Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Science ; 381(6662): eabq5202, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37676943

RESUMO

Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.


Assuntos
Infecções Transmitidas por Sangue , Células Gigantes , Células de Kupffer , Cirrose Hepática , Animais , Humanos , Camundongos , Células Gigantes/imunologia , Células Gigantes/microbiologia , Células de Kupffer/imunologia , Células de Kupffer/microbiologia , Cirrose Hepática/imunologia , Cirrose Hepática/microbiologia , Cirrose Hepática/patologia , Infecções Transmitidas por Sangue/imunologia , Modelos Animais de Doenças
2.
Front Immunol ; 12: 633324, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262558

RESUMO

In the process of microbial invasion, the inflammation reaction is induced to eliminate the pathogen. However, un-controlled or un-resolved inflammation can lead to tissue damage and death of the host. MicroRNAs (miRNAs) are the signaling regulators that prevent the uncontrolled progress of an inflammatory response. Our previous work strongly indicated that miR-142a-3p is related to the immune regulation in grass carp. In the present study, we found that the expression of miR-142a-3p was down-regulated after infection by Aeromonas hydrophila. tnfaip2 and glut3 were confirmed as be the target genes of miR-142a-3p, which were confirmed by expression correlation analysis, gene overexpression, and dual luciferase reporter assay. The miR-142a-3p can reduce cell viability and stimulate cell apoptosis by targeting tnfaip2 and glut3. In addition, miR-142a-3p also regulates macrophage polarization induced by A. hydrophila. Our results suggest that miR-142a-3p has multiple functions in host antibacterial immune response. Our research provides further understanding of the molecular mechanisms between miRNAs and their target genes, and provides a new insights for the development of pro-resolution strategies for the treatment of complex inflammatory diseases in fish.


Assuntos
Apoptose/genética , Carpas/genética , Citocinas/genética , Transportador de Glucose Tipo 3/genética , Infecções por Bactérias Gram-Negativas/veterinária , Macrófagos/fisiologia , MicroRNAs/genética , Aeromonas hydrophila/imunologia , Aeromonas hydrophila/patogenicidade , Animais , Carpas/imunologia , Carpas/microbiologia , Células Cultivadas , Citocinas/classificação , Infecções por Bactérias Gram-Negativas/imunologia , Inflamação/imunologia , Rim/citologia , Rim/microbiologia , Células de Kupffer/microbiologia , Ativação de Macrófagos , MicroRNAs/classificação , Transdução de Sinais
3.
Front Immunol ; 12: 624360, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841405

RESUMO

The gut-liver axis has been increasingly recognized as a major autoimmunity modulator. However, the implications of intestinal barrier in the pathogenesis of autoimmune hepatitis (AIH) remain elusive. Here, we investigated the functional role of gut barrier and intestinal microbiota for hepatic innate immune response in AIH patients and murine models. In this study, we found that AIH patients displayed increased intestinal permeability and pronounced RIP3 activation of liver macrophages. In mice models, intestinal barrier dysfunction increased intestinal bacterial translocation, thus amplifying the hepatic RIP3-mediated innate immune response. Furthermore, GSK872 dampened RIP3 activation and ameliorated the activation and accumulation of liver macrophages in vitro and in vivo experiments. Strikingly, broad-spectrum antibiotic ablation significantly alleviated RIP3 activation and liver injury, highlighting the causal role of intestinal microbiota for disease progression. Our results provided a potentially novel mechanism of immune tolerance breakage in the liver via the gut-liver axis. In addition, we also explored the therapeutic and research potentials of regulating the intestinal microbiota for the therapy of AIH.


Assuntos
Microbioma Gastrointestinal , Hepatite Autoimune/enzimologia , Intestinos/microbiologia , Fígado/enzimologia , Ativação de Macrófagos , Macrófagos/enzimologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Idoso , Animais , Antibacterianos/farmacologia , Translocação Bacteriana , Células CACO-2 , Estudos de Casos e Controles , Modelos Animais de Doenças , Disbiose , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Hepatite Autoimune/imunologia , Hepatite Autoimune/microbiologia , Hepatite Autoimune/prevenção & controle , Humanos , Imunidade Inata , Células de Kupffer/enzimologia , Células de Kupffer/imunologia , Células de Kupffer/microbiologia , Fígado/imunologia , Fígado/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Permeabilidade , Células RAW 264.7 , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais
4.
J Clin Invest ; 131(4)2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33320839

RESUMO

Patients with acute liver failure (ALF) have systemic innate immune suppression and increased susceptibility to infections. Programmed cell death 1 (PD-1) expression by macrophages has been associated with immune suppression during sepsis and cancer. We therefore examined the role of the programmed cell death 1/programmed death ligand 1 (PD-1/PD-L1) pathway in regulating Kupffer cell (KC) inflammatory and antimicrobial responses in acetaminophen-induced (APAP-induced) acute liver injury. Using intravital imaging and flow cytometry, we found impaired KC bacterial clearance and systemic bacterial dissemination in mice with liver injury. We detected increased PD-1 and PD-L1 expression in KCs and lymphocyte subsets, respectively, during injury resolution. Gene expression profiling of PD-1+ KCs revealed an immune-suppressive profile and reduced pathogen responses. Compared with WT mice, PD-1-deficient mice and anti-PD-1-treated mice with liver injury showed improved KC bacterial clearance, a reduced tissue bacterial load, and protection from sepsis. Blood samples from patients with ALF revealed enhanced PD-1 and PD-L1 expression by monocytes and lymphocytes, respectively, and that soluble PD-L1 plasma levels could predict outcomes and sepsis. PD-1 in vitro blockade restored monocyte functionality. Our study describes a role for the PD-1/PD-L1 axis in suppressing KC and monocyte antimicrobial responses after liver injury and identifies anti-PD-1 immunotherapy as a strategy to reduce infection susceptibility in ALF.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Inibidores de Checkpoint Imunológico/farmacologia , Células de Kupffer/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Acetaminofen/efeitos adversos , Acetaminofen/uso terapêutico , Adulto , Animais , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/genética , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/microbiologia , Feminino , Humanos , Células de Kupffer/microbiologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia
5.
J Cell Physiol ; 236(6): 4360-4368, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33169399

RESUMO

Protein ubiquitination regulates protein stability, cellular localization, and enzyme activity. Deubiquitinases catalyze the removal of ubiquitin from target proteins and reverse ubiquitination. USP13, a deubiquitinase, has been shown to regulate a variety of cellular responses including inflammation; however, the molecular regulation of USP13 has not been demonstrated. In this study, we revealed that USP13 is degraded in response to lipopolysaccharide (LPS) in Kupffer cells. USP13 levels are significantly decreased in inflamed organs, including liver tissues from septic mice. LPS reduces USP13 protein stability, not transcription, in Kupffer cells. Furthermore, LPS increases USP13 polyubiquitination. Inhibition of proteasome, but not lysosome or immunoproteasome, attenuates LPS-induced USP13 degradation, suggesting USP13 degradation is mediated by the ubiquitin-proteasome system. A catalytically inactive form of USP13 exhibits similar degree of degradation compared with USP13 wild-type, suggesting that USP13 degradation is not dependent on its activity. Furthermore, USP13 degradation is dependent on new protein synthesis. Inhibition of c-Jun N-terminal kinase (JNK) attenuates USP13 degradation, indicating that JNK-dependent new protein synthesis is necessary for USP13 degradation. This study reveals a molecular mechanism of regulation of USP13 degradation in Kupffer cells in response to bacterial endotoxin.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células de Kupffer/enzimologia , Sepse/enzimologia , Proteases Específicas de Ubiquitina/metabolismo , Animais , Modelos Animais de Doenças , Ativação Enzimática , Estabilidade Enzimática , Células Hep G2 , Humanos , Células de Kupffer/microbiologia , Células de Kupffer/patologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Células RAW 264.7 , Sepse/induzido quimicamente , Sepse/microbiologia , Sepse/patologia , Transdução de Sinais , Proteases Específicas de Ubiquitina/genética , Ubiquitinação
6.
Front Immunol ; 11: 578654, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33250892

