Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1085895, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153600

RESUMO

Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.


Assuntos
Linfócitos T CD8-Positivos , Proteínas de Ligação ao GTP , Listeria monocytogenes , Animais , Camundongos , Proteínas de Ligação ao GTP/metabolismo , Subunidades Proteicas/metabolismo , Subpopulações de Linfócitos T
2.
FEBS J ; 288(19): 5530-5554, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33215853

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can lead to the induction of apoptosis in tumor or infected cells. However, activation of TRAIL signaling may also trigger nonapoptotic pathways in cancer and in nontransformed cells, that is, immune cells. Here, we review the current knowledge on noncanonical TRAIL signaling. The biological outcomes of TRAIL signaling in immune and malignant cells are presented and explained, with a focus on the role of TRAIL for natural killer (NK) cell function. Furthermore, we highlight the technical difficulties in dissecting the precise molecular mechanisms involved in the switch between apoptotic and nonapoptotic TRAIL signaling. Finally, we discuss the consequences thereof for a therapeutic manipulation of TRAIL in cancer and possible approaches to bypass these difficulties.


Assuntos
Apoptose/genética , Imunidade/genética , Neoplasias/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Apoptose/imunologia , Humanos , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Transdução de Sinais/genética
3.
EMBO Rep ; 21(1): e48789, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31742873

RESUMO

The role of death receptor signaling for pathogen control and infection-associated pathogenesis is multifaceted and controversial. Here, we show that during viral infection, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) modulates NK cell activity independently of its pro-apoptotic function. In mice infected with lymphocytic choriomeningitis virus (LCMV), Trail deficiency led to improved specific CD8+ T-cell responses, resulting in faster pathogen clearance and reduced liver pathology. Depletion experiments indicated that this effect was mediated by NK cells. Mechanistically, TRAIL expressed by immune cells positively and dose-dependently modulates IL-15 signaling-induced granzyme B production in NK cells, leading to enhanced NK cell-mediated T cell killing. TRAIL also regulates the signaling downstream of IL-15 receptor in human NK cells. In addition, TRAIL restricts NK1.1-triggered IFNγ production by NK cells. Our study reveals a hitherto unappreciated immunoregulatory role of TRAIL signaling on NK cells for the granzyme B-dependent elimination of antiviral T cells.


Assuntos
Células Matadoras Naturais , Viroses , Animais , Vírus da Coriomeningite Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/genética
4.
Sci Adv ; 5(7): eaav9732, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31328159

RESUMO

LRH-1 (liver receptor homolog-1/NR5a2) is an orphan nuclear receptor, which regulates glucose and lipid metabolism, as well as intestinal inflammation via the transcriptional control of intestinal glucocorticoid synthesis. Predominantly expressed in epithelial cells, its expression and role in immune cells are presently enigmatic. LRH-1 was found to be induced in immature and mature T lymphocytes upon stimulation. T cell-specific deletion of LRH-1 causes a drastic loss of mature peripheral T cells. LRH-1-depleted CD4+ T cells exert strongly reduced activation-induced proliferation in vitro and in vivo and fail to mount immune responses against model antigens and to induce experimental intestinal inflammation. Similarly, LRH-1-deficient cytotoxic CD8+ T cells fail to control viral infections. This study describes a novel and critical role of LRH-1 in T cell maturation, functions, and immopathologies and proposes LRH-1 as an emerging pharmacological target in the treatment of T cell-mediated inflammatory diseases.


Assuntos
Imunomodulação , Receptores Citoplasmáticos e Nucleares/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Apoptose/genética , Biomarcadores , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Colite/etiologia , Colite/metabolismo , Colite/patologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Suscetibilidade a Doenças , Feminino , Deleção de Genes , Humanos , Isotipos de Imunoglobulinas/imunologia , Masculino , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo
5.
Int J Mol Sci ; 19(3)2018 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-29495595

RESUMO

Neutrophils are key players in the early defense against invading pathogens. Due to their potent effector functions, programmed cell death of activated neutrophils has to be tightly controlled; however, its underlying mechanisms remain unclear. Fas ligand (FASL/CD95L) has been shown to induce neutrophil apoptosis, which is accelerated by the processing of the BH3-only protein BH3 interacting domain death agonist (BID) to trigger mitochondrial apoptotic events, and been attributed a regulatory role during viral and bacterial infections. Here, we show that, in accordance with previous works, mouse neutrophils underwent caspase-dependent apoptosis in response to FASL, and that this cell death was significantly delayed upon loss of BID. However, pan-caspase inhibition failed to protect mouse neutrophils from FASL-induced apoptosis and caused a switch to RIPK3-dependent necroptotic cell death. Intriguingly, such a switch was less evident in the absence of BID, particularly under inflammatory conditions. Delayed neutrophil apoptosis has been implicated in several auto-inflammatory diseases, including inflammatory bowel disease. We show that neutrophil and macrophage driven acute dextran sulfate sodium (DSS) induced colitis was slightly more aggravated in BID-deficient mice, based on significantly increased weight loss compared to wild-type controls. Taken together, our data support a central role for FASL > FAS and BID in mouse neutrophil cell death and further underline the anti-inflammatory role of BID.


Assuntos
Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/deficiência , Sulfato de Dextrana/efeitos adversos , Proteína Ligante Fas/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Redução de Peso/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Inibidores de Caspase/metabolismo , Morte Celular/genética , Colite/etiologia , Colite/metabolismo , Colite/patologia , Lipopolissacarídeos/imunologia , Camundongos , Neutrófilos/imunologia , Ligação Proteica , Receptor fas/metabolismo
6.
Front Immunol ; 9: 23, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29416538

RESUMO

Aberrant interferon gamma (IFNγ) expression is associated with the pathogenesis of numerous autoimmune- and inflammatory disorders, including inflammatory bowel diseases (IBD). However, the requirement of IFNγ for the pathogenesis of chronic intestinal inflammation remains controversial. The aim of this study was thus to investigate the role of IFNγ in experimental mouse models of innate and adaptive immune cell-mediated intestinal inflammation using genetically and microbiota-stabilized hosts. While we find that IFNγ drives acute intestinal inflammation in the anti-CD40 colitis model in an innate lymphoid cell (ILC)-dependent manner, IFNγ secreted by both transferred CD4 T cells and/or cells of the lymphopenic Rag1-/- recipient mice was dispensable for CD4 T cell-mediated colitis. In the absence of IFNγ, intestinal inflammation in CD4 T cell recipient mice was associated with enhanced IL17 responses; consequently, targeting IL17 signaling in IFNγ-deficient mice reduced T cell-mediated colitis. Intriguingly, in contrast to the anti-CD40 model of colitis, depletion of ILC in the Rag1-/- recipients of colitogenic CD4 T cells did not prevent induction of colonic inflammation. Together, our findings demonstrate that IFNγ represents an essential, or a redundant, pro-inflammatory cytokine for the induction of intestinal inflammation, depending on the experimental mouse model used and on the nature of the critical disease inducing immune cell populations involved.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Interferon gama/imunologia , Imunidade Adaptativa , Animais , Células Cultivadas , Colite/patologia , Colo/imunologia , Colo/patologia , Modelos Animais de Doenças , Imunidade Inata , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
IUBMB Life ; 67(4): 275-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25914114

RESUMO

In the mammalian gastrointestinal tract the close vicinity of abundant immune effector cells and trillions of commensal microbes requires sophisticated barrier and regulatory mechanisms to maintain vital host-microbial interactions and tissue homeostasis. During co-evolution of the host and its intestinal microbiota a protective multilayered barrier system was established to segregate the luminal microbes from the intestinal mucosa with its potent immune effector cells, limit bacterial translocation into host tissues to prevent tissue damage, while ensuring the vital functions of the intestinal mucosa and the luminal gut microbiota. In the present review we will focus on the different layers of protection in the intestinal tract that allow the successful mutualism between the microbiota and the potent effector cells of the intestinal innate and adaptive immune system. In particular, we will review some of the recent findings on the vital functions of the mucus layer and its site-specific adaptations to the changing quantities and complexities of the microbiota along the (gastro-) intestinal tract. Understanding the regulatory pathways that control the establishment of the mucus layer, but also its degradation during intestinal inflammation may be critical for designing novel strategies aimed at maintaining local tissue homeostasis and supporting remission from relapsing intestinal inflammation in patients with inflammatory bowel diseases.


Assuntos
Fenômenos Fisiológicos Bacterianos , Homeostase/fisiologia , Mucosa Intestinal/fisiologia , Intestinos/fisiologia , Humanos , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Muco/metabolismo
8.
Transplantation ; 97(11): 1102-9, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24804996

RESUMO

BACKGROUND: Ischemia-reperfusion injury (IRI) significantly contributes to graft dysfunction after liver transplantation. Natural killer (NK) cells are crucial innate effector cells in the liver and express tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a potent inducer of hepatocyte cell death. Here, we investigated if TRAIL expression on NK cells contributes to hepatic IRI. METHODS: The outcome after partial hepatic IRI was assessed in TRAIL-null mice and contrasted to C57BL/6J wild-type mice and after NK cell adoptive transfer in RAG2/common gamma-null mice that lack T, B, and NK cells. Liver IRI was assessed by histological analysis, alanine aminotransferase, hepatic neutrophil activation by myeloperoxidase activity, and cytokine secretion at specific time points. NK cell cytotoxicity and differentiation were assessed in vivo and in vitro. RESULTS: Twenty-four hours after reperfusion, TRAIL-null mice exhibited significantly higher serum transaminases, histological signs of necrosis, neutrophil infiltration, and serum levels of interleukin-6 compared to wild-type animals. Adoptive transfer of TRAIL-null NK cells into immunodeficient RAG2/common gamma-null mice was associated with significantly elevated liver damage compared to transfer of wild-type NK cells. In TRAIL-null mice, NK cells exhibit higher cytotoxicity and decreased differentiation compared to wild-type mice. In vitro, cytotoxicity against YAC-1 and secretion of interferon gamma by TRAIL-null NK cells were significantly increased compared to wild-type controls. CONCLUSIONS: These experiments reveal that expression of TRAIL on NK cells is protective in a murine model of hepatic IRI through modulation of NK cell cytotoxicity and NK cell differentiation.


Assuntos
Células Matadoras Naturais/metabolismo , Fígado/patologia , Traumatismo por Reperfusão/patologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Transferência Adotiva , Animais , Morte Celular , Diferenciação Celular , Citocinas/metabolismo , Granulócitos/citologia , Hepatócitos/citologia , Interleucina-6/sangue , Células Matadoras Naturais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Necrose/patologia , Neutrófilos/citologia , Estresse Oxidativo , Peroxidase/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transaminases/sangue
9.
Oncoimmunology ; 1(4): 529-530, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22754774

RESUMO

Expression or release of immunosuppressive molecules may protect tumor cells from the recognition and destruction by the immune system. New findings indicate that colorectal tumors produce immunoregulatory glucocorticoids and thereby suppress immune cell activation. The nuclear receptor LRH-1 plays a critical role in the regulation of colorectal tumor proliferation and glucocorticoid synthesis.

10.
J Immunol ; 188(2): 916-22, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22156338

RESUMO

The proapoptotic Bcl-2 homolog Bim was shown to control the apoptosis of both T cells and hepatocytes. This dual role of Bim might be particularly relevant for the development of viral hepatitis, in which both the sensitivity of hepatocytes to apoptosis stimuli and the persistence of cytotoxic T cells are essential factors for the outcome of the disease. The relevance of Bim in regulating survival of cytotoxic T cells or induction of hepatocyte death has only been investigated in separate systems, and their relative contributions to the pathogenesis of T cell-mediated hepatitis remain unclear. Using the highly dynamic model system of lymphocytic choriomeningitis virus-mediated hepatitis and bone marrow chimeras, we found that Bim has a dual role in the development of lymphocytic choriomeningitis virus-induced, T cell-mediated hepatitis. Although the absence of Bim in parenchymal cells led to markedly attenuated liver damage, loss of Bim in the lymphoid compartment moderately enhanced hepatitis. However, when both effects were combined in Bim(-/-) mice, the effect of Bim deficiency in the lymphoid compartment was overcompensated for by the reduced sensitivity of Bim(-/-) hepatocytes to T cell-induced apoptosis, resulting in the protection of Bim(-/-) mice from hepatitis.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose/imunologia , Hepatite Viral Animal/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Hepatite Viral Animal/patologia , Hepatite Viral Animal/virologia , Hepatócitos/imunologia , Hepatócitos/patologia , Hepatócitos/virologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Família Multigênica , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Índice de Gravidade de Doença , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia
11.
Int J Nanomedicine ; 6: 1793-800, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21980242

RESUMO

BACKGROUND: Experimental tissue fusion benefits from the selective heating of superparamagnetic iron oxide nanoparticles (SPIONs) under high frequency irradiation. However, the metabolic pathways of SPIONs for tissue fusion remain unknown. Hence, the goal of this in vivo study was to analyze the distribution of SPIONs in different organs by means of magnetic resonance imaging (MRI) and histological analysis after a SPION-containing patch implantation. METHODS: SPION-containing patches were implanted in rats. Three animal groups were studied histologically over six months. Degradation assessment of the SPION-albumin patch was performed in vivo using MRI for iron content localization and biodistribution. RESULTS: No SPION degradation or accumulation into the reticuloendothelial system was detected by MRI, MRI relaxometry, or histology, outside the area of the implantation patch. Concentrations from 0.01 µg/mL to 25 µg/mL were found to be hyperintense in T1-like gradient echo sequences. The best differentiation of concentrations was found in T2 relaxometry, susceptibility-sensitive gradient echo sequences, and in high repetition time T2 images. Qualitative and semiquantitative visualization of small concentrations and accumulation of SPIONs by MRI are feasible. In histological liver samples, Kupffer cells were significantly correlated with postimplantation time, but no differences were observed between sham-treated and induction/no induction groups. Transmission electron microscopy showed local uptake of SPIONs in macrophages and cells of the reticuloendothelial system. Apoptosis staining using caspase showed no increased toxicity compared with sham-treated tissue. Implanted SPION patches were relatively inert with slow, progressive local degradation over the six-month period. No distant structural alterations in the studied tissue could be observed. CONCLUSION: Systemic bioavailability may play a role in specific SPION implant toxicity and therefore the local degradation process is a further aspect to be assessed in future studies.


Assuntos
Compostos Férricos/farmacocinética , Nanopartículas de Magnetita/administração & dosagem , Animais , Compostos Férricos/metabolismo , Histocitoquímica , Implantes Experimentais , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/química , Masculino , Ratos , Ratos Wistar , Distribuição Tecidual
12.
Hepatology ; 53(1): 282-92, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20872776

RESUMO

UNLABELLED: Fas/CD95 is a critical mediator of cell death in many chronic and acute liver diseases and induces apoptosis in primary hepatocytes in vitro. In contrast, the proinflammatory cytokine tumor necrosis factor α (TNFα) fails to provoke cell death in isolated hepatocytes but has been implicated in hepatocyte apoptosis during liver diseases associated with chronic inflammation. Here we report that TNFα sensitizes primary murine hepatocytes cultured on collagen to Fas ligand (FasL)-induced apoptosis. This synergism is time-dependent and is specifically mediated by TNFα. Fas itself is essential for the sensitization, but neither Fas up-regulation nor endogenous FasL is responsible for this effect. Although FasL is shown to induce Bid-independent apoptosis in hepatocytes cultured on collagen, the sensitizing effect of TNFα is clearly dependent on Bid. Moreover, both c-Jun N-terminal kinase activation and Bim, another B cell lymphoma 2 homology domain 3 (BH3)-only protein, are crucial mediators of TNFα-induced apoptosis sensitization. Bim and Bid activate the mitochondrial amplification loop and induce cytochrome c release, a hallmark of type II apoptosis. The mechanism of TNFα-induced sensitization is supported by a mathematical model that correctly reproduces the biological findings. Finally, our results are physiologically relevant because TNFα also induces sensitivity to agonistic anti-Fas-induced liver damage. CONCLUSION: Our data suggest that TNFα can cooperate with FasL to induce hepatocyte apoptosis by activating the BH3-only proteins Bim and Bid.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Hepatopatias/etiologia , Proteínas de Membrana/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Receptor fas , Animais , Anticorpos/farmacologia , Proteína 11 Semelhante a Bcl-2 , Caspase 3/metabolismo , Caspase 7/metabolismo , Células Cultivadas , Proteína Ligante Fas/fisiologia , Hepatócitos/patologia , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/fisiologia , Receptor fas/imunologia
13.
J Exp Med ; 207(5): 1057-66, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20439544

RESUMO

Although tumor necrosis factor (alpha) (TNF) exerts proinflammatory activities in a variety of diseases, including inflammatory bowel disease, there is increasing evidence for antiinflammatory actions of TNF. In contrast, glucocorticoids (GCs) are steroid hormones that suppress inflammation, at least in part by regulating the expression and action of TNF. We report that TNF induces extraadrenal production of immunoregulatory GCs in the intestinal mucosa during acute intestinal inflammation. The absence of TNF results in a lack of colonic GC synthesis and exacerbation of dextran sodium sulfate-induced colitis. TNF seems to promote local steroidogenesis by directly inducing steroidogenic enzymes in intestinal epithelial cells. Therapeutic administration of TNF induces GC synthesis in oxazolone-induced colitis and ameliorates intestinal inflammation, whereas inhibition of intestinal GC synthesis abrogates the therapeutic effect of TNF. These data show that TNF suppresses the pathogenesis of acute intestinal inflammation by promoting local steroidogenesis.


Assuntos
Glucocorticoides/biossíntese , Inflamação/prevenção & controle , Enteropatias/prevenção & controle , Fator de Necrose Tumoral alfa/farmacologia , Animais , Doença de Crohn/prevenção & controle , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Doenças Inflamatórias Intestinais/prevenção & controle , Enteropatias/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Camundongos
14.
FASEB J ; 24(5): 1340-6, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20056718

RESUMO

Stringent control of immune responses in the intestinal mucosa is critical for the maintenance of immune homeostasis and prevention of tissue damage, such as observed during inflammatory bowel disease. Intestinal epithelial cells, primarily thought to form a simple physical barrier, critically regulate intestinal immune cell functions by producing immunoregulatory glucocorticoids on T-cell activation. In this study we investigated whether stimulation of cells of the innate immune system results in the induction of intestinal glucocorticoids synthesis and what role TNF-alpha plays in this process. Stimulation of the innate immune system with lipopolysaccharide (LPS) led to an up-regulation of colonic steroidogenic enzymes and the induction of intestinal glucocorticoid synthesis. The observed induction was dependent on macrophage effector functions, as depletion of macrophages using clodronate-containing liposomes, but not absence of T and B cells, inhibited intestinal glucocorticoid synthesis. LPS-induced glucocorticoid synthesis was critically dependent on TNF-alpha as it was significantly decreased in TNF-alpha-deficient animals. Both TNF receptor-1 and -2 were found to be equally involved in LPS- and T-cell-induced intestinal GC synthesis. These results describe a novel and critical role of TNF-alpha in immune cell-induced intestinal glucocorticoid synthesis.


Assuntos
Glucocorticoides/biossíntese , Intestinos/imunologia , Lipopolissacarídeos/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linfócitos B/imunologia , Glucocorticoides/genética , Imunidade Inata , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Mutantes , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/genética
15.
Ann N Y Acad Sci ; 1171: 50-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19723037

RESUMO

The death ligand members of the tumor necrosis factor (TNF) family are potent inducers of apoptosis in a variety of cell types. In particular, TNF-related apoptosis-inducing ligand (TRAIL) has recently received much scientific and commercial attention because of its potent tumor cell-killing activity while leaving normal untransformed cells mostly unaffected. Furthermore, TRAIL strongly synergizes with conventional chemotherapeutic drugs in inducing tumor cell apoptosis, making it a most promising candidate for future cancer therapy. Increasing evidence indicates, however, that TRAIL may also induce or modulate apoptosis in primary cells. A particular concern is the potential side effect of TRAIL-based tumor therapies in the liver. In this review we summarize some of the recent findings on the role of TRAIL in tumor cell and hepatocyte apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Humanos , Camundongos , Neoplasias/patologia , Ligante Indutor de Apoptose Relacionado a TNF/efeitos adversos , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Vet Res ; 40(6): 58, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19631032

RESUMO

Little is currently known about the lymphocyte populations in the normal and diseased canine gut. The aim of this study was thus the phenotypical and functional characterization of canine intestinal intraepithelial lymphocytes (IEL). IEL were isolated from full-thickness biopsies of 15 adult Swiss Beagle dogs (mean age 8.2 +/-2.8 years) and compared to mesenteric lymph node cells. The phenotypical characterization by multi-parameter flow cytometry revealed that canine IEL differ substantially from lymph node T cells, and consist of various unconventional lymphocyte subsets, unique to mucosal surfaces. These include gammasigma T cells, and CD4(-)CD8(-) and CD8alphaalpha(+) T cells. IEL populations in adult dogs were also compared to those isolated from neonatal Beagle dogs. Analysis revealed a high frequency of undifferentiated CD4(-)CD8(-) T cells in newborn dogs whereas mature CD4(+) and CD8(+) T cells predominate in adult dogs, indicating maturation of the intestinal immune system during development. As IEL in other species are thought to exhibit regulatory functions, we investigated the role of IEL on the activation-induced proliferation of lymph node T cells. While IEL alone did not show activation-induced proliferation, they significantly inhibited the proliferation of activated lymph node T cells in a cell number-dependent manner. These findings are the first to demonstrate that canine intestinal IEL have an immunoregulatory phenotype, which may contribute to the maintenance of intestinal immune homeostasis and may, therefore, be lost in canine chronic enteropathies.


Assuntos
Mucosa Intestinal/citologia , Linfócitos/classificação , Linfócitos/fisiologia , Animais , Cães , Feminino , Regulação da Expressão Gênica , Masculino , Mitógenos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo
17.
Semin Immunopathol ; 31(2): 267-77, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19533131

RESUMO

Liver diseases represent an important cause of morbidity and mortality in the world. Death of hepatocytes and other hepatic cell types is a characteristic feature of several forms of liver injury such as cholestasis, viral hepatitis, drug- or toxin-induced injury, and alcohol-induced liver damage. Moreover, irrespectively of the reason, liver injury seems to be facilitated by similar immune effector mechanisms common to these various liver diseases. Indeed, common immune effector mechanisms may explain the high prevalence of cirrhosis and cancer development in most forms of liver disease. Improved understanding of the immune cell-mediated mechanisms involved in hepatocyte cell death could be beneficial for the development of common therapeutic strategies against different forms of liver diseases. In this review, we will discuss novel findings on the role of different immune cells in liver disease and immune cell-induced death executioner mechanisms involved in hepatocyte cell death.


Assuntos
Hepatócitos/imunologia , Imunidade Celular , Hepatopatias/imunologia , Fígado/imunologia , Fígado/lesões , Animais , Morte Celular/imunologia , Hepatócitos/patologia , Humanos , Fígado/patologia , Hepatopatias/patologia
18.
J Exp Med ; 205(13): 3065-77, 2008 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19064696

RESUMO

Mononuclear phagocytes have been attributed a crucial role in the host defense toward influenza virus (IV), but their contribution to influenza-induced lung failure is incompletely understood. We demonstrate for the first time that lung-recruited "exudate" macrophages significantly contribute to alveolar epithelial cell (AEC) apoptosis by the release of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in a murine model of influenza-induced pneumonia. Using CC-chemokine receptor 2-deficient (CCR2(-/-)) mice characterized by defective inflammatory macrophage recruitment, and blocking anti-CCR2 antibodies, we show that exudate macrophage accumulation in the lungs of influenza-infected mice is associated with pronounced AEC apoptosis and increased lung leakage and mortality. Among several proapoptotic mediators analyzed, TRAIL messenger RNA was found to be markedly up-regulated in alveolar exudate macrophages as compared with peripheral blood monocytes. Moreover, among the different alveolar-recruited leukocyte subsets, TRAIL protein was predominantly expressed on macrophages. Finally, abrogation of TRAIL signaling in exudate macrophages resulted in significantly reduced AEC apoptosis, attenuated lung leakage, and increased survival upon IV infection. Collectively, these findings demonstrate a key role for exudate macrophages in the induction of alveolar leakage and mortality in IV pneumonia. Epithelial cell apoptosis induced by TRAIL-expressing macrophages is identified as a major underlying mechanism.


Assuntos
Apoptose/imunologia , Influenza Humana/imunologia , Pulmão , Macrófagos/imunologia , Orthomyxoviridae/imunologia , Pneumonia/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Quimera , Humanos , Leucócitos Mononucleares/imunologia , Pulmão/imunologia , Pulmão/fisiologia , Pulmão/virologia , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orthomyxoviridae/patogenicidade , Fagócitos/imunologia , Pneumonia/mortalidade , Pneumonia/virologia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Ligante Indutor de Apoptose Relacionado a TNF/genética
20.
FASEB J ; 22(12): 4117-25, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18711026

RESUMO

Glucocorticoids are anti-inflammatory steroids with important applications in the treatment of inflammatory diseases. Endogenous glucocorticoids are mainly produced by the adrenal glands, although there is increasing evidence for extra-adrenal sources. Recent findings show that intestinal crypt cells produce glucocorticoids, which contribute to the maintenance of intestinal immune homeostasis. Intestinal glucocorticoid synthesis is critically regulated by the transcription factor liver receptor homologue-1 (LRH-1). As expression of steroidogenic enzymes and LRH-1 is restricted to the proliferating cells of the crypts, we aimed to investigate the role of the cell cycle in the regulation of LRH-1 activity and intestinal glucocorticoid synthesis. We here show that either pharmacological or molecular modulation of cell cycle progression significantly inhibited expression of steroidogenic enzymes and synthesis of glucocorticoids in intestinal epithelial cells. Synchronization of intestinal epithelial cells in the cell cycle revealed that expression of steroidogenic enzymes is preferentially induced at the G(1)/S stage. Differentiation of immature intestinal epithelial cells to mature nonproliferating cells also resulted in reduced expression of steroidogenic enzymes. This cell cycle-related effect on intestinal steroidogenesis was found to be mediated through the regulation of LRH-1 transcriptional activity. This mechanism may restrict intestinal glucocorticoid synthesis to the proliferating cells of the crypts.


Assuntos
Ciclo Celular/fisiologia , Glucocorticoides/biossíntese , Mucosa Intestinal/metabolismo , Animais , Afidicolina/farmacologia , Proteína Quinase CDC2/fisiologia , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular , Linhagem Celular , Enzima de Clivagem da Cadeia Lateral do Colesterol/biossíntese , Colforsina/farmacologia , Ciclina B/biossíntese , Ciclina B1 , Ciclina D1/biossíntese , Doxorrubicina/farmacologia , Mucosa Intestinal/citologia , Cinetina/farmacologia , Camundongos , Nocodazol/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteína do Retinoblastoma/fisiologia , Esteroide 11-beta-Hidroxilase/biossíntese , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA