Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 212(6): 974-981, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38251917

RESUMO

Activation and clonal expansion of the Ag-specific adaptive immune response in the draining lymph node is essential to clearing influenza A virus infections. Activation sufficient for virus clearance is dependent on the lymph node's architectural organization that is maintained by stromal cells, chiefly fibroblastic reticular cells. During an analysis of influenza A virus clearance in leptin receptor knockout (DB/DB) mice, we observed that the DB/DB mice have markedly reduced numbers of lymph node fibroblastic reticular cells at the steady state. The reduction in lymph node fibroblastic reticular cells resulted in abnormal lymph node organization and diminished numbers of adaptive immune cells in the lymph nodes under homeostatic conditions. As a consequence, the DB/DB mice were impaired in their ability to generate an effective influenza-specific adaptive immune response, which prevented virus clearance. Using leptin receptor mutant mice with point mutations at distinct signaling sites in the leptin receptor, we were able to link the leptin receptor's signaling domain tyrosine 985, which does not contribute to obesity, to lymph node fibroblastic reticular cell development and function. These results demonstrate a novel role for leptin receptor signaling in regulating lymph node development in a manner that is crucial to the generation of Ag-specific adaptive immune responses.


Assuntos
Imunidade Adaptativa , Receptores para Leptina , Camundongos , Animais , Receptores para Leptina/genética , Linfonodos , Transdução de Sinais , Camundongos Endogâmicos C57BL , Leptina
2.
Front Endocrinol (Lausanne) ; 14: 1150118, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37274349

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is an important public health problem with growing numbers of NAFLD patients worldwide. Pathological conditions are different in each stage of NAFLD due to various factors. Preclinical and clinical studies provide evidence for a crucial role of immune cells in NAFLD progression. Liver-resident macrophages, kupffer cells (KCs), and monocytes-derived macrophages are the key cell types involved in the progression of NAFLD, non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). Their unique polarization contributes to the progression of NAFLD. KCs are phagocytes with self-renewal abilities and play a role in regulating and maintaining homeostasis. Upon liver damage, KCs are activated and colonized at the site of the damaged tissue. The secretion of inflammatory cytokines and chemokines by KCs play a pivotal role in initiating NAFLD pathogenesis. This review briefly describes the role of immune cells in the immune system in NAFLD, and focuses on the pathological role and molecular pathways of KCs and recruited macrophages. In addition, the relationship between macrophages and insulin resistance is described. Finally, the latest therapeutics that target KCs and macrophages are summarized for the prevention and treatment of NAFLD.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Macrófagos/metabolismo
3.
Clin Mol Hepatol ; 29(1): 65-76, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35957546

RESUMO

Hepatitis C virus (HCV) infection is remarkably efficient in establishing viral persistence, leading to the development of liver cirrhosis and hepatocellular carcinoma (HCC). Direct-acting antiviral agents (DAAs) are promising HCV therapies to clear the virus. However, recent reports indicate potential increased risk of HCC development among HCV patients with cirrhosis following DAA therapy. CD8+ T-cells participate in controlling HCV infection. However, in chronic hepatitis C patients, severe CD4+ and CD8+ T-cell dysfunctions have been observed. This suggests that HCV may employ mechanisms to counteract or suppress the host T-cell responses. The primary site of viral replication is within hepatocytes where infection can trigger the expression of costimulatory molecules and the secretion of immunoregulatory cytokines. Numerous studies indicate that HCV infection in hepatocytes impairs antiviral host immunity by modulating the expression of immunoregulatory molecules. Hepatocytes expressing whole HCV proteins upregulate the ligands of programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and transforming growth factor ß (TGF-ß) synthesis compared to those in hepatocytes in the absence of the HCV genome. Importantly, HCV-infected hepatocytes are capable of inducing regulatory CD4+ T-cells, releasing exosomes displaying TGF-ß on exosome surfaces, and generating follicular regulatory T-cells. Recent studies report that the expression profile of exosome microRNAs provides biomarkers of HCV infection and HCV-related chronic liver diseases. A better understanding of the immunoregulatory mechanisms and identification of biomarkers associated with HCV infection will provide insight into designing vaccine against HCV to bypass HCV-induced immune dysregulation and prevent development of HCV-associated chronic liver diseases.


Assuntos
Carcinoma Hepatocelular , Hepatite C Crônica , Hepatite C , Neoplasias Hepáticas , Humanos , Hepacivirus/genética , Antivirais/uso terapêutico , Carcinoma Hepatocelular/patologia , Hepatite C Crônica/complicações , Hepatite C Crônica/patologia , Neoplasias Hepáticas/patologia , Hepatite C/complicações , Hepatócitos/metabolismo , Cirrose Hepática/complicações , Fator de Crescimento Transformador beta/metabolismo , Biomarcadores , Microambiente Tumoral
4.
Viral Immunol ; 35(3): 223-235, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35467430

RESUMO

Lipid accumulation and inflammation act together to induce, sustain, and further development of chronic liver disease. Hepatitis C virus (HCV) infection induces metabolic and immune changes in liver macrophages, promoting lipid accumulation and inflammation that synergize and culminate in the development of steatohepatitis and fibrogenesis. Chronic HCV patients have increased liver macrophages with disruptions in cholesterol metabolism and alterations in inflammatory mediators. While HCV-induced changes in inflammatory mediators are well documented, how HCV triggers metabolic change in macrophages is unknown. In this report, we examined the mechanism of macrophage sensing of HCV to cause metabolic impairment and subsequent immune dysfunction. We demonstrate that HCV protein and RNA kinetics in macrophages are distinct from hepatocytes. In macrophages, HCV RNAs and protein accumulate rapidly after exposure but internalized RNAs quickly decline to a low-level set point. Notably, exposure of macrophages to HCV resulted in increased lipids and cholesterol and activation of cholesterol-sensing, immunomodulatory liver X receptors (LXRs). Furthermore, we provide evidence that HCV RNA accumulation in macrophages occurs through scavenging receptors. These results suggest that HCV released from infected hepatocytes stimulates accumulation of lipids and activation of LXR in macrophages contributing to metabolic changes involved in HCV-induced chronic liver disease. Our results provide novel insight into mechanisms through which impaired lipid metabolism in macrophages associated with HCV infection promotes development of liver steatohepatitis and fibrosis.


Assuntos
Fígado Gorduroso , Hepatite C Crônica , Hepatite C , Colesterol/metabolismo , Hepacivirus , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos , Macrófagos , RNA/metabolismo , Receptores Depuradores/metabolismo
5.
Front Immunol ; 13: 842535, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185933

RESUMO

Myeloid-derived suppressor cells (MDSCs) are generated under biological stress such as cancer, inflammatory tissue damage, and viral infection. In recent years, with occurrence of global infectious diseases, new discovery on MDSCs functions has been significantly expanded during viral infection and COVID-19. For a successful viral infection, pathogens viruses develop immune evasion strategies to avoid immune recognition. Numerous viruses induce the differentiation and expansion of MDSCs in order to suppress host immune responses including natural killer cells, antigen presenting cells, and T-cells. Moreover, MDSCs play an important role in regulation of immunopathogenesis by balancing viral infection and tissue damage. In this review article, we describe the overview of immunomodulation and genetic regulation of MDSCs during viral infection in the animal model and human studies. In addition, we include up-to-date review of role of MDSCs in SARS-CoV-2 infection and COVID-19. Finally, we discuss potential therapeutics targeting MDSCs.


Assuntos
Imunomodulação/imunologia , Macrófagos/imunologia , Células Supressoras Mieloides/imunologia , Neutrófilos/imunologia , SARS-CoV-2/imunologia , Animais , COVID-19/imunologia , Modelos Animais de Doenças , Humanos , Evasão da Resposta Imune/imunologia , Macrófagos/citologia , Monócitos/citologia , Monócitos/imunologia , Células Supressoras Mieloides/citologia
6.
J Cell Signal ; 2(3): 221-227, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34671766

RESUMO

Hepatocellular carcinoma (HCC) is a significant cause of cancer mortality worldwide. Chronic hepatic inflammation and fibrosis play a critical role in the development of HCC. Liver fibrosis develops as a result of response to injury such that a persistent and excessive wound healing response induces extracellular matrix (ECM) deposition leading to HCC. PAI-1 is a fibrinolysis inhibitor involved in regulating protein degradation and homeostasis while assisting wound healing. PAI-1 presents increased levels in various diseases such as fibrosis, cancer, obesity and metabolic syndrome. Moreover, PAI-1 has been extensively studied for developing potential therapies against fibrosis. In the present review, we summarize how PAI-1 affects oncogenesis during liver disease progression based on the recently published literatures. Although there are controversies regarding the role of PAI-1 and approaches to treatment, this review suggests that proper manipulation of PAI-1 activity could provide a novel therapeutic option on the development of chronic liver disease via modulation of cancer stem-like cells (CSCs) differentiation.

7.
Hepatology ; 74(6): 3409-3426, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34218459

RESUMO

BACKGROUND AND AIMS: HCV infection is a major risk factor that can lead to chronic liver disease, including fibrosis, cirrhosis, and HCC. Progression of chronic liver disease by HCV infection is caused by a complex intercellular reaction. Especially, exosomes and microRNAs (miRNAs) from HCV-infected hepatocytes play a role in the pathogenesis of liver disease by facilitating cellular communication between parenchymal and nonparenchymal cells. However, the underlying mechanism of secretions of exosome and miRNAs during HCV infection is still open for study. APPROACH AND RESULTS: In this study, we demonstrated a pathway for the release of exosome and exosomal miRNAs through caspase-3/pannexin 1 (Panx1)/P2X4 activation during HCV infection in hepatocytes. We found that HCV infection induced the stimulation of exosome release and activation of the caspase-3/Panx1/P2X4 pathway in Huh7.5.1 cells. In addition, miR-122 and miR-146a levels in extracellular exosomes from HCV-infected cells were dramatically increased whereas intracellular miR122 and miR-146a expression had no large changes. Notably, secretions of exosomes and exosomal miRNAs were decreased by inhibition of caspase 3, Panx1, and P2X4 whereas inhibition of ROCK-1 cleavage did not affect these during HCV infection in Huh7.5.1 cells. CONCLUSIONS: These results suggested that HCV infection caused secretions of exosomes and exosomal miRNAs dependent on the caspase 3/Panx1/P2X4 pathway. Our study provides a possible therapeutic intervention using Panx1 suppression for liver disease development mediated by exosomes from HCV-infected hepatocytes.


Assuntos
Conexinas/metabolismo , Hepatite C Crônica/genética , Hepatócitos/virologia , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Purinérgicos P2X4/metabolismo , Western Blotting , Linhagem Celular , Exossomos/metabolismo , Citometria de Fluxo , Hepatócitos/metabolismo , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real
8.
J Virol ; 95(10)2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33627392

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is a critical factor that regulates protein synthesis and degradation. The increased PAI-1 levels are detectable in the serum of patients with chronic hepatitis C virus (HCV) liver disease. The differentiation state and motility of HCV-induced cancer stem-like cells (CSC) play a major role in severe liver disease progression. However, the role of PAI-1 in the pathological process of chronic liver diseases remains unknown. In this study, we determined how PAI-1 affects the differentiation of CSC state in hepatocytes upon HCV infection. We found that HCV infection induced the expression of PAI-1 while decreasing miR-30c expression in Huh7.5.1 cells. Similar results were obtained from isolated hepatocytes from humanized liver mice after HCV infection. Moreover, decreased miR-30c expression in HCV-infected hepatocytes was associated with the increased levels of PAI-1 mRNA and protein. Notably, the increased PAI-1 levels resulted in the activation of Protein Kinase B/AKT, a major mediator of cell proliferation, in HCV-infected hepatocytes along with the increased expression of CSC markers such as Human Differentiated Protein (CD) 133, Epithelial cell adhesion molecule (EpCAM), Octamer 4 (Oct4), Nanog, Cyclin D1, and MYC. Moreover, blockade of PAI-1 activity by miR-30c mimic and anti-PAI-1 mAb abrogated the AKT activation with decreased expression of CSC markers. Our findings suggest that HCV infection induces the CSC state via PAI-1-mediated AKT activation in hepatocytes. It implicates that the manipulation of PAI-1 activity could provide potential therapeutics to prevent the development of HCV-associated chronic liver diseases.IMPORTANCEThe progression of chronic liver disease by HCV infection is considered a major risk factor for hepatocellular carcinoma (HCC), one of the major causes of death from cancer. Recent studies have demonstrated that increased CSC properties in HCV-infected hepatocytes are associated with the progression of HCC. Since proteins and miRNAs production by HCV-infected hepatocytes can play various roles in physiological processes, investigating these factors can potentially lead to new therapeutic targets. However, the mechanism of HCV associated progression of hepatocytes to CSC remains unclear. Here we identify the roles of PAI-1 and miR-30c in the progression of CSC during HCV infection in hepatocytes. Our data shows that increased secretion of PAI-1 following HCV infection promotes this CSC state and activation of AKT. We report that the inhibition of PAI-1 by miR-30c mimic reduces HCV associated CSC properties in hepatocytes. Taken together, targeting this interaction of secreted PAI-1 and miR-30c in HCV-infected hepatocytes may provide a potential therapeutic intervention against the progression to chronic liver diseases and HCC.

9.
Pharmaceutics ; 12(4)2020 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-32316169

RESUMO

The number of therapeutic peptides for human treatment is growing rapidly. However, their development faces two major issues: the poor yield of large peptides from conventional solid-phase synthesis, and the intrinsically short serum half-life of peptides. To address these issues, we investigated a platform for the production of a recombinant therapeutic peptide with an extended serum half-life involving the site-specific conjugation of human serum albumin (HSA). HSA has an exceptionally long serum half-life and can be used to extend the serum half-lives of therapeutic proteins and peptides. We used glucagon-like-peptide 1 (GLP-1) as a model peptide in the present study. A "clickable" non-natural amino acid-p-azido-l-phenylalanine (AzF)-was incorporated into three specific sites (V16, Y19, and F28) of a GLP-1 variant, followed by conjugation with HSA through strain-promoted azide-alkyne cycloaddition. All three HSA-conjugated GLP-1 variants (GLP1_16HSA, GLP1_19HSA, and GLP1_28HSA) exhibited comparable serum half-lives in vivo. However, the three GLP1_HSA variants had different in vitro biological activities and in vivo glucose-lowering effects, demonstrating the importance of site-specific HSA conjugation. The platform described herein could be used to develop other therapeutic peptides with extended serum half-lives.

10.
J Biol Chem ; 294(40): 14591-14602, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31375561

RESUMO

Zika virus (ZIKV)3 is an enveloped, single-stranded, positive-sense RNA virus of the Flaviviridae family that has emerged as a public health threat because of its global transmission and link to microcephaly. Currently there is no vaccine for this virus. Conversion of cholesterol to 25-hydroxycholesterol by cholesterol 25-hydroxylase (CH25H) has been shown to have broad antiviral properties. However, the molecular basis of induction of CH25H in humans is not known. Elucidation of signaling and transcriptional events for induction of CH25H expression is critical for designing therapeutic antiviral agents. In this study, we show that CH25H is induced by ZIKV infection or Toll-like receptor stimulation. Interestingly, CH25H is induced by pro-inflammatory cytokines, including IL-1ß, tumor necrosis factor α, and IL-6, and this induction depends on the STAT1 transcription factor. Additionally, we observed that cAMP-dependent transcription factor (ATF3) weakly binds to the CH25H promoter, suggesting cooperation with STAT1. However, ZIKV-induced CH25H was independent of type I interferon. These findings provide important information for understanding how the Zika virus induces innate inflammatory responses and promotes the expression of anti-viral CH25H protein.


Assuntos
Fator 3 Ativador da Transcrição/genética , Fator de Transcrição STAT1/genética , Esteroide Hidroxilases/genética , Infecção por Zika virus/genética , Zika virus/genética , Antivirais/química , Antivirais/metabolismo , Citocinas/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Regulação Enzimológica da Expressão Gênica , Humanos , Inflamação/enzimologia , Inflamação/genética , Inflamação/virologia , Interferon Tipo I/genética , Interleucina-1beta/genética , Interleucina-6/genética , Macrófagos/virologia , Esteroide Hidroxilases/química , Receptores Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Replicação Viral/genética , Zika virus/patogenicidade , Infecção por Zika virus/enzimologia , Infecção por Zika virus/virologia
11.
Methods Mol Biol ; 1911: 453-458, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30593645

RESUMO

Hepatitis C virus-mediated immune suppression is an underlying feature leading to the establishment of viral persistence and chronic infection. In particular, HCV core protein has been shown to exhibit significant immunosuppressive activity of T cells and antigen presenting cells. Using an HCV core transgenic mouse system, in which liver hepatocytes express core protein, it is possible to study the effects of core-mediated immune suppression in vivo during viral infection. In this protocol, we describe the procedures for evaluating antigen-specific CD8+ T cell responses in response to recombinant adenovirus infection in HCV core transgenic mice.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Hepacivirus/imunologia , Hepatite C/imunologia , Adenoviridae/genética , Animais , Separação Celular/instrumentação , Separação Celular/métodos , Centrifugação com Gradiente de Concentração/instrumentação , Centrifugação com Gradiente de Concentração/métodos , Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Vetores Genéticos/genética , Hepatite C/virologia , Hepatócitos , Leucócitos Mononucleares , Fígado/citologia , Fígado/imunologia , Fígado/virologia , Camundongos , Camundongos Transgênicos , Cultura Primária de Células/instrumentação , Cultura Primária de Células/métodos , Transdução Genética/instrumentação , Transdução Genética/métodos , Proteínas do Core Viral/imunologia
13.
Hepatology ; 67(1): 71-85, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28749570

RESUMO

Hepatitis C virus (HCV) is a global health concern that can cause severe liver disease, such as cirrhosis and hepatocellular carcinoma. Control of HCV requires vigorous T-cell responses, yet CD4+ T cells in chronic HCV patients are dysfunctional. T follicular regulatory (Tfr) cells are a subset of regulatory T cells that suppress T follicular helper (Tfh) cells and the generation of high affinity antibody-producing B cells. In this study, we examined the accumulation of Tfr cells in the liver compartment during chronic HCV infection and defined the cellular and molecular mechanisms underlying their expansion. Our analysis revealed a substantial population of Tfr cells in livers of chronic HCV patients that is absent in liver tissues from nonviral hepatitis or healthy subjects. Coculture of PBMCs from healthy subjects with HCV-infected hepatoma cells resulted in preferential expansion of circulating Tfr cells, leading to suppression of Tfh cells. Additionally, coculture of tonsillar cells with infected hepatoma cells lead to an expansion of germinal center Tfr. Notably, expansion was mediated by transforming growth factor beta (TGF-ß)-containing exosomes released from HCV-infected hepatocytes given that blockade of exosome-associated TGF-ß or inhibition of exosome release abrogated Tfr expansion. CONCLUSION: These results show that liver-derived exosomes play a pivotal role in the accumulation of Tfr cells, likely leading to suppression of Tfh responses in HCV-infected patients. Our study identifies a novel pathway in which HCV infection in hepatocytes exacerbates Tfr cell responses to subvert antiviral immunity. (Hepatology 2018;67:71-85).


Assuntos
Proliferação de Células/fisiologia , Exossomos/imunologia , Hepatite C/imunologia , Hepatócitos/imunologia , Linfócitos T Reguladores/imunologia , Biópsia por Agulha , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Exossomos/metabolismo , Citometria de Fluxo , Hepacivirus/imunologia , Hepatite C/patologia , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Estatísticas não Paramétricas , Linfócitos T Reguladores/metabolismo
14.
Sci Rep ; 7(1): 18041, 2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29269881

RESUMO

Extension of the serum half-life is an important issue in developing new therapeutic proteins and expanding applications of existing therapeutic proteins. Conjugation of fatty acid, a natural human serum albumin ligand, to a therapeutic protein/peptide was developed as a technique to extend the serum half-life in vivo by taking advantages of unusually long serum half-life of human serum albumin (HSA). However, for broad applications of fatty acid-conjugation, several issues should be addressed, including a poor solubility of fatty acid and a substantial loss in the therapeutic activity. Therefore, herein we systematically investigate the conditions and components in conjugation of fatty acid to a therapeutic protein resulting in the HSA binding capacity without compromising therapeutic activities. By examining the crystal structure and performing dye conjugation assay, two sites (W160 and D112) of urate oxidase (Uox), a model therapeutic protein, were selected as sites for fatty acid-conjugation. Combination of site-specific incorporation of a clickable p-azido-L-phenylalanine to Uox and strain-promoted azide-alkyne cycloaddition allowed the conjugation of fatty acid (palmitic acid analog) to Uox with the HSA binding capacity and retained enzyme activity. Deoxycholic acid, a strong detergent, greatly enhanced the conjugation yield likely due to the enhanced solubility of palmitic acid analog.


Assuntos
Ácidos Graxos/metabolismo , Albumina Sérica Humana/metabolismo , Sítios de Ligação , Reação de Cicloadição , Humanos , Ligação Proteica
15.
Front Immunol ; 8: 301, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28373874

RESUMO

NK cells represent an important first line of defense against viral infection and cancer and are also involved in tissue homeostasis. Studies of NK cell activation in the last decade have revealed that they are able to respond to the inflammatory stimuli evoked by tissue damage and contribute to both progression and resolution of diseases. Exacerbation of the inflammatory response through interactions between immune effector cells facilitates the progression of non-alcoholic fatty liver disease (NAFLD) into steatosis, cirrhosis, and hepatocellular carcinoma (HCC). When hepatic damage is incurred, macrophage activation is crucial for initiating cross talk with neighboring cells present in the liver, including hepatocytes and NK cells, and the importance of this interaction in shaping the immune response in liver disease is increasingly recognized. Inflicted structural damage can be in part regenerated via the process of self-limiting fibrosis, though persistent hepatic damage will lead to chronic fibrosis and loss of tissue organization and function. The cytotoxic activity of NK cells plays an important role in inducing hepatic stellate cell apoptosis and thus curtailing the progression of fibrosis. Alternatively, in some diseases, such as HCC, NK cells may become dysregulated, promoting an immunosuppressive state where tumors are able to escape immune surveillance. This review describes the current understanding of the contributions of NK cells to tissue inflammation and metabolic liver diseases and the ongoing effort to develop therapeutics that target the immunoregulatory function of NK cells.

16.
J Immunol ; 198(7): 2589-2601, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28219886

RESUMO

Glomerular damage mediated by glomerulus-infiltrating myeloid-derived cells is a key pathogenic event in lupus nephritis (LN), but the process is poorly understood. Confocal microscopy of kidney sections and flow cytometry analysis of glomerular cells from magnetic bead-purified glomeruli have identified glomerulus-infiltrating leukocyte populations in NZM2328 (NZM) lupus-prone mice with spontaneous chronic glomerulonephritis (GN) and anti-glomerular basement membrane-induced nephritis. The occurrence of a major glomerulus-infiltrating CD11b+F4/80-I-A- macrophage population exhibiting the markers programmed death ligand-1 (PD-L1), Mac-2, and macrophage mannose receptor (CD206) and producing Klf4, Il10, Retnla, Tnf, and Il6 mRNA, which are known to be expressed by alternatively activated (M2b) macrophages, correlated with proteinuria status. In NZM mice with spontaneous LN, glomerular macrophage infiltration is predominant. CD11b+F4/80-I-A- intraglomerular macrophages and polymorphonuclear neutrophils (PMN) are important in inducing GN, as anti-CD11b and -ICAM-1 mAb inhibited both proteinuria and macrophage and PMN infiltration. The predominant and high expression of PD-L1 by CD11b+F4/80-I-A- glomerular macrophages in kidneys of mice with GN and the inhibition of proteinuria by anti-PD-L1 mAb supported the pathogenic role of these macrophages but not the PD-L1- PMN in GN development and in inducing podocyte damage. In NZM mice with spontaneous chronic GN and severe proteinuria, few glomerulus-infiltrating PMN were found, leaving macrophages and, to a less extent, dendritic cells as the major infiltrating leukocytes. Taken together, these data support the important pathogenic effect of CD11b+F4/80-I-A- M2b-like glomerulus-infiltrating macrophages in LN and reinforce macrophages as a promising target for GN treatment.


Assuntos
Glomérulos Renais/imunologia , Nefrite Lúpica/imunologia , Macrófagos/imunologia , Animais , Antígeno B7-H1/imunologia , Células da Medula Óssea/imunologia , Separação Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Glomérulos Renais/patologia , Fator 4 Semelhante a Kruppel , Nefrite Lúpica/patologia , Antígeno de Macrófago 1/imunologia , Macrófagos/patologia , Camundongos , Camundongos Mutantes , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real
17.
J Immunol ; 198(6): 2341-2351, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28159899

RESUMO

Cross-presentation is a modular series of intracellular events dictating the internalization and subsequent MHC class I (MHC I) display of extracellular Ags. This process has been defined in dendritic cells and plays a fundamental role in the induction of CD8+ T cell immunity during viral, intracellular bacterial, and antitumor responses. Herein, acute viral infection of murine liver with adenovirus, a model for intrahepatic cross-presentation, confirms hepatocytes directly contribute to cross-presentation of Ags and priming the pool of naive CD8+ T cells within the liver microenvironment. Processing of soluble and cell-associated Ags into peptide displayed by MHC I is however defective in hepatocytes lacking collectrin, an intracellular chaperone protein that localizes within the endoplasmic reticulum-Golgi intermediate compartment. Loss of hepatic collectrin expression leads to the diminished cross-priming and expansion of cytolytic antiviral CD8+ T cells. This study demonstrates that collectrin positively regulates processing of engulfed Ags into MHC I:peptide complexes within hepatocytes. Collectrin-mediated cross-presentation supports intrahepatic adaptive antiviral immune responses and may lead to insights into the nature of how the liver acts as a primary site of CD8+ T cell activation.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada , Hepatócitos/imunologia , Fígado/imunologia , Glicoproteínas de Membrana/metabolismo , Doença Aguda , Animais , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/virologia , Espaço Extracelular/imunologia , Hepatócitos/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Fígado/virologia , Ativação Linfocitária/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Solubilidade , Quimeras de Transplante
18.
J Leukoc Biol ; 101(1): 329-338, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27493244

RESUMO

The liver contains 2 transcriptionally distinct group 1 ILC subsets: CD49a+ ILC1s and CD49b+ NK cells. However, little is known about how group 1 ILCs contribute to hepatic immune responses. Therefore, we characterized murine liver-resident group 1 ILCs and found that CD49a+ ILC1s express high levels of the inhibitory receptor NKG2A and localize near DCs in perivascular spaces surrounding the portal triads. Upon hepatic viral infection, NKG2A signaling in group 1 ILCs, especially in CD49a+ ILC1s, inhibits CXCL9 expression required for robust accumulation of IFN-γ+CD49b+ NK cells. As a consequence, NKG2A-/- mice showed increased numbers of IFN-γ-producing NK cells that preferentially activate liver CD103+ DCs, leading to the sustained proliferation of adoptively transferred, virus-specific CD8+ T cells. Collectively, these data suggest that group 1 ILCs play a role in maintaining the liver as a tolerogenic site by limiting the recruitment of peripheral NK cells during the early phase of viral infection. Furthermore, our findings implicate that the inhibition of NKG2A signaling on group 1 ILCs may be a novel vaccine strategy to induce robust CD8+ T cell responses against persistent liver pathogens.


Assuntos
Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Imunidade Inata , Fígado/citologia , Linfócitos/citologia , Adenoviridae/metabolismo , Animais , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Contagem de Células , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Quimiocina CXCL9/biossíntese , Fatores Quimiotáticos/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Epitopos/imunologia , Feminino , Imunidade Inata/efeitos dos fármacos , Cadeias alfa de Integrinas/metabolismo , Integrina alfa1/metabolismo , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Subfamília C de Receptores Semelhantes a Lectina de Células NK/deficiência , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo
19.
Hepatology ; 65(1): 32-43, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27770558

RESUMO

The liver maintains an immunologically tolerant environment as a result of continuous exposure to food and bacterial constituents from the digestive tract. Hepatotropic pathogens can take advantage of this niche and establish lifelong chronic infections causing hepatic fibrosis and hepatocellular carcinoma. Macrophages (Mϕ) play a critical role in regulation of immune responses to hepatic infection and regeneration of tissue. However, the factors crucial for Mϕ in limiting hepatic inflammation or resolving liver damage have not been fully understood. In this report, we demonstrate that expression of C-type lectin receptor scavenger receptor-AI (SR-AI) is crucial for promoting M2-like Mϕ activation and polarization during hepatic inflammation. Liver Mϕ uniquely up-regulated SR-AI during hepatotropic viral infection and displayed increased expression of alternative Mϕ activation markers, such as YM-1, arginase-1, and interleukin-10 by activation of mer receptor tyrosine kinase associated with inhibition of mammalian target of rapamycin. Expression of these molecules was reduced on Mϕ obtained from livers of infected mice deficient for the gene encoding SR-AI (msr1). Furthermore, in vitro studies using an SR-AI-deficient Mϕ cell line revealed impeded M2 polarization and decreased phagocytic capacity. Direct stimulation with virus was sufficient to activate M2 gene expression in the wild-type (WT) cell line, but not in the knockdown cell line. Importantly, tissue damage and fibrosis were exacerbated in SR-AI-/- mice following hepatic infection and adoptive transfer of WT bone-marrow-derived Mϕ conferred protection against fibrosis in these mice. CONCLUSION: SR-AI expression on liver Mϕ promotes recovery from infection-induced tissue damage by mediating a switch to a proresolving Mϕ polarization state. (Hepatology 2017;65:32-43).


Assuntos
Hepatite/etiologia , Cirrose Hepática/etiologia , Ativação de Macrófagos , Receptores Depuradores Classe A/biossíntese , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL
20.
PLoS One ; 11(6): e0156996, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280294

RESUMO

Hepatitis C is a chronic liver disease that contributes to progressive metabolic dysfunction. Infection of hepatocytes by hepatitis C virus (HCV) results in reprogramming of hepatic and serum lipids. However, the specific contribution of these distinct pools of lipids to HCV infection remains ill defined. In this study, we investigated the role of hepatic lipogenesis in HCV infection by targeting the rate-limiting step in this pathway, which is catalyzed by the acetyl-CoA carboxylase (ACC) enzymes. Using two structurally unrelated ACC inhibitors, we determined that blockade of lipogenesis resulted in reduced viral replication, assembly, and release. Supplementing exogenous lipids to cells treated with ACC inhibitors rescued HCV assembly with no effect on viral replication and release. Intriguingly, loss of viral RNA was not recapitulated at the protein level and addition of 2-bromopalmitate, a competitive inhibitor of protein palmitoylation, mirrored the effects of ACC inhibitors on reduced viral RNA without a concurrent loss in protein expression. These correlative results suggest that newly synthesized lipids may have a role in protein palmitoylation during HCV infection.


Assuntos
Ácidos Graxos/metabolismo , Hepacivirus/fisiologia , Hepatite C/virologia , Hepatócitos/metabolismo , Lipogênese/fisiologia , Acetil-CoA Carboxilase/antagonistas & inibidores , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Hepatite C/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , RNA Viral/genética , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...