxidation-Reduction , Up-Regulation , Valerates/blood , Valerates/toxicity , Young Adult , gamma-Butyrobetaine Dioxygenase/genetics , gamma-Butyrobetaine Dioxygenase/metabolism
17.
Hepatology ; 71(4): 1228-1246, 2020 04.
Article in English | MEDLINE | ID: mdl-31469911

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic steatohepatitis (NASH) is a progressive liver disease that is characterized by liver injury, inflammation, and fibrosis. NASH pathogenesis is linked to reprogramming of chromatin landscape in the liver that predisposes hepatocytes to stress-induced tissue injury. However, the molecular nature of the putative checkpoint that maintains chromatin architecture and preserves hepatocyte health remains elusive. APPROACH AND RESULTS: Here we show that heterogeneous nuclear ribonucleoprotein U (hnRNPU), a nuclear matrix protein that governs chromatin architecture and gene transcription, is a critical factor that couples chromatin disruption to NASH pathogenesis. RNA-seq and chromatin immunoprecipitation-seq studies revealed an extensive overlap between hnRNPU occupancy and altered gene expression during NASH. Hepatocyte-specific inactivation of hnRNPU disrupted liver chromatin accessibility, activated molecular signature of NASH, and sensitized mice to diet-induced NASH pathogenesis. Mechanistically, hnRNPU deficiency stimulated the expression of a truncated isoform of TrkB (TRKB-T1) that promotes inflammatory signaling in hepatocytes and stress-induced cell death. Brain-derived neurotrophic factor treatment reduced membrane TRKB-T1 protein and protected mice from diet-induced NASH. CONCLUSIONS: These findings illustrate a mechanism through which disruptions of chromatin architecture drive the emergence of disease-specific signaling patterns that promote liver injury and exacerbate NASH pathogenesis.


Subject(s)
Chromatin Assembly and Disassembly , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Membrane Glycoproteins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Brain-Derived Neurotrophic Factor/therapeutic use , Disease Models, Animal , Hepatocytes/metabolism , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/therapy , Protein-Tyrosine Kinases/genetics , Transcriptome
18.
Proc Natl Acad Sci U S A ; 116(43): 21732-21738, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31594848

ABSTRACT

Endoplasmic reticulum (ER) stress plays an important role in metabolic diseases like obesity and type 2 diabetes mellitus (T2DM), although the underlying mechanisms and regulatory pathways remain to be elucidated. Here, we induced chronic low-grade ER stress in lean mice to levels similar to those in high-fat diet (HFD)-fed obese mice and found that it promoted hyperglycemia due to enhanced hepatic gluconeogenesis. Mechanistically, sustained ER stress up-regulated the deubiquitinating enzyme ubiquitin-specific peptidase 14 (USP14), which increased the stability and levels of 3',5'-cyclic monophosphate-responsive element binding (CREB) protein (CBP) to enhance glucagon action and hepatic gluconeogenesis. Exogenous overexpression of USP14 in the liver significantly increased hepatic glucose output. Consistent with this, liver-specific knockdown of USP14 abrogated the effects of ER stress on glucose metabolism, and also improved hyperglycemia and glucose intolerance in obese mice. In conclusion, our findings show a mechanism underlying ER stress-induced disruption of glucose homeostasis, and present USP14 as a potential therapeutic target against T2DM.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Glucagon/metabolism , Hyperglycemia/pathology , Obesity/pathology , Ubiquitin Thiolesterase/metabolism , Animals , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Endoplasmic Reticulum/pathology , Gene Knockdown Techniques , Gluconeogenesis/physiology , Glucose/metabolism , Glucose Intolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Obese , Ubiquitin Thiolesterase/genetics
19.
Nat Metab ; 1(2): 251-260, 2019 02.
Article in English | MEDLINE | ID: mdl-31535079

ABSTRACT

Thermogenesis is an important contributor to whole body energy expenditure and metabolic homeostasis. Although circulating factors that promote energy expenditure are known, endocrine molecules that suppress energy expenditure have remained largely elusive. Here we show that Tsukushi (TSK) is a liver-enriched secreted factor that is highly inducible in response to increased energy expenditure. Hepatic Tsk expression and plasma TSK levels are elevated in obesity. TSK deficiency increases sympathetic innervation and norepinephrine release in adipose tissue, leading to enhanced adrenergic signaling and thermogenesis, attenuation of brown fat whitening and protection from diet-induced obesity in mice. Our work reveals TSK as part of a negative feedback mechanism that gates thermogenic energy expenditure and highlights TSK as a potential target for therapeutic intervention in metabolic disease.


Subject(s)
Adipose Tissue, Brown/metabolism , Energy Metabolism/physiology , Liver/metabolism , Proteoglycans/metabolism , Animals , Mice , Obesity/metabolism , Thermogenesis/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL