Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Hepatology ; 70(4): 1134-1149, 2019 10.
Article in English | MEDLINE | ID: mdl-30891779

ABSTRACT

Although mortality due to acute alcoholic hepatitis (AH) correlates with Model for End-Stage Liver Disease (MELD) scores, biomarkers are critically needed to manage this disease. Increases in inflammatory markers and macrophage activation are associated with acute AH and could be potential biomarkers of clinical events and/or mortality. We enrolled 89 clinically diagnosed AH patients in four US academic medical centers. Plasma from AH patients had a significant increase in gut microbial translocation indicators (endotoxin, bacterial 16S ribosomal DNA) and host response indicators (soluble cluster of differentiation 14 [sCD14] and lipopolysaccharide binding protein [LBP]) compared to controls. Patient MELD score and Glasgow Alcoholic Hepatitis score (GAHS) correlated with endotoxin levels. AH patients also had a significant increase in high mobility group protein 1 (HMGB1), a sterile danger signal molecule, and osteopontin (OPN), a multifunctional phosphoprotein involved in neutrophil activation, compared to controls. Increased levels of OPN positively correlated with increasing MELD score, GAHS, and LBP levels. Consistent with these results, AH patients had significantly increased circulating levels of macrophage activation (sCD163 and sCD206) markers compared to healthy controls, and sCD163 and sCD206 significantly and positively correlated with OPN, HMGB1, and LBP levels as well as with MELD score and GAHS. These findings indicate a connection between microbial translocation, immune cell activation, and AH severity. Plasma sCD14, OPN, sCD163, and sCD206 levels were significantly higher in nonsurvivors than survivors. In multivariate regression models, we identified sCD14, sCD163, and OPN as independent predictors of 90-day mortality, infection, and organ failure development, respectively. Conclusion: Our study suggests that sCD14, LBP, OPN, sCD163, and sCD206 are biomarkers to indicate severity and predict clinical outcomes in AH.


Subject(s)
Acute-Phase Proteins/metabolism , Antigens, CD/metabolism , Carrier Proteins/metabolism , Disease Progression , Hepatitis, Alcoholic/mortality , Hepatitis, Alcoholic/physiopathology , Macrophage Activation/immunology , Membrane Glycoproteins/metabolism , Academic Medical Centers , Adult , Aged , Biomarkers/blood , Case-Control Studies , Cause of Death , Enzyme-Linked Immunosorbent Assay , Hepatitis, Alcoholic/blood , Humans , Kaplan-Meier Estimate , Liver Function Tests , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Prognosis , Proportional Hazards Models , ROC Curve , Risk Assessment , Severity of Illness Index , Survival Analysis , United States
2.
Hepatology ; 69(3): 1105-1121, 2019 03.
Article in English | MEDLINE | ID: mdl-30179264

ABSTRACT

Kupffer cell and macrophage (MØ) activation contributes to steatosis, inflammation, and fibrosis in alcoholic liver disease (ALD). We found increased frequency of MØ, T cells, and expression of C-C chemokine receptor type 2 (Ccr2) and C-C chemokine receptor type 5 (Ccr5) in the livers of patients with ALD, and increased circulating chemokines, C-C chemokine ligand types 2 (CCL2), and C-C chemokine ligand types 5 (CCL5) in patients with alcoholic hepatitis. We hypothesized that inhibition of CCL2 signaling with the dual CCR2/5 inhibitor, cenicriviroc (CVC), would attenuate ALD. In a mouse model of ALD, liver injury (alanine aminotransferase [ALT]) and steatosis were prevented by CVC whether administered as "prevention" throughout the alcohol feeding or as "treatment" started after the development of ALD. Alcohol-induced increases in early liver fibrosis markers (sirius red, hydroxyproline, and collagen-1) were normalized by both modes of CVC administration. We found that prevention and treatment with CVC reversed alcohol-related increases in liver mRNA and protein expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and CCL2. CVC administration regimens prevented the increase in infiltrating MØ (F4/80lo CD11bhi ) and reduced proinflammatory Ly6Chi MØ in livers of alcohol-fed mice. CVC increased liver T-cell numbers and attenuated Il-2 expression without an effect on CD69+ or CD25+ T-cell expression. In vitro, CVC inhibited CCL2-induced increases in hepatocyte fatty acid synthase (Fasn) and adipose differentiation-related protein (Adrp), whereas it augmented acyl-coenzyme A oxidase 1 (Acox-1), proliferator-activated receptor gamma co-activator alpha (Pgc1α) and uncoupling protein 2 expression, suggesting mechanisms for attenuated hepatocyte steatosis. We found that CCL2 and CCL5 sensitized hepatocytes to lipopolysaccharide-induced liver injury (TNF-α, ALT, and lactate dehydrogenase release). Alcohol feeding induced apoptosis (poly ADP-ribose polymerase [PARP] and caspase-3 [CASP-3] cleavage) and pyroptosis (gasdermin D [GSDMD] cleavage) in livers, and CVC prevented both of these forms of cell death. Conclusion: Together, our data demonstrate preclinical evidence for CCR2/CCR5 inhibition with CVC as a potent intervention to ameliorate alcohol-induced steatohepatitis and liver damage.


Subject(s)
CCR5 Receptor Antagonists/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Receptors, CCR2/antagonists & inhibitors , Animals , Fatty Liver, Alcoholic/drug therapy , Female , Hepatitis, Alcoholic/drug therapy , Liver Cirrhosis, Alcoholic/drug therapy , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
3.
Hepatology ; 67(5): 1986-2000, 2018 05.
Article in English | MEDLINE | ID: mdl-29251792

ABSTRACT

A salient feature of alcoholic liver disease (ALD) is Kupffer cell (KC) activation and recruitment of inflammatory monocytes and macrophages (MØs). These key cellular events of ALD pathogenesis may be mediated by extracellular vesicles (EVs). EVs transfer biomaterials, including proteins and microRNAs, and have recently emerged as important effectors of intercellular communication. We hypothesized that circulating EVs from mice with ALD have a protein cargo characteristic of the disease and mediate biological effects by activating immune cells. The total number of circulating EVs was increased in mice with ALD compared to pair-fed controls. Mass spectrometric analysis of circulating EVs revealed a distinct signature for proteins involved in inflammatory responses, cellular development, and cellular movement between ALD EVs and control EVs. We also identified uniquely important proteins in ALD EVs that were not present in control EVs. When ALD EVs were injected intravenously into alcohol-naive mice, we found evidence of uptake of ALD EVs in recipient livers in hepatocytes and MØs. Hepatocytes isolated from mice after transfer of ALD EVs, but not control EVs, showed increased monocyte chemoattractant protein 1 mRNA and protein expression, suggesting a biological effect of ALD EVs. Compared to control EV recipient mice, ALD EV recipient mice had increased numbers of F4/80hi cluster of differentiation 11b (CD11b)lo KCs and increased percentages of tumor necrosis factor alpha-positive/interleukin 12/23-positive (inflammatory/M1) KCs and infiltrating monocytes (F4/80int CD11bhi ), while the percentage of CD206+ CD163+ (anti-inflammatory/M2) KCs was decreased. In vitro, ALD EVs increased tumor necrosis factor alpha and interleukin-1ß production in MØs and reduced CD163 and CD206 expression. We identified heat shock protein 90 in ALD EVs as the mediator of ALD-EV-induced MØ activation. CONCLUSION: Our study indicates a specific protein signature of ALD EVs and demonstrates a functional role of circulating EVs containing heat shock protein 90 in mediating KC/MØ activation in the liver. (Hepatology 2018;67:1986-2000).


Subject(s)
Extracellular Vesicles/metabolism , HSP90 Heat-Shock Proteins/metabolism , Liver Diseases, Alcoholic/metabolism , Macrophages/metabolism , Animals , Cytokines/metabolism , Female , Hepatocytes/metabolism , Liver/metabolism , Liver/pathology , Macrophage Activation/genetics , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL
4.
J Immunol ; 198(5): 1974-1984, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122964

ABSTRACT

Monocytes and macrophages (MΦs) play a central role in the pathogenesis of chronic hepatitis C virus (HCV) infection. The tissue microenvironment triggers monocyte differentiation into MΦs, with polarization ranging within the spectrum of M1 (classical) to M2 (alternative) activation. Recently, we demonstrated that HCV infection leads to monocyte differentiation into polarized MΦs that mediate stellate cell activation via TGF-ß. In this study, we aimed to identify the viral factor(s) that mediate monocyte-to-MΦ differentiation. We performed coculture experiments using healthy monocytes with exosome-packaged HCV, cell-free HCV, or HCV ssRNA. Coculture of monocytes with exosome-packaged HCV, cell-free HCV, or HCV ssRNA induced differentiation into MΦs with high M2 surface marker expression and production of pro- and anti-inflammatory cytokines. The HCV ssRNA-induced monocyte activation and differentiation into MΦs could be prevented by TLR7 or TLR8 knockdown. Furthermore, TLR7 or TLR8 stimulation, independent of HCV, caused monocyte differentiation and M2 MΦ polarization. In vivo, in chronic HCV-infected patients, we found increased expression of TLR7/8 in circulating monocytes that was associated with increased intracellular expression of procollagen. Furthermore, knockdown of TLR8 completely attenuated collagen expression in monocytes exposed to HCV, and knockdown of TLR7 partially attenuated this expression, suggesting roles for TLR7/8 in induction of fibrocytes in HCV infection. We identified TLR7/8 as mediators of monocyte differentiation and M2 MΦ polarization during HCV infection. Further, we demonstrated that HCV ssRNA and other TLR7/8 ligands promote MΦ polarization and generation of circulating fibrocytes.


Subject(s)
Hepacivirus/physiology , Hepatitis C, Chronic/immunology , Macrophages/physiology , Monocytes/physiology , RNA, Viral/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Cytokines/metabolism , Exosomes/immunology , Exosomes/virology , Fibroblasts/physiology , Gene Knockdown Techniques , Humans , Macrophages/virology , Monocytes/virology , Th2 Cells/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics
5.
J Hepatol ; 69(5): 1145-1154, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30030149

ABSTRACT

BACKGROUND & AIMS: Neutrophil extracellular traps (NETs) are an important strategy utilized by neutrophils to immobilize and kill invading microorganisms. Herein, we studied NET formation and the process of neutrophil cell death (NETosis), as well as the clearance of NETs by macrophages (MΦ) (efferocytosis) in acute sepsis following binge drinking. METHODS: Healthy volunteers consumed 2 ml of vodka/kg body weight, before blood endotoxin and 16 s rDNA were measured. Peripheral neutrophils were isolated and exposed to alcohol followed by phorbol 12-myristate 13-acetate (PMA) stimulation. Mice were treated with three alcohol binges and intraperitoneal lipopolysaccharide (LPS) to assess the dynamics of NET formation and efferocytosis. In vivo, anti-Ly6G antibody (IA8) was used for neutrophil depletion. RESULTS: Inducers of NETs (endotoxin and bacterial DNA) significantly increased in the circulation after binge alcohol drinking in humans. Ex vivo, alcohol alone increased NET formation, but upon PMA stimulation alcohol attenuated NET formation. Binge alcohol in mice resulted in a biphasic response to LPS. Initially, binge alcohol reduced LPS-induced NET formation and resulted in a diffuse distribution of neutrophils in the liver compared to alcohol-naïve mice. Moreover, indicators of NET formation including citrullinated histone H3, neutrophil elastase, and neutrophil myeloperoxidase were decreased at an early time point after LPS challenge in mice receiving binge alcohol, suggesting decreased NET formation. However, in the efferocytosis phase (15 h after LPS) citrullinated histone-H3 was increased in the liver in alcohol binge mice, suggesting decreased clearance of NETs. In vitro alcohol treatment reduced efferocytosis and phagocytosis of NETotic neutrophils and promoted expression of CD206 on MΦ. Finally, depletion of neutrophils prior to binge alcohol ameliorated LPS-induced systemic inflammation and liver injury in mice. CONCLUSIONS: Dysfunctional NETosis and efferocytosis following binge drinking exacerbate liver injury associated with sepsis. LAY SUMMARY: Disease severity in alcoholic liver disease (ALD) is associated with a significant presence of neutrophils (a type of immune cell) in the liver. It remains unknown how alcohol affects the capacity of neutrophils to control infection, a major hallmark of ALD. We found that binge alcohol drinking impaired important strategies used by neutrophils to contain and resolve infection, resulting in increased liver injury during ALD.


Subject(s)
Binge Drinking/complications , Extracellular Traps/physiology , Hepatitis, Alcoholic/etiology , Macrophages/physiology , Phagocytosis , Sepsis/etiology , Animals , HMGB1 Protein/physiology , Humans , Lipopolysaccharides , Mice , Mice, Inbred C57BL
6.
Mol Pharm ; 15(11): 5263-5276, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30354145

ABSTRACT

A series of polymer-drug conjugates based on 2-methacryloyloxyethyl phosphorylcholine (MPC) was prepared with the glioblastoma drug temozolomide (TMZ) as pendent groups. Random and block copolymers were synthesized by reversible addition-fragmentation chain-transfer (RAFT) polymerization using a TMZ-containing methacrylate monomer. The solution properties of the polyMPC-TMZ copolymers were investigated by dynamic light scattering and transmission electron microscopy, revealing well-defined nanostructures from the block copolymers. Conjugation of TMZ to polyMPC enhanced drug stability, with decomposition half-life values ranging from 2- to 19-times longer than that of free TMZ. The cytotoxicity of polyMPC-TMZ was evaluated in both chemosensitive (U87MG) and chemoresistant (T98G) glioblastoma cell lines. Furthermore, the polyMPC-TMZ platform was expanded considerably by the preparation of redox-sensitive polyMPC-TMZ copolymers utilizing disulfides as the polymer-to-drug linker.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Brain Neoplasms/drug therapy , Drug Carriers/chemistry , Glioblastoma/drug therapy , Temozolomide/administration & dosage , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Compounding/methods , Drug Screening Assays, Antitumor , Glioblastoma/pathology , Humans , Methacrylates/chemistry , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Polymerization , Polymers/chemistry
7.
J Biol Chem ; 291(1): 149-59, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26527689

ABSTRACT

Membrane-coated extracellular vesicles (EVs) released by cells can serve as vehicles for delivery of biological materials and signals. Recently, we demonstrated that alcohol-treated hepatocytes cross-talk with immune cells via exosomes containing microRNA (miRNAs). Here, we hypothesized that alcohol-exposed monocytes can communicate with naive monocytes via EVs. We observed increased numbers of EVs, mostly exosomes, secreted by primary human monocytes and THP-1 monocytic cells in the presence of alcohol in a concentration- and time-dependent manner. EVs derived from alcohol-treated monocytes stimulated naive monocytes to polarize into M2 macrophages as indicated by increased surface expression of CD68 (macrophage marker), M2 markers (CD206 (mannose receptor) and CD163 (scavenger receptor)), secretion of IL-10, and TGFß and increased phagocytic activity. miRNA profiling of the EVs derived from alcohol-treated THP-1 monocytes revealed high expression of the M2-polarizing miRNA, miR-27a. Treatment of naive monocytes with control EVs overexpressing miR-27a reproduced the effect of EVs from alcohol-treated monocytes on naive monocytes and induced M2 polarization, suggesting that the effect of alcohol EVs was mediated by miR-27a. We found that miR-27a modulated the process of phagocytosis by targeting CD206 expression on monocytes. Importantly, analysis of circulating EVs from plasma of alcoholic hepatitis patients revealed increased numbers of EVs that contained high levels of miR-27a as compared with healthy controls. Our results demonstrate the following: first, alcohol increases EV production in monocytes; second, alcohol-exposed monocytes communicate with naive monocytes via EVs; and third, miR-27a cargo in monocyte-derived EVs can program naive monocytes to polarize into M2 macrophages.