RESUMO

Background and Aims: Mirtazapine is an atypical antidepressant with antagonist activity for serotonin and histamine receptors. Clinical and experimental evidence suggests that, in addition to treating depression, mirtazapine also alters liver innate immunity and suppresses immune-driven hepatic macrophage activation. Liver macrophages, Kupffer cells, represent the largest collection of fixed macrophages in the body and are critical in regulating hepatic immunity. In addition to their capacity to regulate inflammation, Kupffer cells are key sentinels for clearing blood-borne pathogens, preventing their dissemination within the body. This process involves pathogen capture, phagocytosis, and activation-induced killing via reactive oxygen species (ROS) production. Therefore, we speculated that mirtazapine might adversely alter Kupffer cell pathogen-associated activation and killing. Methods: Mice were treated with mirtazapine and time-dependent changes in Kupffer cells were characterized using intravital microscopy. Macrophage and neutrophil responses, bacterial dissemination, and liver damage were assessed following i.v. infection with a pathogenic strain of S. aureus. Results: Mirtazapine rapidly (within 1.5 h) activates Kupffer cells, indicated by a loss of elongated shape with cellular rounding. However, this shape change did not result in impaired pathogen capture function, and, in fact, generated enhanced ROS production in response to S. aureus-induced sepsis. Neutrophil dynamics were altered with reduced cellular recruitment to the liver following infection. Bacterial dissemination post-intravenous administration was not altered by mirtazapine treatment; however, hepatic abscess formation was significantly reduced. Conclusions: Mirtazapine rapidly activates Kupffer cells, associated with preserved bacterial capture functions and enhanced ROS generation capacity. Moreover, these changes in Kupffer cells were linked to a beneficial reduction in hepatic abscess size. In contrast to our initial speculation, mirtazapine may have beneficial effects in sepsis and warrants further exploration.


Assuntos
Antidepressivos/farmacologia , Células de Kupffer/efeitos dos fármacos , Abscesso Hepático/tratamento farmacológico , Fígado/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Mirtazapina/farmacologia , Fagocitose/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/patogenicidade , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Células de Kupffer/metabolismo , Células de Kupffer/microbiologia , Fígado/metabolismo , Fígado/microbiologia , Fígado/patologia , Abscesso Hepático/metabolismo , Abscesso Hepático/microbiologia , Abscesso Hepático/patologia , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Fatores de Tempo
7.
Front Immunol ; 11: 1086, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32612603

RESUMO

While the interactions between HIV and various liver cell populations have been explored, the relevance of these interactions when patients are well-controlled on ART is less clear. Therefore, we focus this perspective on HIV-related alterations that may drive hepatic inflammation and fibrosis in aviremic patients, with a focus on Kupffer cells and Hepatic Stellate Cells. Persistent CD4+ T cell depletion in the gut resulting in increased gut permeability has been postulated to play a role in systemic immune activation in HIV patients. The liver, with its unique location, remains the gatekeeper between the gut and the systemic circulation. The resident liver macrophage, Kupffer cell, is responsible for clearing and responding to these products. We propose that changes in Kupffer cell biology, in the context of HIV infection, creates a mileu that drives hepatic inflammation and fibrosis in response to microbial translocation. Targeting these pathways may be helpful in improving liver-related outcomes in HIV patients.


Assuntos
Infecções por HIV/complicações , Infecções por HIV/imunologia , Hepatite/etiologia , Hepatite/imunologia , Células de Kupffer/imunologia , Cirrose Hepática/etiologia , Cirrose Hepática/imunologia , Microbioma Gastrointestinal/imunologia , HIV-1 , Células Estreladas do Fígado/imunologia , Células Estreladas do Fígado/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Células de Kupffer/microbiologia , Células de Kupffer/virologia , Modelos Imunológicos , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia
8.
Nat Commun ; 11(1): 3535, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669568

RESUMO

Macrophages are professional phagocytes known to play a vital role in controlling Mycobacterium tuberculosis (Mtb) infection and disease progression. Here we compare Mtb growth in mouse alveolar (AMs), peritoneal (PMs), and liver (Kupffer cells; KCs) macrophages and in bone marrow-derived monocytes (BDMs). KCs restrict Mtb growth more efficiently than all other macrophages and monocytes despite equivalent infections through enhanced autophagy. A metabolomics comparison of Mtb-infected macrophages indicates that ornithine and imidazole are two top-scoring metabolites in Mtb-infected KCs and that acetylcholine is the top-scoring in Mtb-infected AMs. Ornithine, imidazole and atropine (acetylcholine inhibitor) inhibit Mtb growth in AMs. Ornithine enhances AMPK mediated autophagy whereas imidazole directly kills Mtb by reducing cytochrome P450 activity. Intranasal delivery of ornithine or imidazole or the two together restricts Mtb growth. Our study demonstrates that the metabolic differences between Mtb-infected AMs and KCs lead to differences in the restriction of Mtb growth.


Assuntos
Autofagia/efeitos dos fármacos , Ornitina/farmacologia , Tuberculose/tratamento farmacológico , Ureia/química , Amônia/química , Animais , Apoptose , Arginase/química , Atropina/farmacologia , Proliferação de Células , Progressão da Doença , Feminino , Imidazóis/farmacologia , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/microbiologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/microbiologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/química , Fosfatidilserinas/química , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/química
9.
Nat Commun ; 10(1): 4566, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31594939

RESUMO

Fungal dissemination into the bloodstream is a critical step leading to invasive fungal infections. Here, using intravital imaging, we show that Kupffer cells (KCs) in the liver have a prominent function in the capture of circulating Cryptococcus neoformans and Candida albicans, thereby reducing fungal dissemination to target organs. Complement C3 but not C5, and complement receptor CRIg but not CR3, are involved in capture of C. neoformans. Internalization of C. neoformans by KCs is subsequently mediated by multiple receptors, including CR3, CRIg, and scavenger receptors, which work synergistically along with C5aR signaling. Following phagocytosis, the growth of C. neoformans is inhibited by KCs in an IFN-γ independent manner. Thus, the liver filters disseminating fungi from circulation via KCs, providing a mechanistic explanation for the enhanced risk of cryptococcosis among individuals with liver diseases, and suggesting a therapeutic strategy to prevent fungal dissemination through enhancing KC functions.


Assuntos
Infecções Fúngicas Invasivas/imunologia , Células de Kupffer/imunologia , Fígado/imunologia , Fagocitose , Animais , Candida albicans/imunologia , Candida albicans/isolamento & purificação , Candida albicans/patogenicidade , Complemento C3/genética , Complemento C3/imunologia , Complemento C3/metabolismo , Cryptococcus neoformans/imunologia , Cryptococcus neoformans/isolamento & purificação , Cryptococcus neoformans/patogenicidade , Modelos Animais de Doenças , Feminino , Humanos , Microscopia Intravital , Infecções Fúngicas Invasivas/sangue , Infecções Fúngicas Invasivas/microbiologia , Células de Kupffer/metabolismo , Células de Kupffer/microbiologia , Fígado/citologia , Fígado/diagnóstico por imagem , Masculino , Camundongos , Camundongos Knockout , Microscopia Confocal , Receptores de Complemento/genética , Receptores de Complemento/imunologia , Receptores de Complemento/metabolismo
10.
Cell Microbiol ; 20(11): e12869, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29904997

RESUMO

Staphylococcus lugdunensis is a commensal bacterium that can cause serious infection suggesting an ability to circumvent aspects of host immunity. We demonstrate here that macrophages fail to kill ingested S. lugdunensis and the bacteria persist for extended periods, without replicating, within mature LAMP-1-positive phagolysosomes. Phagocytosed S. lugdunensis also do not intoxicate host cells in contrast to Staphylococcus aureus. Optimal survival of S. lugdunensis requires O-acetylated peptidoglycan because an oatA mutant, which is more sensitive to killing by lysozyme than wild type, survived to a lesser extent in macrophages. In vitro models of macrophage infection reveal that viable intracellular S. lugdunensis bacteria can be made to grow by pharmacologic perturbation of phagosome function or by phagocyte intoxication by S. aureus toxins. Remarkably, replicating S. lugdunensis is not constrained by LAMP-1 and phosphatidylserine-positive endomembranes, which is distinct from S. aureus that replicates within phagolysosomes. In vivo, S. lugdunensis can also reside in the murine Kupffer cell where the bacteria persist without replicating and require oatA to resist killing in vivo. The intracellular environment of the macrophage represents a niche where S. lugdunensis can exist while protected from extracellular immune factors and may serve as a reservoir from which these bacteria could disseminate.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Evasão da Resposta Imune , Macrófagos/microbiologia , Infecções Estafilocócicas/imunologia , Staphylococcus lugdunensis/patogenicidade , Animais , Toxinas Bacterianas/farmacologia , Células Cultivadas , Feminino , Humanos , Células de Kupffer/microbiologia , Células de Kupffer/patologia , Proteínas de Membrana Lisossomal/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Peptidoglicano/genética , Peptidoglicano/metabolismo , Fagossomos/microbiologia , Células RAW 264.7 , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Staphylococcus aureus/química , Staphylococcus aureus/patogenicidade , Staphylococcus lugdunensis/fisiologia
11.
Immunity ; 47(2): 374-388.e6, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813662

RESUMO

The liver is positioned at the interface between two routes traversed by pathogens in disseminating infection. Whereas blood-borne pathogens are efficiently cleared in hepatic sinusoids by Kupffer cells (KCs), it is unknown how the liver prevents dissemination of peritoneal pathogens accessing its outer membrane. We report here that the hepatic capsule harbors a contiguous cellular network of liver-resident macrophages phenotypically distinct from KCs. These liver capsular macrophages (LCMs) were replenished in the steady state from blood monocytes, unlike KCs that are embryonically derived and self-renewing. LCM numbers increased after weaning in a microbiota-dependent process. LCMs sensed peritoneal bacteria and promoted neutrophil recruitment to the capsule, and their specific ablation resulted in decreased neutrophil recruitment and increased intrahepatic bacterial burden. Thus, the liver contains two separate and non-overlapping niches occupied by distinct resident macrophage populations mediating immunosurveillance at these two pathogen entry points to the liver.


Assuntos
Células de Kupffer/fisiologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Fígado/imunologia , Macrófagos/imunologia , Neutrófilos/imunologia , Peritônio/microbiologia , Animais , Comunicação Celular , Autorrenovação Celular , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Células de Kupffer/microbiologia , Fígado/microbiologia , Fígado/patologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Infiltração de Neutrófilos , Peritônio/patologia
12.
Sci China Life Sci ; 60(11): 1191-1196, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28840534

RESUMO

Gut microbiota play an essential role in shaping immune cell responses. The liver was continuously exposed to metabolic products of intestinal commensal bacterial through portal vein and alteration of gut commensal bateria was always associated with increased risk of liver inflammation and autoimmune disease. Considered as a unique immunological organ, the liver is enriched with a large number of innate immune cells. Herein, we summarize the available literature of gut microbiota in shaping the response of hepatic innate immune cells including NKT cells, NK cells, γδ T cells and Kupffer cells during health and disease. Such knowledge might help to develop novel and innovative strategies for the prevention and therapy of innate immune cell-related liver disease.


Assuntos
Microbioma Gastrointestinal/imunologia , Imunidade Inata , Células de Kupffer/microbiologia , Hepatopatias/imunologia , Fígado/imunologia , Linfócitos/microbiologia , Animais , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Células de Kupffer/imunologia , Hepatopatias/microbiologia , Linfócitos/imunologia , Transdução de Sinais
13.
Methods ; 128: 12-19, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28522327

RESUMO

It is central to the field of bacterial pathogenesis to define how bacteria are killed by phagocytic cells. During phagocytosis, the microbe is localized to the phagolysosome where crucial defense mechanisms such as acidification and production of reactive oxygen species (ROS) are initiated. This process has extensively been studied in vitro, however many resident tissue phagocytes will phenotypically change upon isolation from their natural environment. Therefore, interrogation of phagocytosis and phagosomal function of cells in the context of their natural tissue environment enhances our understanding of the biological process in vivo. This article outlines a real-time intravital microscopy protocol that utilizes fluorescent dyes to study the process of phagocytosis, which reveals acidification and oxidation of individual bacteria inside host cells of living animals. The novelty of this technique exists in use of bacteria that are covalently labelled with the fluorescent dyes Oxyburst and pHrodo, which respectively report on oxidation or acidification. Intravital microscopy is applied to visualize the uptake and subsequent oxidation or acidification of reporter bacteria in the organ of interest. Fluorescently labelled antibodies can be used to counter stain for host immune cells such as neutrophils and macrophages, along with reference stains to identify all bacteria. Although these assays were originally developed to assess the uptake and survival ofStaphylococcus aureusin liver resident macrophages (Kupffer cells), this protocol may be adapted to investigate any bacterium-host cell interaction.


Assuntos
Microscopia Intravital/métodos , Células de Kupffer/microbiologia , Fagossomos/microbiologia , Coloração e Rotulagem/métodos , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Proteínas de Fluorescência Verde/análise , Células de Kupffer/química , Células de Kupffer/fisiologia , Camundongos , Fagossomos/química , Fagossomos/fisiologia , Staphylococcus aureus/química
14.
J Clin Invest ; 127(6): 2249-2261, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28463232

RESUMO

Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.


Assuntos
Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Biespecíficos , Antígenos de Bactérias/imunologia , Carga Bacteriana , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Proteínas do Sistema Complemento/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Células de Kupffer/microbiologia , Pulmão/irrigação sanguínea , Pulmão/microbiologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Microvasos/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose , Proteínas Citotóxicas Formadoras de Poros/imunologia , Infecções por Pseudomonas/imunologia , Receptores Fc/metabolismo
15.
Bull Exp Biol Med ; 162(2): 264-268, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27905039

RESUMO

The dynamics of pathomorphological changes in response to infection with plasmid variants of Yersinia pseudotuberculosis was studied in experimental animals. Variability of cell injuries in pseudotuberculosis histopathology depended on the plasmid-associated virulence of the infection agent. Infection with highly virulent two-plasmid strain pYV48:pVM82 MDa and Y. pseudotuberculosis strain with low virulence with the only plasmid pVM82 MDa led to the development of cell destruction (necrosis and apoptosis) in the target organs. Apoptosis predominated in response to infection by plasmid variant pVM82 MDa with low virulence.


Assuntos
Necrose/patologia , Plasmídeos/metabolismo , Infecções por Yersinia pseudotuberculosis/patologia , Yersinia pseudotuberculosis/patogenicidade , Animais , Animais não Endogâmicos , Apoptose , Hepatócitos/microbiologia , Hepatócitos/patologia , Humanos , Injeções Intraperitoneais , Células de Kupffer/microbiologia , Células de Kupffer/patologia , Fígado/microbiologia , Fígado/patologia , Pulmão/microbiologia , Pulmão/patologia , Linfonodos/microbiologia , Linfonodos/patologia , Masculino , Camundongos , Necrose/microbiologia , Fagócitos/microbiologia , Fagócitos/patologia , Plasmídeos/química , Especificidade da Espécie , Baço/microbiologia , Baço/patologia , Virulência , Yersinia pseudotuberculosis/genética , Yersinia pseudotuberculosis/crescimento & desenvolvimento , Infecções por Yersinia pseudotuberculosis/microbiologia
16.
Toxins (Basel) ; 8(7)2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27376326

RESUMO

Although Staphylococcus aureus is part of the normal body flora, heavy usage of antibiotics has resulted in the emergence of methicillin-resistant strains (MRSA). MRSA can form biofilms and cause indwelling foreign body infections, bacteremia, soft tissue infections, endocarditis, and osteomyelitis. Using an in vitro assay, we screened 173 clinical blood isolates of MRSA and selected 20 high-biofilm formers (H-BF) and low-biofilm formers (L-BF). These were intravenously administered to mice and the general condition of mice, the distribution of bacteria, and biofilm in the liver, lung, spleen, and kidney were investigated. MRSA count was the highest in the liver, especially within Kupffer cells, which were positive for acid polysaccharides that are associated with intracellular biofilm. After 24 h, the general condition of the mice worsened significantly in the H-BF group. In the liver, bacterial deposition and aggregation and the biofilm-forming spot number were all significantly greater for H-BF group than for L-BF. CFU analysis revealed that bacteria in the H-BF group survived for long periods in the liver. These results indicate that the biofilm-forming ability of MRSA is a crucial factor for intracellular persistence, which could lead to chronic infections.


Assuntos
Biofilmes/crescimento & desenvolvimento , Células de Kupffer/microbiologia , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Infecções Estafilocócicas/microbiologia , Animais , Carga Bacteriana , Feminino , Staphylococcus aureus Resistente à Meticilina/metabolismo , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Polissacarídeos Bacterianos/metabolismo , Infecções Estafilocócicas/sangue , Fatores de Tempo , Virulência
17.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G237-45, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27288425

RESUMO

Recent studies suggest that coinhibitory receptors appear to be important in contributing sepsis-induced immunosuppression. Our laboratory reported that mice deficient in programmed cell death receptor (PD)-1 have increased bacterial clearance and improved survival in experimental sepsis induced by cecal ligation and puncture (CLP). In response to infection, the liver clears the blood of bacteria and produces cytokines. Kupffer cells, the resident macrophages in the liver, are strategically situated to perform the above functions. However, it is not known if PD-1 expression on Kupffer cells is altered by septic stimuli, let alone if PD-1 ligation contributes to the altered microbial handling seen. Here we report that PD-1 is significantly upregulated on Kupffer cells during sepsis. PD-1-deficient septic mouse Kupffer cells displayed markedly enhanced phagocytosis and restoration of the expression of major histocompatibility complex II and CD86, but reduced CD80 expression compared with septic wild-type (WT) mouse Kupffer cells. In response to ex vivo LPS stimulation, the cytokine productive capacity of Kupffer cells derived from PD-1-/- CLP mice exhibited a marked, albeit partial, restoration of the release of IL-6, IL-12, IL-1ß, monocyte chemoattractant protein-1, and IL-10 compared with septic WT mouse Kupffer cells. In addition, PD-1 gene deficiency decreased LPS-induced apoptosis of septic Kupffer cells, as indicated by decreased levels of cleaved caspase-3 and reduced terminal deoxynucleotidyl transferase dUTP nick end-labeling-positive cells. Exploring the signal pathways involved, we found that, after ex vivo LPS stimulation, septic PD-1-/- mouse Kupffer cells exhibited an increased Akt phosphorylation and a reduced p38 phosphorylation compared with septic WT mouse Kupffer cells. Together, these results indicate that PD-1 appears to play an important role in regulating the development of Kupffer cell dysfunction seen in sepsis.


Assuntos
Coinfecção/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Sepse/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Células Cultivadas , Coinfecção/genética , Coinfecção/microbiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Células de Kupffer/microbiologia , Fígado/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Fenótipo , Fosforilação , Receptor de Morte Celular Programada 1/deficiência , Receptor de Morte Celular Programada 1/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sepse/genética , Sepse/microbiologia , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Int J Nanomedicine ; 10: 4173-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26170657

RESUMO

BACKGROUND: Zinc oxide engineered nanoparticles (ZnO ENPs) have potential as nanomedicines due to their inherent properties. Studies have described their pulmonary impact, but less is known about the consequences of ZnO ENP interactions with the liver. This study was designed to describe the effects of ZnO ENPs on the liver and Kupffer cells after intravenous (IV) administration. MATERIALS AND METHODS: First, pharmacokinetic studies were conducted to determine the tissue distribution of neutron-activated (65)ZnO ENPs post-IV injection in Wistar Han rats. Then, a noninvasive in vivo method to assess Kupffer cell phagosomal motility was employed using ferromagnetic iron particles and magnetometry. We also examined whether prior IV injection of ZnO ENPs altered Kupffer cell bactericidal activity on circulating Pseudomonas aeruginosa. Serum and liver tissues were collected to assess liver-injury biomarkers and histological changes, respectively. RESULTS: We found that the liver was the major site of initial uptake of (65)ZnO ENPs. There was a time-dependent decrease in tissue levels of (65)Zn in all organs examined, refecting particle dissolution. In vivo magnetometry showed a time-dependent and transient reduction in Kupffer cell phagosomal motility. Animals challenged with P. aeruginosa 24 hours post-ZnO ENP injection showed an initial (30 minutes) delay in vascular bacterial clearance. However, by 4 hours, IV-injected bacteria were cleared from the blood, liver, spleen, lungs, and kidneys. Seven days post-ZnO ENP injection, creatine phosphokinase and aspartate aminotransferase levels in serum were significantly increased. Histological evidence of hepatocyte damage and marginated neutrophils were observed in the liver. CONCLUSION: Administration of ZnO ENPs transiently inhibited Kupffer cell phagosomal motility and later induced hepatocyte injury, but did not alter bacterial clearance from the blood or killing in the liver, spleen, lungs, or kidneys. Our data show that diminished Kupffer cell organelle motion correlated with ZnO ENP-induced liver injury.


Assuntos
Células de Kupffer , Fígado , Nanopartículas Metálicas , Fagossomos/efeitos dos fármacos , Óxido de Zinco , Animais , Células de Kupffer/citologia , Células de Kupffer/efeitos dos fármacos , Células de Kupffer/microbiologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Nanopartículas Metálicas/química , Nanopartículas Metálicas/toxicidade , Ratos , Ratos Wistar , Óxido de Zinco/química , Óxido de Zinco/toxicidade
19.
Dig Dis Sci ; 60(11): 3340-50, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26143342

RESUMO

BACKGROUND AND AIMS: Gut microbiota may be associated with the pathogenesis of nonalcoholic steatohepatitis (NASH). This study aimed to investigate the protective effects and possible mechanisms of Lactobacillus paracasei on NASH. METHODS: Thirty male C57BL/6 mice were randomized into three groups and maintained for 10 weeks: control group (standard chow), NASH model group (high fat + 10 % fructose diet), and the L. paracasei group (NASH model with L. paracasei). Liver histology, serum aminotransferase levels, and hepatic gene expression levels were measured. Intestinal permeability was investigated using urinary (51)Creatinine Ethylenediaminetetraacetic acid ((51)Cr-EDTA) clearance. Total Kupffer cell counts and their composition (M1 vs. M2 Kupffer cells) were measured using flow cytometry with F4/80 and CD206 antibodies. RESULTS: Hepatic fat deposition, serum ALT level, and (51)Cr-EDTA clearance were significantly lower in the L. paracasei group than the NASH group (p < 0.05). The L. paracasei group had lower expression in Toll-like receptor-4 (TLR-4), NADPH oxidase-4 (NOX-4), tumor necrosis factor alpha (TNF-α), monocyte chemotactic protein-1 (MCP-1), interleukin 4 (IL-4), peroxisome proliferator activated receptor gamma (PPAR-γ), and PPAR-δ compared with the NASH group (p < 0.05). The total number of F4/80(+) Kupffer cells was lower in the L. paracasei group than the NASH group. L. paracasei induced the fraction of F4/80(+)CD206(+) cells (M2 Kupffer cells) while F4/80(+)CD206(-) cells (M1 Kupffer cells) were higher in the NASH group (F4/80(+)CD206(+) cell: 44% in NASH model group vs. 62% in L. paracasei group, p < 0.05). CONCLUSIONS: Lactobacillus paracasei attenuates hepatic steatosis with M2-dominant Kupffer cell polarization in a NASH model.


Assuntos
Intestinos/microbiologia , Células de Kupffer/microbiologia , Lactobacillus/fisiologia , Hepatopatia Gordurosa não Alcoólica/terapia , Probióticos , Animais , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células de Kupffer/imunologia , Células de Kupffer/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/imunologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/microbiologia , Permeabilidade , Fenótipo , Transdução de Sinais , Fatores de Tempo
20.
Dig Dis ; 33(4): 570-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26159275

RESUMO

BACKGROUND: Microbial infections are a relevant problem for patients with liver cirrhosis. Different types of bacteria are responsible for different kinds of infections: Escherichia coli and Klebsiella pneumoniae are frequently observed in spontaneous bacterial peritonitis or urinary tract infections, and Streptococcus pneumoniae and Mycoplasma pneumoniae in pulmonary infections. Mortality is up to 4-fold higher in infected patients with liver cirrhosis than in patients without infections. Key Messages: Infections in patients with liver cirrhosis are due to three major reasons: bacterial translocation, immune deficiency and an increased incidence of systemic infections. Nonparenchymal liver cells like Kupffer cells, sinusoidal endothelial cells and hepatic stellate cells are the first liver cells to come into contact with microbial products when systemic infection or bacterial translocation occurs. Kupffer cell (KC) activation by Toll-like receptor (TLR) agonists and endothelial sinusoidal dysfunction have been shown to be important mechanisms increasing portal pressure following intraperitoneal lipopolysaccharide pretreatment in cirrhotic rat livers. Reduced intrahepatic vasodilation and increased intrahepatic vasoconstriction are the relevant pathophysiological pathways. Thromboxane A2 and leukotriene (LT) C4/D4 have been identified as important vasoconstrictors. Accordingly, treatment with montelukast to inhibit the cysteinyl-LT1 receptor reduced portal pressure in cirrhotic rat livers. Clinical studies have demonstrated that activation of KCs, estimated by the amount of soluble CD163 in the blood, correlates with the risk for variceal bleeding. Additionally, intestinal decontamination with rifaximin in patients with alcohol-associated liver cirrhosis reduced the portal pressure and the risk for variceal bleeding. CONCLUSIONS: TLR activation of nonparenchymal liver cells by pathogens results in portal hypertension. This might explain the pathophysiologic correlation between microbial infections and portal hypertension in patients with liver cirrhosis. These findings are the basis for both better risk stratifying and new treatment options, such as specific inhibition of TLR for patients with liver cirrhosis and portal hypertension.


Assuntos
Infecções por Bactérias Gram-Negativas/fisiopatologia , Hipertensão Portal/microbiologia , Cirrose Hepática/microbiologia , Acetatos/uso terapêutico , Animais , Anti-Infecciosos/uso terapêutico , Antígenos CD/sangue , Antígenos de Diferenciação Mielomonocítica/sangue , Translocação Bacteriana , Ciclopropanos , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Varizes Esofágicas e Gástricas , Hemorragia Gastrointestinal/tratamento farmacológico , Hemorragia Gastrointestinal/etiologia , Infecções por Bactérias Gram-Negativas/complicações , Infecções por Bactérias Gram-Negativas/microbiologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/microbiologia , Humanos , Hipertensão Portal/tratamento farmacológico , Células de Kupffer/metabolismo , Células de Kupffer/microbiologia , Antagonistas de Leucotrienos/uso terapêutico , Leucotrienos/metabolismo , Fígado/irrigação sanguínea , Fígado/metabolismo , Fígado/microbiologia , Cirrose Hepática/complicações , Cirrose Hepática/mortalidade , Pressão na Veia Porta , Quinolinas/uso terapêutico , Ratos , Receptores de Superfície Celular/sangue , Rifamicinas/uso terapêutico , Rifaximina , Sulfetos , Tromboxano A2/metabolismo , Vasoconstrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA