Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Clin Invest ; 134(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38488009

ABSTRACT

Uncontrolled accumulation of extracellular matrix leads to tissue fibrosis and loss of organ function. We previously demonstrated in vitro that the DNA/RNA-binding protein fused in sarcoma (FUS) promotes fibrotic responses by translocating to the nucleus, where it initiates collagen gene transcription. However, it is still not known whether FUS is profibrotic in vivo and whether preventing its nuclear translocation might inhibit development of fibrosis following injury. We now demonstrate that levels of nuclear FUS are significantly increased in mouse models of kidney and liver fibrosis. To evaluate the direct role of FUS nuclear translocation in fibrosis, we used mice that carry a mutation in the FUS nuclear localization sequence (FUSR521G) and the cell-penetrating peptide CP-FUS-NLS that we previously showed inhibits FUS nuclear translocation in vitro. We provide evidence that FUSR521G mice or CP-FUS-NLS-treated mice showed reduced nuclear FUS and fibrosis following injury. Finally, differential gene expression analysis and immunohistochemistry of tissues from individuals with focal segmental glomerulosclerosis or nonalcoholic steatohepatitis revealed significant upregulation of FUS and/or collagen genes and FUS protein nuclear localization in diseased organs. These results demonstrate that injury-induced nuclear translocation of FUS contributes to fibrosis and highlight CP-FUS-NLS as a promising therapeutic option for organ fibrosis.


Subject(s)
Amyotrophic Lateral Sclerosis , RNA , Animals , Mice , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mutation , DNA , Fibrosis , Collagen/metabolism , Amyotrophic Lateral Sclerosis/genetics
2.
Hepatology ; 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38373106

ABSTRACT

BACKGROUND AND AIMS: Sustained inflammation and hepatocyte injury in chronic liver disease activate HSCs to transdifferentiate into fibrogenic, contractile myofibroblasts. We investigated the role of protocadherin 7 (PCDH7), a cadherin family member not previously characterized in the liver, whose expression is restricted to HSCs. APPROACH AND RESULTS: We created a PCDH7 fl/fl mouse line, which was crossed to lecithin retinol acyltransferase-Cre mice to generate HSC-specific PCDH7 knockout animals. HSC contraction in vivo was tested in response to the HSC-selective vasoconstrictor endothelin-1 using intravital multiphoton microscopy. To establish a PCDH7 null HSC line, cells were isolated from PCDH7 fl/fl mice and infected with adenovirus-expressing Cre. Hepatic expression of PCDH7 was strictly restricted to HSCs. Knockout of PCDH7 in vivo abrogated HSC-mediated sinusoidal contraction in response to endothelin-1. In cultured HSCs, loss of PCDH7 markedly attenuated contractility within collagen gels and led to altered gene expression in pathways governing adhesion and vasoregulation. Loss of contractility in PCDH7 knockout cells was impaired Rho-GTPase signaling, as demonstrated by altered gene expression, reduced assembly of F-actin fibers, and loss of focal adhesions. CONCLUSIONS: The stellate cell-specific cadherin, PCDH7, is a novel regulator of HSC contractility whose loss leads to cytoskeletal remodeling and sinusoidal relaxation.

3.
Cancer Immunol Res ; 11(12): 1630-1641, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37844197

ABSTRACT

T cells recognize several types of antigens in tumors, including aberrantly expressed, nonmutated proteins, which are therefore shared with normal tissue and referred to as self/shared-antigens (SSA), and mutated proteins or oncogenic viral proteins, which are referred to as tumor-specific antigens (TSA). Immunotherapies such as immune checkpoint blockade (ICB) can activate T-cell responses against TSA, leading to tumor control, and also against SSA, causing immune-related adverse events (irAE). To improve anti-TSA immunity while limiting anti-SSA autoreactivity, we need to understand how tumor-specific CD8+ T cells (TST) and SSA-specific CD8+ T (SST) cells differentiate in response to cognate antigens during tumorigenesis. Therefore, we developed a genetic cancer mouse model in which we can track TST and SST differentiation longitudinally as liver cancers develop. We found that both TST and SST lost effector function over time, but while TST persisted long term and had a dysfunctional/exhausted phenotype (including expression of PD1, CD39, and TOX), SST exited cell cycle prematurely and disappeared from liver lesions. However, SST persisted in spleens in a dysfunctional TCF1+PD-1- state: unable to produce effector cytokines or proliferate in response to ICB targeting PD-1 or PD-L1. Thus, our studies identify a dysfunctional T-cell state occupied by T cells reactive to SSA: a TCF1+PD-1- state lacking in effector function, demonstrating that the type/specificity of tumor antigen may determine tumor-reactive T-cell differentiation.


Subject(s)
Liver Neoplasms , Programmed Cell Death 1 Receptor , Animals , Mice , CD8-Positive T-Lymphocytes , Cytokines/metabolism , Lymphocyte Activation , Antigens
4.
Sci Signal ; 16(787): eadf6696, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37253090

ABSTRACT

Organ size is maintained by the controlled proliferation of distinct cell populations. In the mouse liver, hepatocytes in the midlobular zone that are positive for cyclin D1 (CCND1) repopulate the parenchyma at a constant rate to preserve liver mass. Here, we investigated how hepatocyte proliferation is supported by hepatic stellate cells (HSCs), pericytes that are in close proximity to hepatocytes. We used T cells to ablate nearly all HSCs in the murine liver, enabling the unbiased characterization of HSC functions. In the normal liver, complete loss of HSCs persisted for up to 10 weeks and caused a gradual reduction in liver mass and in the number of CCND1+ hepatocytes. We identified neurotrophin-3 (Ntf-3) as an HSC-produced factor that induced the proliferation of midlobular hepatocytes through the activation of tropomyosin receptor kinase B (TrkB). Treating HSC-depleted mice with Ntf-3 restored CCND1+ hepatocytes in the midlobular region and increased liver mass. These findings establish that HSCs form the mitogenic niche for midlobular hepatocytes and identify Ntf-3 as a hepatocyte growth factor.


Subject(s)
Hepatic Stellate Cells , Liver , Neurotrophin 3 , Animals , Mice , Cell Proliferation , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver/metabolism , Neurotrophin 3/metabolism
5.
Hepatology ; 78(3): 820-834, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36988383

ABSTRACT

BACKGROUND AND AIMS: The scarcity of suitable donor livers highlights a continuing need for innovation to recover organs with reversible injuries in liver transplantation. APPROACH AND RESULTS: Explanted human donor livers (n = 5) declined for transplantation were supported using xenogeneic cross-circulation of whole blood between livers and xeno-support swine. Livers and swine were assessed over 24 hours of xeno-support. Livers maintained normal global appearance, uniform perfusion, and preservation of histologic and subcellular architecture. Oxygen consumption increased by 75% ( p = 0.16). Lactate clearance increased from -0.4 ± 15.5% to 31.4 ± 19.0% ( p = 0.02). Blinded histopathologic assessment demonstrated improved injury scores at 24 hours compared with 12 hours. Vascular integrity and vasoconstrictive function were preserved. Bile volume and cholangiocellular viability markers improved for all livers. Biliary structural integrity was maintained. CONCLUSIONS: Xenogeneic cross-circulation provided multisystem physiological regulation of ex vivo human livers that enabled functional rehabilitation, histopathologic recovery, and improvement of viability markers. We envision xenogeneic cross-circulation as a complementary technique to other organ-preservation technologies in the recovery of marginal donor livers or as a research tool in the development of advanced bioengineering and pharmacologic strategies for organ recovery and rehabilitation.


Subject(s)
Liver Transplantation , Liver , Humans , Swine , Animals , Liver/pathology , Liver Transplantation/methods , Bile , Perfusion/methods , Organ Preservation/methods
6.
J Intern Med ; 291(1): 11-31, 2022 01.
Article in English | MEDLINE | ID: mdl-34564899

ABSTRACT

Non-alcoholic fatty liver disease is comprised of either simple steatosis (non-alcoholic fatty liver) or a more advanced inflammatory and fibrogenic stage (non-alcoholic steatohepatitis [NASH]). NASH affects a growing proportion of the global adult and pediatric population, leading to rising rates of liver fibrosis and hepatocellular carcinoma. NASH is a multifactorial disease that is part of a systemic metabolic disorder. Here, we provide an overview of the metabolic underpinnings of NASH pathogenesis and established drivers of inflammation and fibrosis. Clarification of underlying fibrogenic and inflammatory mechanisms will advance the development of novel treatment strategies as there are no approved therapies at present. We discuss emerging experimental approaches and potential novel investigational strategies derived from animal models including the inflammasome, epigenetic reprogramming, Hippo signaling, Notch signaling, engineered T cells to remove fibrogenic HSCs, and HSC-specific targeting therapies. Recently completed and ongoing clinical trials and antifibrotics are discussed, illuminating the growing expectation that one or more therapies will yield clinical benefit in NASH in the coming years.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Child , Fibrosis , Hippo Signaling Pathway , Humans , Liver/pathology , Liver Cirrhosis/therapy , Non-alcoholic Fatty Liver Disease/therapy
7.
Nat Cell Biol ; 23(12): 1240-1254, 2021 12.
Article in English | MEDLINE | ID: mdl-34887515

ABSTRACT

Extracellular vesicles and exomere nanoparticles are under intense investigation as sources of clinically relevant cargo. Here we report the discovery of a distinct extracellular nanoparticle, termed supermere. Supermeres are morphologically distinct from exomeres and display a markedly greater uptake in vivo compared with small extracellular vesicles and exomeres. The protein and RNA composition of supermeres differs from small extracellular vesicles and exomeres. Supermeres are highly enriched with cargo involved in multiple cancers (glycolytic enzymes, TGFBI, miR-1246, MET, GPC1 and AGO2), Alzheimer's disease (APP) and cardiovascular disease (ACE2, ACE and PCSK9). The majority of extracellular RNA is associated with supermeres rather than small extracellular vesicles and exomeres. Cancer-derived supermeres increase lactate secretion, transfer cetuximab resistance and decrease hepatic lipids and glycogen in vivo. This study identifies a distinct functional nanoparticle replete with potential circulating biomarkers and therapeutic targets for a host of human diseases.


Subject(s)
Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Nanoparticles/metabolism , Alzheimer Disease/pathology , Angiotensin-Converting Enzyme 2/metabolism , Biological Transport/physiology , Biomarkers/metabolism , COVID-19/pathology , Cardiovascular Diseases/pathology , Cell Communication/physiology , Cell Line, Tumor , HeLa Cells , Humans , Lactic Acid/metabolism , MicroRNAs/genetics , Nanoparticles/classification , Neoplasms/pathology , Tumor Microenvironment
9.
J Hepatol ; 75(4): 865-878, 2021 10.
Article in English | MEDLINE | ID: mdl-33992698

ABSTRACT

BACKGROUND AND AIMS: Non-alcoholic steatohepatitis (NASH)-related hepatocellular carcinoma (HCC) is increasing globally, but its molecular features are not well defined. We aimed to identify unique molecular traits characterising NASH-HCC compared to other HCC aetiologies. METHODS: We collected 80 NASH-HCC and 125 NASH samples from 5 institutions. Expression array (n = 53 NASH-HCC; n = 74 NASH) and whole exome sequencing (n = 52 NASH-HCC) data were compared to HCCs of other aetiologies (n = 184). Three NASH-HCC mouse models were analysed by RNA-seq/expression-array (n = 20). Activin A receptor type 2A (ACVR2A) was silenced in HCC cells and proliferation assessed by colorimetric and colony formation assays. RESULTS: Mutational profiling of NASH-HCC tumours revealed TERT promoter (56%), CTNNB1 (28%), TP53 (18%) and ACVR2A (10%) as the most frequently mutated genes. ACVR2A mutation rates were higher in NASH-HCC than in other HCC aetiologies (10% vs. 3%, p <0.05). In vitro, ACVR2A silencing prompted a significant increase in cell proliferation in HCC cells. We identified a novel mutational signature (MutSig-NASH-HCC) significantly associated with NASH-HCC (16% vs. 2% in viral/alcohol-HCC, p = 0.03). Tumour mutational burden was higher in non-cirrhotic than in cirrhotic NASH-HCCs (1.45 vs. 0.94 mutations/megabase; p <0.0017). Compared to other aetiologies of HCC, NASH-HCCs were enriched in bile and fatty acid signalling, oxidative stress and inflammation, and presented a higher fraction of Wnt/TGF-ß proliferation subclass tumours (42% vs. 26%, p = 0.01) and a lower prevalence of the CTNNB1 subclass. Compared to other aetiologies, NASH-HCC showed a significantly higher prevalence of an immunosuppressive cancer field. In 3 murine models of NASH-HCC, key features of human NASH-HCC were preserved. CONCLUSIONS: NASH-HCCs display unique molecular features including higher rates of ACVR2A mutations and the presence of a newly identified mutational signature. LAY SUMMARY: The prevalence of hepatocellular carcinoma (HCC) associated with non-alcoholic steatohepatitis (NASH) is increasing globally, but its molecular traits are not well characterised. In this study, we uncovered higher rates of ACVR2A mutations (10%) - a potential tumour suppressor - and the presence of a novel mutational signature that characterises NASH-related HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Molecular Biology/statistics & numerical data , Non-alcoholic Fatty Liver Disease/genetics , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/etiology , Female , Humans , Liver Neoplasms/etiology , Liver Neoplasms/genetics , Male , Middle Aged , Molecular Biology/methods , Non-alcoholic Fatty Liver Disease/complications , Risk Factors
10.
Am J Pathol ; 191(2): 309-319, 2021 02.
Article in English | MEDLINE | ID: mdl-33159885

ABSTRACT

A unique and complex microstructure underlies the diverse functions of the liver. Breakdown of this organization, as occurs in fibrosis and cirrhosis, impairs liver function and leads to disease. The role of integrin ß1 was examined both in establishing liver microstructure and recreating it after injury. Embryonic deletion of integrin ß1 in the liver disrupts the normal development of hepatocyte polarity, specification of cell-cell junctions, and canalicular formation. This in turn leads to the expression of transforming growth factor ß (TGF-ß) and widespread fibrosis. Targeted deletion of integrin ß1 in adult hepatocytes prevents recreation of normal hepatocyte architecture after liver injury, with resultant fibrosis. In vitro, integrin ß1 is essential for canalicular formation and is needed to prevent stellate cell activation by modulating TGF-ß. Taken together, these findings identify integrin ß1 as a key determinant of liver architecture with a critical role as a regulator of TGF-ß secretion. These results suggest that disrupting the hepatocyte-extracellular matrix interaction is sufficient to drive fibrosis.


Subject(s)
Integrin beta1/metabolism , Liver Regeneration/physiology , Liver/metabolism , Transforming Growth Factor beta/metabolism , Animals , Extracellular Matrix/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/metabolism , Mice , Mice, Transgenic
11.
JCI Insight ; 52019 04 11.
Article in English | MEDLINE | ID: mdl-30973829

ABSTRACT

Extracellular mRNAs (ex-mRNAs) potentially supersede extracellular miRNAs (ex-miRNAs) and other RNA classes as biomarkers. We performed conventional small-RNA-sequencing (sRNA-seq) and sRNA-seq with T4 polynucleotide kinase (PNK) end-treatment of total exRNA isolated from serum and platelet-poor EDTA, ACD, and heparin plasma to study the effect on ex-mRNA capture. Compared to conventional sRNA-seq PNK-treatment increased the detection of informative ex-mRNAs reads up to 50-fold. The exRNA pool was dominated by hematopoietic cells and platelets, with additional contribution from the liver. About 60% of the 15- to 42-nt reads originated from the coding sequences, in a pattern reminiscent of ribosome-profiling. Blood sample type had a considerable influence on the exRNA profile. On average approximately 350 to 1,100 distinct ex-mRNA transcripts were detected depending on plasma type. In serum, additional transcripts from neutrophils and hematopoietic cells increased this number to near 2,300. EDTA and ACD plasma showed a destabilizing effect on ex mRNA and non-coding RNA ribonucleoprotein complexes compared to other plasma types. In a proof-of-concept study, we investigated differences between the exRNA profiles of patients with acute coronary syndrome (ACS) and healthy controls. The improved tissue resolution of ex mRNAs after PNK-treatment enabled us to detect a neutrophil-signature in ACS that escaped detection by ex miRNA analysis.


Subject(s)
Acute Coronary Syndrome/genetics , Blood Cells/metabolism , Cell-Free Nucleic Acids/genetics , Liver/metabolism , Muscle, Skeletal/metabolism , RNA-Seq/methods , Acute Coronary Syndrome/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Cell-Free Nucleic Acids/blood , Citric Acid , Edetic Acid , Erythrocytes/metabolism , Female , Glucose/analogs & derivatives , Heparin , High-Throughput Nucleotide Sequencing , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Monocytes/metabolism , Neutrophils/metabolism , Plasma , Polynucleotide 5'-Hydroxyl-Kinase , Proof of Concept Study , Sequence Analysis, RNA/methods , Serum , Specimen Handling
12.
Hepatology ; 70(6): 2107-2122, 2019 12.
Article in English | MEDLINE | ID: mdl-31016744

ABSTRACT

The growing burden of liver fibrosis and lack of effective antifibrotic therapies highlight the need for identification of pathways and complementary model systems of hepatic fibrosis. A rare, monogenic disorder in which children with mutations in mannose phosphate isomerase (MPI) develop liver fibrosis led us to explore the function of MPI and mannose metabolism in liver development and adult liver diseases. Herein, analyses of transcriptomic data from three human liver cohorts demonstrate that MPI gene expression is down-regulated proportionate to fibrosis in chronic liver diseases, including nonalcoholic fatty liver disease and hepatitis B virus. Depletion of MPI in zebrafish liver in vivo and in human hepatic stellate cell (HSC) lines in culture activates fibrotic responses, indicating that loss of MPI promotes HSC activation. We further demonstrate that mannose supplementation can attenuate HSC activation, leading to reduced fibrogenic activation in zebrafish, culture-activated HSCs, and in ethanol-activated HSCs. Conclusion: These data indicate the prospect that modulation of mannose metabolism pathways could reduce HSC activation and improve hepatic fibrosis.


Subject(s)
Hepatic Stellate Cells/physiology , Liver Cirrhosis/etiology , Mannose-6-Phosphate Isomerase/physiology , Mannose/pharmacology , Animals , Cells, Cultured , Glycosylation , Humans , Male , Platelet-Derived Growth Factor/physiology , Signal Transduction/physiology , Zebrafish
13.
Hepatology ; 69(5): 2196-2213, 2019 05.
Article in English | MEDLINE | ID: mdl-30520052

ABSTRACT

Autophagy is important for hepatic homeostasis, nutrient regeneration, and organelle quality control. We investigated the mechanisms by which liver injury occurred in the absence of autophagy function. We found that mice deficient in autophagy because of the lack of autophagy-related gene 7 or autophagy-related gene 5, key autophagy-related genes, manifested intracellular cholestasis with increased levels of serum bile acids, a higher ratio of tauromuricholic acid/taurocholic acid in the bile, increased hepatic bile acid load, abnormal bile canaliculi, and altered expression of hepatic transporters. In determining the underlying mechanism, we found that autophagy sustained and promoted the basal and up-regulated expression of farnesoid X receptor (Fxr) in the fed and starved conditions, respectively. Consequently, expression of Fxr and its downstream genes, particularly bile salt export pump, and the binding of FXR to the promoter regions of these genes, were suppressed in autophagy-deficient livers. In addition, codeletion of nuclear factor erythroid 2-related factor 2 (Nrf2) in autophagy deficiency status reversed the FXR suppression. Furthermore, the cholestatic injury of autophagy-deficient livers was reversed by enhancement of FXR activity or expression, or by Nrf2 deletion. Conclusion: Together with earlier reports that FXR can suppress autophagy, our findings indicate that autophagy and FXR form a regulatory loop and deficiency of autophagy causes abnormal FXR functionality, leading to the development of intracellular cholestasis and liver injury.


Subject(s)
Autophagy , Cholestasis, Intrahepatic/etiology , NF-E2-Related Factor 2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/blood , Cholestasis, Intrahepatic/metabolism , Female , Food Deprivation , Liver/ultrastructure , Male , Mice, Transgenic
14.
Nat Commun ; 9(1): 4962, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30470740

ABSTRACT

Activation of the Hippo pathway effector Yap underlies many liver cancers, however no germline or somatic mutations have been identified. Autophagy maintains essential metabolic functions of the liver, and autophagy-deficient murine models develop benign adenomas and hepatomegaly, which have been attributed to activation of the p62/Sqstm1-Nrf2 axis. Here, we show that Yap is an autophagy substrate and mediator of tissue remodeling and hepatocarcinogenesis independent of the p62/Sqstm1-Nrf2 axis. Hepatocyte-specific deletion of Atg7 promotes liver size, fibrosis, progenitor cell expansion, and hepatocarcinogenesis, which is rescued by concurrent deletion of Yap. Our results shed new light on mechanisms of Yap degradation and the sequence of events that follow disruption of autophagy, which is impaired in chronic liver disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Hepatocytes/cytology , Liver Neoplasms/metabolism , Liver Neoplasms/physiopathology , Liver/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Carcinogenesis , Cell Cycle Proteins , Cell Differentiation , Female , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/pathology , Liver Neoplasms/genetics , Male , Mice , Phosphoproteins/genetics , Proteolysis , Transcription Factors , YAP-Signaling Proteins
16.
J Hepatol ; 69(2): 385-395, 2018 08.
Article in English | MEDLINE | ID: mdl-29572095

ABSTRACT

BACKGROUND AND AIMS: Although the majority of patients with non-alcoholic fatty liver disease (NAFLD) have only steatosis without progression, a sizeable fraction develop non-alcoholic steatohepatitis (NASH), which can lead to cirrhosis and hepatocellular carcinoma (HCC). Many established diet-induced mouse models for NASH require 24-52 weeks, which makes testing for drug response costly and time consuming. METHODS: We have sought to establish a murine NASH model with rapid progression of extensive fibrosis and HCC by using a western diet (WD), which is high-fat, high-fructose and high-cholesterol, combined with low weekly dose of intraperitoneal carbon tetrachloride (CCl4), which serves as an accelerator. RESULTS: C57BL/6J mice were fed a normal chow diet ±â€¯CCl4 or WD ±â€¯CCl4 for 12 and 24 weeks. Addition of CCl4 exacerbated histological features of NASH, fibrosis, and tumor development induced by WD, which resulted in stage 3 fibrosis at 12 weeks and HCC development at 24 weeks. Furthermore, whole liver transcriptomic analysis indicated that dysregulated molecular pathways in WD/CCl4 mice and immunologic features were similar to those of human NASH. CONCLUSIONS: Our mouse NASH model exhibits rapid progression of advanced fibrosis and HCC, and mimics histological, immunological and transcriptomic features of human NASH, suggesting that it will be a useful experimental tool for preclinical drug testing. LAY SUMMARY: A carefully characterized model has been developed in mice that recapitulates the progressive stages of human fatty liver disease, from simple steatosis, to inflammation, fibrosis and cancer. The functional pathways of gene expression and immune abnormalities in this model closely resemble human disease. The ease and reproducibility of this model make it ideal to study disease pathogenesis and test new treatments.


Subject(s)
Diet, Western , Fatty Liver , Liver Cirrhosis , Liver Neoplasms , Mice, Inbred C57BL , Animals , Carbon Tetrachloride/pharmacology , Disease Models, Animal , Disease Progression , Fatty Liver/etiology , Fatty Liver/immunology , Fatty Liver/pathology , Gene Expression Profiling/methods , Inflammation/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Neoplasms/etiology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Reproducibility of Results
17.
Sci Rep ; 7: 42563, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256512

ABSTRACT

We have used a computational approach to identify anti-fibrotic therapies by querying a transcriptome. A transcriptome signature of activated hepatic stellate cells (HSCs), the primary collagen-secreting cell in liver, and queried against a transcriptomic database that quantifies changes in gene expression in response to 1,309 FDA-approved drugs and bioactives (CMap). The flavonoid apigenin was among 9 top-ranked compounds predicted to have anti-fibrotic activity; indeed, apigenin dose-dependently reduced collagen I in the human HSC line, TWNT-4. To identify proteins mediating apigenin's effect, we next overlapped a 122-gene signature unique to HSCs with a list of 160 genes encoding proteins that are known to interact with apigenin, which identified C1QTNF2, encoding for Complement C1q tumor necrosis factor-related protein 2, a secreted adipocytokine with metabolic effects in liver. To validate its disease relevance, C1QTNF2 expression is reduced during hepatic stellate cell activation in culture and in a mouse model of alcoholic liver injury in vivo, and its expression correlates with better clinical outcomes in patients with hepatitis C cirrhosis (n = 216), suggesting it may have a protective role in cirrhosis progression.These findings reinforce the value of computational approaches to drug discovery for hepatic fibrosis, and identify C1QTNF2 as a potential mediator of apigenin's anti-fibrotic activity.


Subject(s)
Antifibrinolytic Agents/pharmacology , Apigenin/pharmacology , Drug Discovery , Drug Repositioning , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Transcriptome , Animals , Biomarkers , Cell Line , Humans , Mice
18.
Semin Liver Dis ; 36(1): 87-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26870935

ABSTRACT

Fibrosis is the wound-healing response of tissues to injury. Extensive characterization of organ fibrosis mechanisms has identified common core pathways in renal, pulmonary, skin, and liver fibrosis that offer novel antifibrotic approaches across tissues, in addition to organ-specific and/or disease-specific pathways. A growing number of small molecules and biologics have been identified that are reaching clinical trials for one or more fibrotic diseases, making new antifibrotic options for liver fibrosis an emerging reality. The accelerating pace of drug development, which will also include drug repurposing or combination therapies, heightens the need for novel methods for noninvasive fibrosis assessment without liver biopsy, which is critical to establishing surrogate endpoints for patients in clinical trials who have a low risk of hepatic decompensation. In this article the authors review mechanisms of liver fibrosis and outline potential therapeutic targets and antifibrotic therapies in preclinical studies and clinical trials.


Subject(s)
Liver Cirrhosis/drug therapy , Liver/drug effects , Molecular Targeted Therapy , Animals , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/metabolism , Molecular Targeted Therapy/adverse effects , Signal Transduction/drug effects , Treatment Outcome
19.
Cell Metab ; 24(6): 771-772, 2016 12 13.
Article in English | MEDLINE | ID: mdl-28068221

ABSTRACT

Nonalcoholic steatohepatitis leads to cirrhosis and cancer in a rising number of patients with metabolic syndrome. In this issue of Cell Metabolism, Wang et al. (2016) identify the transcriptional co-activator Taz as a driver of inflammation and fibrosis through the induction of Indian hedgehog in hepatocytes, which stimulates fibrogenesis by hepatic stellate cells.


Subject(s)
Fatty Liver , Non-alcoholic Fatty Liver Disease , Hepatic Stellate Cells , Hepatocytes/drug effects , Humans , Inflammation , Liver Cirrhosis
20.
Gut ; 64(5): 830-41, 2015 May.
Article in English | MEDLINE | ID: mdl-25681399

ABSTRACT

Reversibility of hepatic fibrosis and cirrhosis following antiviral therapy for hepatitis B or C has advanced the prospect of developing antifibrotic therapies for patients with chronic liver diseases, especially non-alcoholic steatohepatitis. Mechanisms of fibrosis have focused on hepatic stellate cells, which become fibrogenic myofibroblasts during injury through 'activation', and are at the nexus of efforts to define novel drug targets. Recent studies have clarified pathways of stellate cell gene regulation and epigenetics, emerging pathways of fibrosis regression through the recruitment and amplification of fibrolytic macrophages, nuanced responses of discrete inflammatory cell subsets and the identification of the 'ductular reaction' as a marker of severe injury and repair. Based on our expanded knowledge of fibrosis pathogenesis, attention is now directed towards strategies for antifibrotic therapies and regulatory challenges for conducting clinical trials with these agents. New therapies are attempting to: 1) Control or cure the primary disease or reduce tissue injury; 2) Target receptor-ligand interactions and intracellular signaling; 3) Inhibit fibrogenesis; and 4) Promote resolution of fibrosis. Progress is urgently needed in validating non-invasive markers of fibrosis progression and regression that can supplant biopsy and shorten the duration of clinical trials. Both scientific and clinical challenges remain, however the past three decades of steady progress in understanding liver fibrosis have contributed to an emerging translational success story, with realistic hopes for antifibrotic therapies to treat patients with chronic liver disease in the near future.


Subject(s)
Drug Discovery/methods , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Clinical Trials as Topic/methods , Hepatic Stellate Cells/physiology , Humans , Patient Selection , Translational Research, Biomedical/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...