Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 475
Filtrar
1.
Nat Commun ; 12(1): 7172, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34887405

RESUMO

Complement receptor of immunoglobulin superfamily (CRIg) is expressed on liver macrophages and directly binds complement component C3b or Gram-positive bacteria to mediate phagocytosis. CRIg plays important roles in several immune-mediated diseases, but it is not clear how its pathogen recognition and phagocytic functions maintain homeostasis and prevent disease. We previously associated cytolysin-positive Enterococcus faecalis with severity of alcohol-related liver disease. Here, we demonstrate that CRIg is reduced in liver tissues from patients with alcohol-related liver disease. CRIg-deficient mice developed more severe ethanol-induced liver disease than wild-type mice; disease severity was reduced with loss of toll-like receptor 2. CRIg-deficient mice were less efficient than wild-type mice at clearing Gram-positive bacteria such as Enterococcus faecalis that had translocated from gut to liver. Administration of the soluble extracellular domain CRIg-Ig protein protected mice from ethanol-induced steatohepatitis. Our findings indicate that ethanol impairs hepatic clearance of translocated pathobionts, via decreased hepatic CRIg, which facilitates progression of liver disease.


Assuntos
Enterococcus faecalis/imunologia , Infecções por Bactérias Gram-Positivas/imunologia , Hepatopatias Alcoólicas/imunologia , Macrófagos/imunologia , Receptores de Complemento 3b/imunologia , Receptores de Complemento/imunologia , Animais , Translocação Bacteriana , Complemento C3b/imunologia , Enterococcus faecalis/fisiologia , Etanol/efeitos adversos , Feminino , Trato Gastrointestinal/microbiologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/microbiologia , Hepatopatias Alcoólicas/etiologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento/deficiência , Receptores de Complemento/genética , Receptores de Complemento 3b/genética
2.
Sci Rep ; 11(1): 22811, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34819599

RESUMO

Camel milk (CM) is considered to protect the liver in the practice of traditional medicine in nomadic areas. The purpose of the present study was to investigate the effects of CM on the hepatic biochemical and multiple omics alterations induced by chronic alcoholic liver disease (ALD). An intragastric gavage mice Lieber DeCarli + Gao binge model (NIAAA model) was employed to investigate the inflammatory mechanism of camel milk on the liver tissue of mice. A gut microbiota of the feces of mice and transcriptomic and proteomic analyses of the liver of mice were performed. Analysis of serum and liver biochemical indexes revealed that camel milk not only prevents alcohol-induced colonic dysfunction and lipid accumulation, but also regulates oxidative stress and inflammatory cytokine production to protect against chronic ALD in mouse. The gut microbial community of mice treated with camel milk was more similar to the untreated control group than to the model group, indicating that the intake of camel milk pre- and post-alcohol gavage effectively prevents and alleviates the intestinal microbial disorder caused by chronic alcoholism in mice. Furthermore, the results of the transcriptomic and proteomic analyses of the liver tissue showed that camel milk can improve alcoholic liver injury in mice by regulating inflammatory factors and immune system disruptions. This study provides insights into the molecular mechanism by which camel milk can be developed as a potential functional food with no side effects and against liver injury.


Assuntos
Anti-Inflamatórios/administração & dosagem , Camelus , Mediadores da Inflamação/metabolismo , Intestinos/metabolismo , Hepatopatias Alcoólicas/prevenção & controle , Fígado/metabolismo , Leite , Animais , Consumo Excessivo de Bebidas Alcoólicas , Modelos Animais de Doenças , Disbiose , Alimento Funcional , Microbioma Gastrointestinal , Intestinos/imunologia , Intestinos/microbiologia , Metabolismo dos Lipídeos , Fígado/imunologia , Fígado/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Proteoma , Transcriptoma
3.
Front Immunol ; 12: 695491, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489943

RESUMO

Patients with liver disease are susceptible to infection with Vibrio vulnificus (V. vulnificus), but the specific reasons remain elusive. Through RNA-seq, we found that when mice with alcoholic liver disease (ALD) were infected with V. vulnificus by gavage, compared with the Pair group, the small intestinal genes affecting intestinal permeability were upregulated; and the number of differentially expressed genes related to immune functions (e.g., such as cell chemotaxis, leukocyte differentiation, and neutrophil degranulation) decreased in the liver, spleen, and blood. Further analysis showed that the number of white blood cells decreased in the Pair group, whereas those in the ALD mice did not change significantly. Interestingly, the blood bacterial load in the ALD mice was about 100 times higher than that of the Pair group. After the ALD mice were infected with V. vulnificus, the concentrations of T cell proliferation-promoting cytokines (IL-2, IL-23) decreased. Therefore, unlike the Pair group, ALD mice had weaker immune responses, lower T cell proliferation-promoting cytokines, and higher bacterial loads post-infection, possibly increasing their susceptibility to V. vulnificus infection. These new findings we presented here may help to advance the current understanding of the reasons why patients with liver disease are susceptible to V. vulnificus infection and provides potential targets for further investigation in the context of treatment options for V. vulnificus sepsis in liver disease patient.


Assuntos
Citocinas/metabolismo , Hepatopatias Alcoólicas/imunologia , Transcriptoma , Vibrioses/imunologia , Vibrio vulnificus/patogenicidade , Animais , Carga Bacteriana , Proliferação de Células , Citocinas/genética , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Ativação Linfocitária , Camundongos Endogâmicos C57BL , RNA-Seq , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia , Vibrioses/genética , Vibrioses/metabolismo , Vibrio vulnificus/crescimento & desenvolvimento , Vibrio vulnificus/imunologia
4.
Eur Rev Med Pharmacol Sci ; 25(13): 4563-4569, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34286499

RESUMO

OBJECTIVE: Sepsis is one of the most common complications and causes of death in patients with Alcohol-related Liver Disease. This narrative review will focus on several aspects of sepsis in the context of Alcohol-related Liver Disease. The pathophysiology of the increased susceptibility to infections consists mainly of impaired innate and adaptive immunity, changes in gut microbiota with consequent gut translocation of bacteria due to both alcohol abuse and the underlying liver disease. The diagnosis of sepsis in the context of Alcohol-related Liver Disease is challenging. Moreover, the use of classical acute-phase serum proteins (e.g., C-reactive protein and procalcitonin) has several limitations in this setting. The early administration of an adequate antibiotic treatment is pivotal. Finally, measures of infection control and prevention are needed because the prognosis of sepsis in patients affected by Alcohol-related Liver Disease is poor.


Assuntos
Antibacterianos/uso terapêutico , Suscetibilidade a Doenças/imunologia , Hepatopatias Alcoólicas/complicações , Sepse/imunologia , Proteína C-Reativa/análise , Suscetibilidade a Doenças/sangue , Suscetibilidade a Doenças/diagnóstico , Humanos , Hepatopatias Alcoólicas/sangue , Hepatopatias Alcoólicas/imunologia , Pró-Calcitonina/sangue , Prognóstico , Sepse/sangue , Sepse/diagnóstico , Sepse/tratamento farmacológico , Resultado do Tratamento
5.
Int Immunopharmacol ; 95: 107471, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33756231

RESUMO

Alcoholic liver disease (ALD) is the most common chronic liver disease worldwide. Currently, there is no definitive treatment for alcohol-induced liver injury (ALI). Inflammatory response and oxidative stress play a crucial role in ALI. Cyclooxygenase 2 (COX-2) can be induced by inflammation and it has been reported that the enhanced expression of COX-2 in alcoholic liver injury. Rutaecarpine (RUT) was extracted from evodia rutaecarpa. RUT has a wide range of pharmacological activities. In order to increase its anti-inflammatory activity, our group introduced sulfonyl group to synthesized the 3-[2-(trifluoromethoxy)benzenesulfonamide]-rutaecarpine (3-B-RUT). In this study, we explored the protective effect of 3-B-RUT on alcoholic liver injury in vivo and in vitro and preliminarily explore its mechanism. Mice ALI model was established according to the chronic-plus-binge ethanol model. Results showed that 3-B-RUT (20 µg/kg) attenuated alcohol-induced liver injury and suppressed liver inflammation and oxidative stress, and the effect was comparable to RUT (20 mg/kg). In vitro results are consistent with in vivo results. Mechanistically, the 3-B-RUT might suppress inflammatory response and oxidative stress by regulating activation of NF-κB/COX-2 pathway. In summary, 3-B-RUT, a derivative of RUT, may be a promising clinical candidate for ALI treatment.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Alcaloides Indólicos/uso terapêutico , Hepatopatias Alcoólicas/tratamento farmacológico , Quinazolinas/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Alcaloides Indólicos/farmacologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , Estresse Oxidativo/efeitos dos fármacos , Quinazolinas/farmacologia , Células RAW 264.7
6.
Int J Biol Sci ; 17(1): 307-327, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33390852

RESUMO

Uncovering the intricacies of the gut microbiome and how it interacts with the host immune system has opened up pathways in the search for the treatment of disease conditions. Alcohol-associated liver disease is a major cause of death worldwide. Research has shed light on the breakdown of the protective gut barriers, translocation of gut microbes to the liver and inflammatory immune response to microbes all contributing to alcohol-associated liver disease. This knowledge has opened up avenues for alternative therapies to alleviate alcohol-associated liver disease based on the interaction of the commensal gut microbiome as a key player in the regulation of the immune response. This review describes the relevance of the intestinal immune system, the gut microbiota, and specialized and non-specialized intestinal cells in the regulation of intestinal homeostasis. It also reflects how these components are altered during alcohol-associated liver disease and discusses new approaches for potential future therapies in alcohol-associated liver disease.


Assuntos
Microbioma Gastrointestinal , Mucosa Intestinal/imunologia , Hepatopatias Alcoólicas/imunologia , Animais , Disbiose/terapia , Homeostase , Humanos , Mucosa Intestinal/microbiologia , Fígado/imunologia , Hepatopatias Alcoólicas/microbiologia , Hepatopatias Alcoólicas/prevenção & controle
7.
Int Immunopharmacol ; 88: 106968, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33182058

RESUMO

Alcohol-induced liver injury is characterized by abnormal liver dysfunction and excessive inflammation response. Recent years a wealth of data have been yielded indicating that EtOH (ethyl alcohol)-induced macrophage activation along with liver inflammation plays a dominating role in the progression of alcohol-induced liver injury. Here we found high expression of NLRP12 (Nucleotide-binding oligomerization domain protein 12, which is generally considered to be a negative regulator of inflammatory response) in EtOH-fed mouse liver tissue, primary Kupffer cells and EtOH-induced RAW264.7 cells. Additionally, overexpression of NLRP12 following Ad (adenovirus)-NLRP12-EGFP contributed to the attenuation of steatosis and inflammation in EtOH-fed mice model and EtOH-primed RAW264.7 cells. In parallel, Knockdown of NLRP12 aggravated the inflammatory response in RAW264.7 cells triggered by EtOH. Meanwhile, after administration of overexpression or inhibition of NLRP12 expression in vitro, the expression of phosphorylated protein of NF-kB signaling pathway was significantly affected. After increasing or decreasing the expression of NLRP12 in RAW264.7 cells, AML-12 cells were cultured with the supernatant of RAW264.7 cells stimulated by EtOH, and the percent of apoptosis ratio of AML-12 cells was remarkably altered. The study suggested that reduced inflammatory response induced by NLRP12-mediated inhibition of NF-kB pathway participated in the decrease of hepatocyte apoptosis in alcohol-induced liver injury. Collectively, these findings suggested the significance of NLRP12-mediated macrophage activation in alcohol-induced liver injury.


Assuntos
Apoptose , Hepatócitos/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Hepatopatias Alcoólicas/imunologia , Ativação de Macrófagos , NF-kappa B/imunologia , Animais , Linhagem Celular , Citocinas/imunologia , Etanol , Fígado/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
8.
Clin Sci (Lond) ; 134(14): 1935-1956, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32639005

RESUMO

The regulation of macrophages during inflammatory responses is a crucial process in alcoholic liver disease (ALD) and aberrant macrophage DNA methylation is associated with inflammation. Our preliminary screening results of macrophage methylation in the present study demonstrated the zinc finger SWI2/SNF2 and MuDR (SWIM)-domain containing 3 (ZSWIM3) were hypermethylated in the 5' untranslated region (5'-UTR) region. ZSWIM3, a novel zinc finger-chelate domain of SWIM, is predicted to function in DNA-binding and protein-binding interactions. Its expression was found to be consistently decreased in macrophages isolated from livers of ethyl alcohol (EtOH)-fed mice and in EtOH+lipopolysaccharide (LPS)-induced RAW264.7 cells. Over-expression of ZSWIM3 was found to attenuate chronic+binge ethanol feeding-induced liver injury and inhibit inflammatory responses in vivo. Enforced expression of ZSWIM3 in vitro was also found to have anti-inflammatory effects. Aberrant expression of ZSWIM3 in alcohol-induced liver injury (ALI) was found to be associated with hypermethylation. Analysis of CpG prediction indicated the presence of two methylated sites in the ZSWIM3 promoter region and methylation inhibitor and DNA methyltransferases (DNMTs)-siRNA transfection were found to restore down-regulated ZSWIM3. Chromatin immunoprecipitation (ChIP) assay and molecular docking affirmed the role of DNMT 3b (DNMT3b) as a principal regulator of ZSWIM3 expression. Mechanistically, ZSWIM3 might affect inflammation by binding with tumor necrosis factor receptor-associated factor 2 (TRAF2), which further mediates the activation of the nuclear transcription factor κB (NF-κB) pathway. The present study, therefore, provides detailed insights into the possible structure and function of ZSWIM3 and thus, contributes new substantial research in the elucidation of the pathogenesis of ALI.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Hepatopatias Alcoólicas/metabolismo , Macrófagos/metabolismo , Animais , Metilação de DNA , Modelos Animais de Doenças , Hepatopatias Alcoólicas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , NF-kappa B/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , DNA Metiltransferase 3B
9.
Inflamm Res ; 69(8): 789-800, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32451556

RESUMO

OBJECTIVE: Transmembrane protein 88 (TMEM88), a new protein of increasing concern existed in cell membrane, inhibits the typical Wnt/ß-catenin signaling pathway to play a regulatory role on cell proliferation by binding to Dishevelled-1. Until recently, the connection between TMEM88 and alcoholic liver disease is unknown. In this research, we explored the effect of TMEM88 on the secretion of inflammatory cytokines in ethanol (EtOH)-induced RAW264.7 cells, moreover, the function of YAP signaling pathway in EtOH-induced RAW264.7 cells were investigated. METHODS: We administered TMEM88 adenovirus (ADV-TMEM88) by tail vein injection into C57BL/6J mice in vivo. In vitro, RAW264.7 murine macrophages were stimulated with EtOH and were transfected with pEGFP-C1-TMEM88 and TMEM88 siRNA, respectively, protein expression and mRNA expression of IL-6 and IL-1ß were assessed by Western Blotting and RT-qPCR, respectively. RESULTS: Our group found that the overexpression of TMEM88 led to an up-regulation of IL-6 and IL-1ß secretion, hinting that it had the possibility of linking with the initiation, the development, and the end of inflammation. In addition to that, TMEM88 silencing reduced the secretion of IL-6 and IL-1ß in RAW264.7 cells. Moreover, we demonstrated that the YAP signaling pathway under the action of EtOH was activated by TMEM88. CONCLUSIONS: All in all, these experimental outcomes indicated that TMEM88 had an indispensable impact on EtOH-induced secretion of inflammatory cytokines (IL-6 and IL-1ß) in RAW264.7 cells through YAP signaling pathway.


Assuntos
Citocinas/biossíntese , Lipoproteínas/fisiologia , Hepatopatias Alcoólicas/etiologia , Proteínas de Membrana/fisiologia , Transativadores/fisiologia , Animais , Apoptose/efeitos dos fármacos , Etanol/farmacologia , Hepatopatias Alcoólicas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Transdução de Sinais/fisiologia
10.
Alcohol Clin Exp Res ; 44(5): 1061-1074, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32154597

RESUMO

BACKGROUND: Liver is enriched in several innate-like unconventional T cells, but their role in alcohol-related liver disease (ALD) is not fully understood. Studies in several acute alcohol feeding models but not in chronic alcoholic steatohepatitis (ASH) model have shown that invariant natural killer T (iNKT) cells play a pathogenic role in ALD. Here, we investigated the activation of iNKT cells in an intragastric (iG) infusion model of chronic ASH as well as the frequency and cytokine phenotype of 3 different unconventional T cells: iNKT, mucosal-associated invariant T (MAIT), and CD8+ CD161hi Vα7.2- cells in peripheral blood of ALD patients. METHODS: Hepatic iNKT cells were investigated using the iG model of chronic ASH that combines feeding of high-cholesterol/high-fat diet (HCFD) with intragastric feeding of ethanol diet (HCFD + iG Alc). Human iNKT, MAIT, and CD8+ CD161hi Vα7.2- cells were examined by flow cytometry in peripheral blood of patients with severe alcoholic hepatitis (SAH) and chronic alcoholics (ChA) and compared with healthy controls. RESULTS: In the iG model of chronic ASH, IFNγ+ iNKT cells accumulate in their livers compared with pair-fed control mice and activated hepatic iNKT cells show high expression of Fas and FasL. Notably, IFNγ+ iNKT cells are also significantly increased in peripheral blood of ChA patients compared with SAH patients. MAIT cells are significantly reduced in all ALD patients, but CD8+ CD161hi Vα7.2- cells are increased in SAH patients. Although MAIT and CD8+ CD161hi Vα7.2- cells displayed a similar cytokine production profile, the production of IFNγ and TNFα is significantly increased in SAH patients, while significant IL-17A production is found in ChA patients. CONCLUSIONS: We found that the 3 unconventional T cells are activated in ALD patients showing interesting differences in their frequency and cytokine production profile between SAH and ChA patients. In the iG murine model of chronic ASH, iNKT cells are also activated secreting proinflammatory cytokines suggesting their involvement in liver disease.