Subject(s)
Cell Differentiation/drug effects , Cell Polarity/drug effects , Ethanol/pharmacology , Extracellular Vesicles/metabolism , Macrophages/cytology , MicroRNAs/metabolism , Monocytes/cytology , Cell Line , Extracellular Vesicles/drug effects , Hepatitis, Alcoholic/blood , Hepatitis, Alcoholic/genetics , Humans , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Monocytes/drug effects , Monocytes/metabolism , Phagocytosis/drug effects , Phenotype , RNA Transport/drug effects , Receptors, Cell Surface/metabolism , Time Factors , Transforming Growth Factor beta/metabolism
8.
Hepatology ; 64(4): 1057-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27302565

ABSTRACT

UNLABELLED: The spectrum of alcoholic liver disease (ALD) is a major cause of mortality with limited therapies available. Because alcohol targets numerous signaling pathways in hepatocytes and in immune cells, the identification of a master regulatory target that modulates multiple signaling processes is attractive. In this report, we assessed the role of spleen tyrosine kinase (SYK), a nonreceptor tyrosine kinase, which has a central modulatory role in multiple proinflammatory signaling pathways involved in the pathomechanism of ALD. Using mouse disease models that represent various phases in the progression of human ALD, we found that alcohol, in all of these models, induced SYK activation in the liver, both in hepatocytes and liver mononuclear cells. Furthermore, significant SYK activation also occurred in liver samples and peripheral blood mononuclear cells of patients with ALD/alcoholic hepatitis compared to controls. Functional inhibition of SYK activation in vivo abrogated alcohol-induced hepatic neutrophil infiltration, resident immune cell activation, as well as inflammasome and extracellular signal-regulated kinase 1 and 2-mediated nuclear factor kappa B activation in mice. Strikingly, inhibition of SYK activation diminished alcohol-induced hepatic steatosis and interferon regulatory factor 3-mediated apoptosis. CONCLUSION: Our data demonstrate a novel, functional, and multicellular role for SYK phosphorylation in modulating immune cell-driven liver inflammation, hepatocyte cell death, and steatosis at different stages of ALD. These novel findings highlight SYK as a potential multifunctional target in the treatment of alcoholic steatohepatitis. (Hepatology 2016;64:1057-1071).


Subject(s)
Cell Death , Fatty Liver/prevention & control , Hepatocytes/pathology , Inflammation/prevention & control , Liver Diseases, Alcoholic/enzymology , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Syk Kinase/antagonists & inhibitors , Animals , Fatty Liver/etiology , Female , Humans , Inflammation/etiology , Liver Diseases, Alcoholic/complications , Male , Mice , Mice, Inbred C57BL , Middle Aged
9.
J Immunol ; 194(7): 3079-87, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25716995

ABSTRACT

Alcohol abuse is a leading cause of liver disease characterized by liver inflammation, fatty liver, alcoholic hepatitis, or liver cirrhosis. Immunomodulatory effects of alcohol on monocytes and macrophages contribute to alcoholic liver disease. Alcohol use, an independent risk factor for progression of hepatitis C virus (HCV) infection-mediated liver disease, impairs host defense and alters cytokine production and monocyte/macrophage activation. We hypothesized that alcohol and HCV have synergistic effects on the phenotype and function of monocytes. Our data show that acute alcohol binge drinking in healthy volunteers results in increased frequency of CD16(+) and CD68(+) and M2-type (CD206(+), dendritic cell [DC]-SIGN(+)-expressing and IL-10-secreting) circulating CD14(+) monocytes. Expression of HCV-induced CD68 and M2 markers (CD206 and DC-SIGN) in normal monocytes was further enhanced in the presence of alcohol. The levels of microRNA (miR)-27a was significantly upregulated in monocytes cultured in the presence of alcohol or alcohol and HCV as compared with HCV alone. The functional role of miR-27a in macrophage polarization was demonstrated by transfecting monocytes with an miR-27a inhibitor that resulted in reduced alcohol- and HCV- mediated monocyte activation (CD14 and CD68 expression), polarization (CD206 and DC-SIGN expression), and IL-10 secretion. Overexpression of miR-27a in monocytes enhanced IL-10 secretion via activation of the ERK signaling pathway. We found that miR-27a promoted ERK phosphorylation by downregulating the expression of ERK inhibitor sprouty2 in monocytes. Thus, we identified that sprouty2 is a target of miR-27a in human monocytes. In summary, our study demonstrates the regulatory role of miR-27a in alcohol-induced monocyte activation and polarization.


Subject(s)
Cell Differentiation/genetics , Ethanol/metabolism , Gene Expression Regulation , Macrophages/cytology , Macrophages/metabolism , MicroRNAs/genetics , Monocytes/cytology , Monocytes/metabolism , Adult , Cytokines/metabolism , Ethanol/administration & dosage , Ethanol/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Healthy Volunteers , Hepacivirus/physiology , Hepatitis C/genetics , Hepatitis C/immunology , Hepatitis C/metabolism , Humans , Immunophenotyping , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Macrophages/drug effects , Male , Membrane Proteins/metabolism , Middle Aged , Models, Biological , Monocytes/drug effects , Phenotype , Transcriptome , Young Adult
10.
J Hepatol ; 64(6): 1378-87, 2016 06.
Article in English | MEDLINE | ID: mdl-26867493

ABSTRACT

BACKGROUND & AIMS: Alcoholic liver disease (ALD) ranges from fatty liver to inflammation and cirrhosis. miRNA-155 is an important regulator of inflammation. In this study, we describe the in vivo role of miR-155 in ALD. METHODS: Wild-type (WT) (C57/BL6J) or miR-155 knockout (KO) and TLR4 KO mice received Lieber DeCarli diet for 5weeks. Some mice received corn oil or CCl4 for 2 or 9weeks. RESULTS: We found that miR-155 KO mice are protected from alcohol-induced steatosis and inflammation. The reduction in alcohol-induced fat accumulation in miR-155 KO mice was associated with increased peroxisome proliferator-activated receptor response element (PPRE) and peroxisome proliferator-activated receptors (PPAR)α (miR-155 target) binding and decreased MCP1 production. Treatment with a miR-155 inhibitor increased PPARγ expression in naïve and alcohol treated RAW macrophages. Alcohol increased lipid metabolism gene expression (FABP4, LXRα, ACC1 and LDLR) in WT mice and this was prevented in KO mice. Alcohol diet caused an increase in the number of CD163(+) CD206(+) infiltrating macrophages and neutrophils in WT mice, which was prevented in miR-155 KO mice. Kupffer cells isolated from miR-155 KO mice exhibited predominance of M2 phenotype when exposed to M1 polarized signals and this was due to increased C/EBPß. Pro-fibrotic genes were attenuated in miR-155 KO mice after alcohol diet or CCl4 treatment. Compared to WT mice, attenuation in CCl4 induced hydroxyproline and α-SMA was observed in KO mice. Finally, we show TLR4 signaling regulates miR-155 as TLR4 KO mice showed no induction of miR-155 after alcohol diet. CONCLUSIONS: Collectively our results demonstrated the role of miR-155 in alcohol-induced steatohepatitis and fibrosis in vivo.


Subject(s)
Fatty Liver, Alcoholic/etiology , Liver Cirrhosis, Experimental/etiology , MicroRNAs/physiology , Animals , Carbon Tetrachloride , DNA/metabolism , Female , Interferon-gamma/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/metabolism , Response Elements , Toll-Like Receptor 4/physiology
11.
Biochim Biophys Acta ; 1843(11): 2645-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24983769

ABSTRACT

Interferon-gamma (Ifnγ), a known immunomodulatory cytokine, regulates cell proliferation and survival. In this study, the mechanisms leading to the selective susceptibility of some tumor cells to Ifnγ were deciphered. Seven different mouse tumor cell lines tested demonstrated upregulation of MHC class I to variable extents with Ifnγ; however, only the cell lines, H6 hepatoma and L929 fibrosarcoma, that produce higher amounts of nitric oxide (NO) and reactive oxygen species (ROS) are sensitive to Ifnγ-induced cell death. NO inhibitors greatly reduce Ifnγ-induced ROS; however, ROS inhibitors did not affect the levels of Ifnγ-induced NO, demonstrating that NO regulates ROS. Consequently, NO inhibitors are more effective, compared to ROS inhibitors, in reducing Ifnγ-induced cell death. Further analysis revealed that Ifnγ induces peroxynitrite and 3-nitrotyrosine amounts and a peroxynitrite scavenger, FeTPPS, reduces cell death. Ifnγ treatment induces the phosphorylation of c-jun N-terminal kinase (Jnk) in H6 and L929 but not CT26, a colon carcinoma cell line, which is resistant to Ifnγ-mediated death. Jnk activation downstream to NO leads to induction of ROS, peroxynitrite and cell death in response to Ifnγ. Importantly, three cell lines tested, i.e. CT26, EL4 and Neuro2a, that are resistant to cell death with Ifnγ alone become sensitive to the combination of Ifnγ and NO donor or ROS inducer in a peroxynitrite-dependent manner. Overall, this study delineates the key roles of NO as the initiator and Jnk, ROS, and peroxynitrite as the effectors during Ifnγ-mediated cell death. The implications of these findings in the Ifnγ-mediated treatment of malignancies are discussed.

12.
J Hepatol ; 63(5): 1147-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26100496

ABSTRACT

BACKGROUND & AIMS: The inflammasome is a well-characterized inducer of inflammation in alcoholic steatohepatitis (ASH). Inflammasome activation requires two signals for mature interleukin (IL)-1ß production. Here we asked whether metabolic danger signals trigger inflammasome activation in ASH. METHODS: Wild-type mice, ATP receptor 2x7 (P2rx7)-KO mice, or mice overexpressing uricase were fed Lieber-DeCarli ethanol or control diet. We also implemented a pharmacological approach in which mice were treated with probenecid or allopurinol. RESULTS: The sterile danger signals, ATP and uric acid, were increased in the serum and liver of alcohol-fed mice. Depletion of uric acid or ATP, or lack of ATP signaling attenuated ASH and prevented inflammasome activation and its major downstream cytokine, IL-1ß. Pharmacological depletion of uric acid with allopurinol provided significant protection from alcohol-induced inflammatory response, steatosis and liver damage, and additional protection was achieved in mice treated with probenecid, which depletes uric acid and blocks ATP-induced P2rx7 signaling. We found that alcohol-damaged hepatocytes released uric acid and ATP in vivo and in vitro and that these sterile danger signals activated the inflammasome in LPS-exposed liver mononuclear cells. CONCLUSIONS: Our data indicate that the second signal in inflammasome activation and IL-1ß production in ASH results from the endogenous danger signals, uric acid and ATP. Inhibition of signaling triggered by uric acid and ATP may have therapeutic implications in ASH.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Allopurinol/therapeutic use , Fatty Liver, Alcoholic/metabolism , Hepatocytes/metabolism , Inflammasomes/metabolism , Probenecid/therapeutic use , Uric Acid/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Adjuvants, Pharmaceutic/therapeutic use , Animals , Antimetabolites/therapeutic use , Cells, Cultured , Disease Models, Animal , Fatty Liver, Alcoholic/drug therapy , Fatty Liver, Alcoholic/pathology , Female , Hepatocytes/drug effects , Hepatocytes/pathology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Signal Transduction , Uric Acid/metabolism
13.
J Transl Med ; 13: 261, 2015 Aug 12.
Article in English | MEDLINE | ID: mdl-26264599

ABSTRACT

BACKGROUND: It has been well documented that alcohol and its metabolites induce injury and inflammation in the liver. However, there is no potential biomarker to monitor the extent of liver injury in alcoholic hepatitis patients. MicroRNAs (miRNAs) are a class of non-coding RNAs that are involved in various physiologic and pathologic processes. In the circulation, a great proportion of miRNAs is associated with extracellular vesicles (EVs)/exosomes. Here, we hypothesized that the exosome-associated miRNAs can be used as potential biomarkers in alcoholic hepatitis (AH). METHODS: Exosomes were isolated from sera of alcohol-fed mice or pair-fed mice, and plasma of alcoholic hepatitis patients or healthy controls by ExoQuick. The exosomes were characterized by transmission electron microscopy and Western blot and enumerated with a Nanoparticle Tracking Analysis system. Firefly™ microRNA Assay was performed on miRNA extracted from mice sera. TaqMan microRNA assay was used to identify differentially expressed miRNAs in plasma of cohort of patients with AH versus controls followed by construction of receiver operating characteristic (ROC) curves to determine the sensitivity and specificity of the candidates. RESULTS: The total number of circulating EVs was significantly increased in mice after alcohol feeding. Those EVs mainly consisted of exosomes, the smaller size vesicle subpopulation of EVs. By performing microarray screening on exosomes, we found nine inflammatory miRNAs which were deregulated in sera of chronic alcohol-fed mice compared to controls including upregulated miRNAs: miRNA-192, miRNA-122, miRNA-30a, miRNA-744, miRNA-1246, miRNA 30b and miRNA-130a. The ROC analyses indicated excellent diagnostic value of miRNA-192, miRNA-122, and miRNA-30a to identify alcohol-induced liver injury. We further validated findings from our animal model in human samples. Consistent with the animal model, total number of EVs, mostly exosomes, was significantly increased in human subjects with AH. Both miRNA-192 and miRNA-30a were significantly increased in the circulation of subjects with AH. miRNA-192 showed promising value for the diagnosis of AH. CONCLUSION: Elevated level of EVs/exosomes and exosome-associated miRNA signature could serve as potential diagnostic markers for AH. In addition to the biomarker diagnostic capabilities, these findings may facilitate development of novel strategies for diagnostics, monitoring, and therapeutics of AH.


Subject(s)
Exosomes/metabolism , Hepatitis, Alcoholic/blood , Hepatitis, Alcoholic/genetics , MicroRNAs/blood , Alanine Transaminase/metabolism , Animals , Biomarkers/blood , Case-Control Studies , Ethanol , Exosomes/genetics , Exosomes/ultrastructure , Feeding Behavior , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , RNA Stability/genetics , ROC Curve , Software
14.
J Transl Med ; 13: 193, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26077675

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is becoming a pandemic. While multiple 'hits' have been reported to contribute to NAFLD progression to non-alcoholic steatohepatitis (NASH), fibrosis and liver cancer, understanding the natural history of the specific molecular signals leading to hepatocyte damage, inflammation and fibrosis, is hampered by the lack of suitable animal models that reproduce disease progression in humans. The purpose of this study was first, to develop a mouse model that closely mimics progressive NAFLD covering the spectrum of immune, metabolic and histopathologic abnormalities present in human disease; and second, to characterize the temporal relationship between sterile/exogenous danger signals, inflammation, inflammasome activation and NAFLD progression. METHODS: Male C57Bl/6 mice were fed a high fat diet with high cholesterol and a high sugar supplement (HF-HC-HSD) for 8, 27, and 49 weeks and the extent of steatosis, liver inflammation, fibrosis and tumor development were evaluated at each time point. RESULTS: The HF-HC-HSD resulted in liver steatosis at 8 weeks, progressing to steatohepatitis and early fibrosis at 27 weeks, and steatohepatitis, fibrosis, and tumor development at 49 weeks compared to chow diet. Steatohepatitis was characterized by increased levels of MCP-1, TNFα, IL-1ß and increased liver NASH histological score. We found increased serum levels of sterile danger signals, uric acid and HMGB1, as early as 8 weeks, while endotoxin and ATP levels increased only after 49 weeks. Increased levels of these sterile and microbial danger signals paralleled upregulation and activation of the multiprotein complex inflammasome. At 27, 49 weeks of HF-HC-HSD, activation of M1 macrophages and loss of M2 macrophages as well as liver fibrosis were present. Finally, similar to human NASH, liver tumors occurred in 41% of mice in the absence of cirrhosis and livers expressed increased p53 and detectable AFP. CONCLUSIONS: HF-HC-HSD over 49 weeks induces the full spectrum of liver pathophysiologic changes that characterizes the progression of NAFLD in humans. NAFLD progression to NASH, fibrosis and liver tumor follows progressive accumulation of sterile and microbial danger signals, inflammasome activation, altered M1/M2 cell ratios that likely contribute to NASH progression and hepatic tumor formation.


Subject(s)
Disease Progression , Inflammation/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Biomarkers/metabolism , Body Weight , Cholesterol/adverse effects , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dietary Carbohydrates/adverse effects , Disease Models, Animal , Feeding Behavior , Female , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Insulin/metabolism , Kupffer Cells/pathology , Liver Cirrhosis/complications , Liver Neoplasms/complications , Macrophage Activation , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/complications , Organ Size , Time Factors , Up-Regulation
15.
Adv Exp Med Biol ; 815: 197-216, 2015.
Article in English | MEDLINE | ID: mdl-25427909

ABSTRACT

Liver cancers are one of the deadliest known malignancies which are increasingly becoming a major public health problem in both developed and developing countries. Overwhelming evidence suggests a strong role of infection with hepatitis B and C virus (HBV and HCV), alcohol abuse, as well as metabolic diseases such as obesity and diabetes either individually or synergistically to cause or exacerbate the development of liver cancers. Although numerous etiologic mechanisms for liver cancer development have been advanced and well characterized, the lack of definite curative treatments means that gaps in knowledge still exist in identifying key molecular mechanisms and pathways in the pathophysiology of liver cancers. Given the limited success with current therapies and preventive strategies against liver cancer, there is an urgent need to identify new therapeutic options for patients. Targeting HCV and or alcohol-induced signal transduction, or virus-host protein interactions may offer novel therapies for liver cancer. This review summarizes current knowledge on the mechanistic development of liver cancer associated with HCV infection and alcohol abuse as well as highlights potential novel therapeutic strategies.


Subject(s)
Ethanol/toxicity , Hepatitis C/complications , Liver Neoplasms/etiology , Animals , Humans , Immunity, Innate , Killer Cells, Natural/immunology , MicroRNAs/physiology , Oxidative Stress , Virus Replication/drug effects
16.
J Hepatol ; 59(3): 442-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23665181

ABSTRACT

BACKGROUND & AIMS: Interferon-γ (IFN-γ), a cytokine produced by activated natural killer cells (NK) and T lymphocytes, is an important regulator of innate and adaptive immunity during hepatitis C virus (HCV) infection. However, the cellular sources and mechanisms of IFN-γ induction in HCV-infection are not fully understood. METHODS: We cultured normal human peripheral blood mononuclear cells (PBMCs) with different populations of immune cells and JFH-1 HCV-infected HuH7.5 (JFH-1/HuH7.5) cells. RESULTS: We found that PBMCs produced large amounts of IFN-γ after co-culture with JFH-1/HuH7.5 cells. Using intracellular cytokine staining, we confirmed that NK cells and NKT cells (to a lesser extent) were the major IFN-γ producers within PBMCs. Purified NK/NKT cells did not produce IFN-γ in response to JFH-1/HuH7.5 cells and depletion of accessory (HLA-DR(+)) cells prevented IFN-γ induction in PBMCs. Through selective cell depletion of dendritic cells or monocytes from PBMCs, we determined that plasmacytoid dendritic cells (pDCs) were indispensable for NK-IFN-γ induction and the presence of monocytes was needed for maximal NK-IFN-γ induction. We further revealed that NK-IFN-γ induction depended on pDC-derived IFN-α while other IFN-γ inducing cytokines, IL-12, and IL-18, played minimal roles. Close contact between JFH-1/HuH7.5 cells and NK cells was required for IFN-γ production and monocyte-derived IL-15 significantly augmented IFN-γ induction. CONCLUSIONS: We discovered a novel mechanism where NK cells interact with pDCs and monocytes, efficiently producing IFN-γ in response to HCV-infected cells. This indicates that co-operation between NK cells and accessory cells is critical for IFN-γ production and regulation of immunity during HCV infection.


Subject(s)
Hepacivirus/immunology , Hepacivirus/pathogenicity , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Animals , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/immunology , Hepatitis C/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Interferon Type I/biosynthesis , Liver Neoplasms, Experimental/immunology , Liver Neoplasms, Experimental/virology , Monocytes/immunology
17.
Int J Cancer ; 130(4): 865-75, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-21455983

ABSTRACT

Mycobacterium indicus pranii (MIP) is approved for use as an adjuvant (Immuvac/Cadi-05) in the treatment of leprosy. In addition, its efficacy is being investigated in clinical trials on patients with tuberculosis and different tumors. To evaluate and delineate the mechanisms by which autoclaved MIP enhances anti-tumor responses, the growth of solid tumors consisting of Sp2/0 (myeloma) and EL4 (thymoma) cells was studied in BALB/c and C57BL/6 mice, respectively. Treatment of mice with a single intra-dermal (i.d.) injection of MIP 3 days after Sp2/0 implantation greatly suppresses tumor growth. MIP treatment of tumor bearing mice lowers Interleukin (IL)6 but increases IL12p70 and IFNγ amounts in sera. Also, increase in CD8(+) T cell mediated lysis of specific tumor targets and production of high amounts of IL2 and IFNγ by CD4(+) T cells upon stimulation with specific tumor antigens in MIP treated mice is observed. Furthermore, MIP is also effective in reducing the growth of EL4 tumors; however, this efficacy is reduced in Ifnγ(-/-) mice. In fact, several MIP mediated anti-tumor responses are greatly abrogated in Ifnγ(-/-) mice: increase in serum Interleukin (IL)12p70 amounts, induction of IL2 and lysis of EL4 targets by splenocytes upon stimulation with specific tumor antigens. Interestingly, tumor-induced increase in serum IL12p70 and IFNγ and reduction in growth of Sp2/0 and EL4 tumors by MIP are not observed in nonobese diabetic severe combined immunodeficiency mice. Overall, our study clearly demonstrates the importance of a functional immune network, in particular endogenous CD4(+) and CD8(+) T cells and IFNγ, in mediating the anti-tumor responses by MIP.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cytotoxicity, Immunologic , Interferon-gamma/physiology , Mycobacterium/immunology , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Adjuvants, Immunologic/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th1 Cells/immunology
18.
Hepatology ; 50(5): 1547-57, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19739265

ABSTRACT

UNLABELLED: Hepatotoxicity due to overdose of the analgesic and antipyretic acetaminophen (APAP) is a major cause of liver failure in adults. To better understand the contributions of different signaling pathways, the expression and role of Ras activation was evaluated after oral dosing of mice with APAP (400-500 mg/kg). Ras-guanosine triphosphate (GTP) is induced early and in an oxidative stress-dependent manner. The functional role of Ras activation was studied by a single intraperitoneal injection of the neutral sphingomyelinase and farnesyltransferase inhibitor (FTI) manumycin A (1 mg/kg), which lowers induction of Ras-GTP and serum amounts of alanine aminotransferase (ALT). APAP dosing decreases hepatic glutathione amounts, which are not affected by manumycin A treatment. However, APAP-induced activation of c-Jun N-terminal kinase, which plays an important role, is reduced by manumycin A. Also, APAP-induced mitochondrial reactive oxygen species are reduced by manumycin A at a later time point during liver injury. Importantly, the induction of genes involved in the inflammatory response (including iNos, gp91phox, and Fasl) and serum amounts of proinflammatory cytokines interferon-gamma (IFNgamma) and tumor necrosis factor alpha, which increase greatly with APAP challenge, are suppressed with manumycin A. The FTI activity of manumycin A is most likely involved in reducing APAP-induced liver injury, because a specific neutral sphingomyelinase inhibitor, GW4869 (1 mg/kg), did not show any hepatoprotective effect. Notably, a structurally distinct FTI, gliotoxin (1 mg/kg), also inhibits Ras activation and reduces serum amounts of ALT and IFN-gamma after APAP dosing. Finally, histological analysis confirmed the hepatoprotective effect of manumycin A and gliotoxin during APAP-induced liver damage. CONCLUSION: This study identifies a key role for Ras activation and demonstrates the therapeutic efficacy of FTIs during APAP-induced liver injury.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Farnesyltranstransferase/antagonists & inhibitors , Gliotoxin/pharmacology , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , ras Proteins/metabolism , Alanine Transaminase/metabolism , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/metabolism , Gliotoxin/therapeutic use , Guanosine Triphosphate/metabolism , Interferon-gamma/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred BALB C , Oxidative Stress/drug effects , Oxidative Stress/physiology , Polyenes/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
19.
Cytokine ; 50(1): 1-14, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20036577

ABSTRACT

Interferon-gamma (IFNgamma) is a central regulator of the immune response and signals via the Janus Activated Kinase (JAK)-Signal Transducer and Activator of Transcription (STAT) pathway. Phosphorylated STAT1 homodimers translocate to the nucleus, bind to Gamma Activating Sequence (GAS) and recruit additional factors to modulate gene expression. A bioinformatics analysis revealed that greater number of putative promoters of immune related genes and also those not directly involved in immunity contain GAS compared to response elements (RE) for Interferon Regulatory Factor (IRF)1, Nuclear factor kappa B (NFkappaB) and Activator Protein (AP)1. GAS is present in putative promoters of well known IFNgamma-induced genes, IRF1, GBP1, CXCL10, and other genes identified were TLR3, VCAM1, CASP4, etc. Analysis of three microarray studies revealed that the expression of a subset of only GAS containing immune genes were modulated by IFNgamma. As a significant correlation exists between GAS containing immune genes and IFNgamma-regulated gene expression, this strategy may identify novel IFNgamma-responsive immune genes. This analysis is integrated with the literature on the roles of IFNgamma in mediating a plethora of functions: anti-microbial responses, antigen processing, inflammation, growth suppression, cell death, tumor immunity and autoimmunity. Overall, this review summarizes our present knowledge on IFNgamma mediated signaling and functions.


Subject(s)
Gene Expression Regulation , Immunomodulation/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Animals , Antigen Presentation/immunology , Autoimmunity/immunology , Humans , Neoplasms/immunology , Neoplasms/pathology
20.
Cell Mol Gastroenterol Hepatol ; 2(3): 302-316.e8, 2016 May.
Article in English | MEDLINE | ID: mdl-28090562

ABSTRACT

BACKGROUND & AIMS: Monocyte and macrophage (MΦ) activation contributes to the pathogenesis of chronic hepatitis C virus (HCV) infection. Disease pathogenesis is regulated by both liver-resident MΦs and monocytes recruited as precursors of MΦs into the damaged liver. Monocytes differentiate into M1 (classic/proinflammatory) or M2 (alternative/anti-inflammatory) polarized MΦs in response to tissue microenvironment. We hypothesized that HCV-infected hepatoma cells (infected with Japanese fulminant hepatitis-1 [Huh7.5/JFH-1]) induce monocyte differentiation into polarized MΦs. METHODS: Healthy human monocytes were co-cultured with Huh7.5/JFH-1 cells or cell-free virus for 7 days and analyzed for MΦ markers and cytokine levels. A similar analysis was performed on circulating monocytes and liver MΦs from HCV-infected patients and controls. RESULTS: Huh7.5/JFH-1 cells induced monocytes to differentiate into MΦs with increased expression of CD14 and CD68. HCV-MΦs showed M2 surface markers (CD206, CD163, and Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN)) and produced both proinflammatory and anti-inflammatory cytokines. HCV-induced early interleukin 1ß production promoted transforming growth factor (TGF)ß production and MΦ polarization to an M2 phenotype. TGF-ß secreted by M2-MΦ led to hepatic stellate cell activation indicated by increased expression of collagen, tissue inhibitor of metalloproteinase 1, and α-smooth muscle actin. In vivo, we observed a significant increase in M2 marker (CD206) expression on circulating monocytes and in the liver of chronic HCV-infected patients. Furthermore, we observed the presence of a unique collagen-expressing CD14+CD206+ monocyte population in HCV patients that correlated with liver fibrosis. CONCLUSIONS: We show an important role for HCV in induction of monocyte differentiation into MΦs with a mixed M1/M2 cytokine profile and M2 surface phenotype that promote stellate cell activation via TGF-ß. We also identified circulating monocytes expressing M2 marker and collagen in chronic HCV infection that can be explored as a biomarker.

SELECTION OF CITATIONS
SEARCH DETAIL