Assuntos
Hepatopatias Alcoólicas/imunologia , Ativação Linfocitária/imunologia , Células T Matadoras Naturais/imunologia , Linfócitos T/imunologia , Alcoolismo/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Citocinas/metabolismo , Etanol/administração & dosagem , Hepatite Alcoólica/imunologia , Humanos , Fígado/patologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Células T Invariantes Associadas à Mucosa , Subfamília B de Receptores Semelhantes a Lectina de Células NK/análise
11.
Int J Biol Sci ; 16(3): 460-470, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32015682

RESUMO

Mucosal-associated invariant T cells (MAIT cells) are a new population of innate immune cells, which are abundant in the liver and play complex roles in various liver diseases. In this review, we summarize MAIT cells in the liver diseases in recent studies, figure out the role of MAIT cells in various liver disease, including Alcoholic liver disease, Non-alcoholic liver disease, Autoimmune liver diseases, Viral hepatitis and Liver Cancer. Briefly, MAIT cells are involved in anti-bacteria responses in the alcoholic liver diseases. Besides, the activated MAIT cells promote the liver inflammation by secreting inflammatory cytokines and produce regulatory cytokines, which induces anti-inflammatory macrophage polarization. MAIT cells participate in the liver fibrosis via enhancing hepatic stellate cell activation. In viral hepatitis, MAIT cells exhibit a flawed and exhausted phenotype, which results in little effect on controlling the virus and bacteria. In liver cancer, MAIT cells indicate the disease progression and the outcome of therapy. In summary, MAIT cells are attractive biomarkers and therapeutic targets for liver disease.


Assuntos
Doenças Autoimunes/imunologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias/imunologia , Neoplasias Hepáticas/imunologia , Células T Invariantes Associadas à Mucosa/metabolismo , Animais , Humanos
12.
Sci Rep ; 10(1): 2451, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32051453

RESUMO

Murine models of chronic alcohol consumption are frequently used to investigate alcoholic liver injury and define new therapeutic targets. Lieber-DeCarli diet (LD) and Meadows-Cook diet (MC) are the most accepted models of chronic alcohol consumption. It is unclear how similar these models are at the cellular, immunologic, and transcriptome levels. We investigated the common and specific pathways of LD and MC models. Livers from LD and MC mice were subjected to histologic changes, hepatic leukocyte population, hepatic transcripts level related to leukocyte recruitment, and hepatic RNA-seq analysis. Cross-species comparison was performed using the alcoholic liver disease (ALD) transcriptomic public dataset. Despite LD mice have increased liver injury and steatosis by alcohol exposure, the number of CD45+ cells were reduced. Opposite, MC mice have an increased number of monocytes/liver by alcohol. The pattern of chemokine gradient, adhesion molecules, and cytokine transcripts is highly specific for each model, not shared with advanced human alcoholic liver disease. Moreover, hepatic RNA-seq revealed a limited and restricted number of shared genes differentially changed by alcohol exposure in these 2 models. Thus, mechanisms involved in alcohol tissue injury are model-dependent at multiple levels and raise the consideration of significant pathophysiological diversity of human alcoholic liver injury.


Assuntos
Consumo de Bebidas Alcoólicas/patologia , Alcoolismo/patologia , Hepatopatias Alcoólicas/patologia , Fígado/patologia , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/imunologia , Alcoolismo/etiologia , Alcoolismo/genética , Alcoolismo/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Humanos , Fígado/imunologia , Fígado/metabolismo , Hepatopatias Alcoólicas/etiologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
13.
Cell Mol Gastroenterol Hepatol ; 9(1): 145-160, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31562937

RESUMO

BACKGROUND & AIMS: Toll-like receptor 2 (TLR2) and TLR3 regulate hepatic immunity under pathological conditions, but their functions and potential drug targets in alcoholic liver disease (ALD) remain poorly understood. METHODS: ALD-associated liver injury were induced in TLR2 knockout (TLR2-/-), TLR3-/-, TLR2-/- bone marrow transplanted (BMT), TLR3-/- BMT, IL-10-/- mice, and their wild-type littermates through ethanol challenge with or without co-administered epigallocatechin-3-gallate (EGCG). Moreover, Kupffer cells were depleted by GdCl3 injection to evaluate their pathogenic roles in ALD. RESULTS: We identified that deficiency of TLR2 and TLR3 significantly alleviated and aggravated ALD-induced liver injury, respectively. Mechanistically, Kupffer cell inactivation, M1 to M2 polarization, and IL-10 production via STAT3 activation contributed to hepatic protection mediated by concurrent TLR2 inhibition and TLR3 agonism. These findings were further confirmed in TLR2 and TLR3 BMT mice. We also identified a novel ALD-protective agent EGCG which directly interacted with Kupffer cell TLR2/3 to induce IL-10 production. Deficiency of IL-10 aggravated ALD injury and blunted EGCG-mediated hepatoprotection while depletion of Kupffer cells partially recovered liver injury but abolished EGCG's actions. CONCLUSIONS: Altogether, our results illustrate the divergent roles of Kupffer cells TLR2/3 in ALD progression via anti-inflammatory cytokine IL-10 production.


Assuntos
Células de Kupffer/patologia , Hepatopatias Alcoólicas/patologia , Fígado/patologia , Receptor 2 Toll-Like/deficiência , Receptor 3 Toll-Like/deficiência , Animais , Catequina/administração & dosagem , Catequina/análogos & derivados , Progressão da Doença , Etanol/administração & dosagem , Etanol/toxicidade , Humanos , Interleucina-10/imunologia , Interleucina-10/metabolismo , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/imunologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Substâncias Protetoras/administração & dosagem , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/genética , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/genética
14.
Int Immunopharmacol ; 78: 106062, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31830621

RESUMO

BACKGROUND: Alcoholic liver disease (ALD) presents one of the leading causes of cirrhosis worldwide. We have demonstrated that inulin alleviates ALD in mice. However, the exact role of hepatic macrophages in effects of inulin on ALD remains largely unclear. METHODS: In vivo, mice were divided into 4 groups: pair-fed (PF) group (PF/CON), alcohol-fed (AF) group (AF/CON), PF with inulin (INU) group (PF/INU) and AF with INU group (AF/INU). Each group was fed modified Lieber-DeCarli liquid diet with or without alcohol. In vitro, RAW264.7 cell lines were polarized to M1 macrophage (Mψ) or M2 Mψ subsets with lipopolysaccharide (LPS) or interleukin-4 (IL-4) stimulation, respectively. The effects of propionate, butyrate and valeric on macrophage M1/M2 were investigated. RESULTS: The contents of propionate, butyrate and valeric were significantly increased in AF/INU group compared with that in the AF/CON group. M1 Mψ, inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α) in AF/INU group were significantly lower than those in AF/CON group. In contrast, M2 Mψ, arginase-1 (Arg-1), and interleukin-10 (IL-10) were notably increased in AF/INU group. In vitro, sodium propionate, sodium butyrate and sodium valerate can suppress M1 Mψ and increase M2 Mψ polarization. CONCLUSION: In ALD, inulin ameliorates the inflammation via SCFAs-inducing suppression of M1 and facilitation of M2 Mψ, which may potentially contribute to the control of the disease.


Assuntos
Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/imunologia , Inflamação/dietoterapia , Inulina/administração & dosagem , Hepatopatias Alcoólicas/dietoterapia , Macrófagos/imunologia , Administração Oral , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Etanol/administração & dosagem , Etanol/toxicidade , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/imunologia , Fezes/química , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/microbiologia , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/patologia , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Camundongos , Cultura Primária de Células , Células RAW 264.7
15.
J Agric Food Chem ; 68(5): 1237-1247, 2020 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-31722525

RESUMO

Alcoholic liver injury, known as the most general result of chronic alcohol intake, is induced by inflammatory responses, which is activated by intestine-derived endotoxins formed from intestinal dysbiosis. The hepatoprotective activity of rice bran phenolic extract (RBPE) on ethanol-fed mice was investigated for the first time in this study, and the underlying mechanism was explored from gut microbiota, barrier function, and hepatic inflammation. Mice were fed an alcohol-containing liquid diet alone or in mixture with RBPE for 8 weeks. RBPE treatment mitigated ethanol-induced liver damage, evidenced by the declined lipid profile levels and hepatic function markers. Moreover, ethanol intake induced intestinal microbiota dysbiosis, which was attenuated by RBPE supplementation. RBPE treatment improved the alcohol-induced decrease in the expression of ZO-1, Claudin-1, Claudin-4, and Reg3g, revealing the ameliorative effect of RBPE on intestinal barrier dysfunction. Furthermore, RBPE treatment repressed the alcohol-induced trigger of the hepatic endotoxin-TLR4-NF-κB pathway, followed by the mitigated liver inflammation. The findings indicate that RBPE supplementation ameliorates intestinal microbiota dysbiosis and barrier dysfunction, inactivates the endotoxin-TLR4-NF-κB pathway, and represses inflammatory responses in liver, and therefore, intake of RBPE or brown rice may be an effective way to mitigate alcoholic liver injury.


Assuntos
Disbiose/tratamento farmacológico , Mucosa Intestinal/microbiologia , Hepatopatias Alcoólicas/prevenção & controle , NF-kappa B/imunologia , Oryza/química , Fenóis/administração & dosagem , Extratos Vegetais/administração & dosagem , Receptor 4 Toll-Like/imunologia , Animais , Disbiose/genética , Disbiose/imunologia , Disbiose/microbiologia , Endotoxinas/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/imunologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , Substâncias Protetoras/administração & dosagem , Receptor 4 Toll-Like/genética
16.
Front Immunol ; 10: 2840, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31867007

RESUMO

Hepatic macrophages are key components of the liver immunity and consist of two main populations. Liver resident macrophages, known as Kupffer cells in mammals, are crucial for maintaining normal liver homeostasis. Upon injury, they become activated to release proinflammatory cytokines and chemokines and recruit a large population of inflammatory monocyte-derived macrophages to the liver. During the progression of liver diseases, macrophages are highly plastic and have opposing functions depending on the signaling cues that they receive from the microenvironment. A comprehensive understanding of liver macrophages is essential for developing therapeutic interventions that target these cells in acute and chronic liver diseases. Mouse studies have provided the bulk of our current knowledge of liver macrophages. The emergence of various liver disease models and availability of transgenic tools to visualize and manipulate macrophages have made the teleost zebrafish (Danio rerio) an attractive new vertebrate model to study liver macrophages. In this review, we summarize the origin and behaviors of macrophages in healthy and injured livers in zebrafish. We highlight the roles of macrophages in zebrafish models of alcoholic and non-alcoholic liver diseases, hepatocellular carcinoma, and liver regeneration, and how they compare with the roles that have been described in mammals. We also discuss the advantages and challenges of using zebrafish to study liver macrophages.


Assuntos
Modelos Animais de Doenças , Hepatopatias/imunologia , Macrófagos/fisiologia , Animais , Hematopoese , Hepatopatias Alcoólicas/imunologia , Neoplasias Hepáticas/imunologia , Regeneração Hepática , Peixe-Zebra
17.
Mar Drugs ; 17(10)2019 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-31569771

RESUMO

Fucoxanthin (Fx) is a natural extract from marine seaweed that has strong antioxidant activity and a variety of other bioactive effects. This study elucidated the protective mechanism of Fx on alcoholic liver injury. Administration of Fx was associated with lower pathological effects in liver tissue and lower serum marker concentrations for liver damage induced by alcohol. Fx also alleviated oxidative stress, and lowered the level of oxides and inflammation in liver tissue. Results indicate that Fx attenuated alcohol-induced oxidative lesions and inflammatory responses by activating the nuclear factor erythrocyte-2-related factor 2 (Nrf2)-mediated signaling pathway and down-regulating the expression of the toll-like receptor 4 (TLR4)-mediated nuclear factor-kappa B (NF-κB) signaling pathway, respectively. Our findings suggest that Fx can be developed as a potential nutraceutical for preventing alcohol-induced liver injury in the future.


Assuntos
Suplementos Nutricionais , Inflamação/prevenção & controle , Hepatopatias Alcoólicas/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Xantofilas/administração & dosagem , Animais , Modelos Animais de Doenças , Etanol/efeitos adversos , Humanos , Inflamação/imunologia , Inflamação/patologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Alga Marinha/química , Receptor 4 Toll-Like/metabolismo
18.
Int J Mol Sci ; 20(18)2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31540133

RESUMO

Alcoholic liver disease (ALD), a disorder caused by excessive alcohol intake represents a global health care burden. ALD encompasses a broad spectrum of hepatic injuries including asymptomatic steatosis, alcoholic steatohepatitis (ASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The susceptibility of alcoholic patients to develop ALD is highly variable and its progression to more advanced stages is strongly influenced by several hits (i.e., amount and duration of alcohol abuse). Among them, the intestinal microbiota and its metabolites have been recently identified as paramount in ALD pathophysiology. Ethanol abuse triggers qualitative and quantitative modifications in intestinal flora taxonomic composition, mucosal inflammation, and intestinal barrier derangement. Intestinal hypermeability results in the translocation of viable pathogenic bacteria, Gram-negative microbial products, and pro-inflammatory luminal metabolites into the bloodstream, further corroborating the alcohol-induced liver damage. Thus, the premise of this review is to discuss the beneficial effect of gut microbiota modulation as a novel therapeutic approach in ALD management.


Assuntos
Alcoolismo/microbiologia , Microbioma Gastrointestinal/fisiologia , Hepatopatias Alcoólicas/microbiologia , Alcoolismo/genética , Alcoolismo/imunologia , Alcoolismo/fisiopatologia , Animais , Antibacterianos/efeitos adversos , Ácidos e Sais Biliares/metabolismo , Dieta , Suplementos Nutricionais/microbiologia , Disbiose/imunologia , Disbiose/metabolismo , Transplante de Microbiota Fecal , Hepatócitos/metabolismo , Humanos , Intestinos/microbiologia , Fígado/metabolismo , Fígado/fisiopatologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/metabolismo , Hepatopatias Alcoólicas/fisiopatologia
19.
Front Immunol ; 10: 1328, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31244862

RESUMO

Accumulating clinical and experimental evidences have demonstrated that both innate and adaptive immunity are involved in the pathogenesis of alcoholic liver disease (ALD), in which the role of immunity is to fuel the inflammation and to drive the progression of ALD. Various immune cells are implicated in the pathogenesis of ALD. The activation of innate immune cells induced by alcohol and adaptive immune response triggered by oxidative modification of hepatic constituents facilitate the persistent hepatic inflammation. Meanwhile, the suppressed antigen-presenting capability of various innate immune cells and impaired function of T cells may consequently lead to an increased risk of infection in the patients with advanced ALD. In this review, we summarized the significant recent findings of immune cells participating in ALD. The pathways and molecules involved in the regulation of specific immune cells, and novel mediators protecting the liver from alcoholic injury via affecting these cells are particularly highlighted. This review aims to update the knowledge about immunity in the pathogenesis of ALD, which may facilitate to enhancement of currently available interventions for ALD treatment.


Assuntos
Inflamação/imunologia , Hepatopatias Alcoólicas/imunologia , Linfócitos/imunologia , Imunidade Adaptativa , Humanos , Estresse Oxidativo
20.
Front Immunol ; 10: 563, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30972062

RESUMO

Non-alcoholic fatty liver disease (NAFLD) and Alcoholic Liver Disease (ALD) are major causes of liver-related morbidity and mortality and constitute important causes of liver transplantation. The spectrum of the liver disease is wide and includes isolated steatosis, steatohepatitis, and cirrhosis. The treatment of NAFLD and ALD remains, however, an unmet need, and therefore it is a public health priority to develop effective treatments for these diseases. Alcoholic and non-alcoholic liver disease share common complex pathogenetic pathways that involve different organs and systems beyond the liver, including the gut, the adipose tissue, and the immune system, which cross-talk to generate damage. Myeloid-derived cells have been widely studied in the setting of NAFLD and ALD and are implicated at different levels in the onset and progression of this disease. Among these cells, monocytes and macrophages have been found to be involved in the induction of inflammation and in the progression to fibrosis, both in animal models and clinical studies and they have become interesting potential targets for the treatment of both NAFLD and ALD. The different mechanisms by which these cells can be targeted include modulation of Kupffer cell activation, monocyte recruitment in the liver and macrophage polarization and differentiation. Evidence from preclinical studies and clinical trials (some of them already in phase II and III) have shown encouraging results in ameliorating steatohepatitis, fibrosis, and the metabolic profile, individuating promising candidates for the pharmacological treatment of these diseases. The currently available results of myeloid-derived cells targeted treatments in NAFLD and ALD are covered in this review.


Assuntos
Hepatopatias Alcoólicas , Monócitos , Animais , Modelos Animais de Doenças , Humanos , Células de Kupffer/imunologia , Células de Kupffer/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/patologia , Hepatopatias Alcoólicas/terapia , Monócitos/imunologia , Monócitos/patologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Hepatopatia Gordurosa não Alcoólica/patologia , Hepatopatia Gordurosa não Alcoólica/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA