Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(5): e0266980, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35507536

RESUMO

PURPOSE: The solid tumor microenvironment (TME) drives T cell dysfunction and inhibits the effectiveness of immunotherapies such as chimeric antigen receptor-based T cell (CAR T) cells. Early data has shown that modulation of T cell metabolism can improve intratumoral T cell function in preclinical models. EXPERIMENTAL DESIGN: We evaluated GPC3 expression in human normal and tumor tissue specimens. We developed and evaluated BOXR1030, a novel CAR T therapeutic co-expressing glypican-3 (GPC3)-targeted CAR and exogenous glutamic-oxaloacetic transaminase 2 (GOT2) in terms of CAR T cell function both in vitro and in vivo. RESULTS: Cell surface expression of tumor antigen GPC3 was observed by immunohistochemical staining in tumor biopsies from hepatocellular carcinoma, liposarcoma, squamous lung cancer, and Merkel cell carcinoma patients. Compared to control GPC3 CAR alone, BOXR1030 (GPC3-targeted CAR T cell that co-expressed GOT2) demonstrated superior in vivo efficacy in aggressive solid tumor xenograft models, and showed favorable attributes in vitro including an enhanced cytokine production profile, a less-differentiated T cell phenotype with lower expression of stress and exhaustion markers, an enhanced metabolic profile and increased proliferation in TME-like conditions. CONCLUSIONS: Together, these results demonstrated that co-expression of GOT2 can substantially improve the overall antitumor activity of CAR T cells by inducing broad changes in cellular function and phenotype. These data show that BOXR1030 is an attractive approach to targeting select solid tumors. To this end, BOXR1030 will be explored in the clinic to assess safety, dose-finding, and preliminary efficacy (NCT05120271).


Assuntos
Neoplasias Hepáticas , Receptores de Antígenos Quiméricos , Linhagem Celular Tumoral , Glipicanas/genética , Glipicanas/metabolismo , Xenoenxertos , Humanos , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/patologia , Linfócitos T , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Biol Chem ; 292(1): 15-30, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27903651

RESUMO

Modulation of T cell proliferation and function by immunoregulatory myeloid cells are an essential means of preventing self-reactivity and restoring tissue homeostasis. Consumption of amino acids such as arginine and tryptophan by immunoregulatory macrophages is one pathway that suppresses local T cell proliferation. Using a reduced complexity in vitro macrophage-T cell co-culture system, we show that macrophage arginase-1 is the only factor required by M2 macrophages to block T cells in G1, and this effect is mediated by l-arginine elimination rather than metabolite generation. Tracking how T cells adjust their metabolism when deprived of arginine revealed the significance of macrophage-mediated arginine deprivation to T cells. We found mTORC1 activity was unaffected in the initial G1 block. After 2 days of arginine deprivation, mTORC1 activity declined paralleling a selective down-regulation of SREBP target gene expression, whereas mRNAs involved in glycolysis, gluconeogenesis, and T cell activation were unaffected. Cell cycle arrest was reversible at any point by exogenous arginine, suggesting starved T cells remain poised awaiting nutrients. Arginine deprivation-induced cell cycle arrest was mediated in part by Rictor/mTORC2, providing evidence that this nutrient recognition pathway is a central component of how T cells measure environmental arginine.


Assuntos
Arginina/metabolismo , Proteínas de Transporte/metabolismo , Pontos de Checagem do Ciclo Celular/imunologia , Proliferação de Células , Complexos Multiproteicos/metabolismo , Células Mieloides/imunologia , Linfócitos T/imunologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Tolerância Imunológica , Terapia de Imunossupressão , Ativação Linfocitária , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Linfócitos T/metabolismo
3.
JCI Insight ; 1(20): e87446, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27942582

RESUMO

Pentraxin-2 (PTX-2), also known as serum amyloid P component (SAP/APCS), is a constitutive, antiinflammatory, innate immune plasma protein whose circulating level is decreased in chronic human fibrotic diseases. Here we show that recombinant human PTX-2 (rhPTX-2) retards progression of chronic kidney disease in Col4a3 mutant mice with Alport syndrome, reducing blood markers of kidney failure, enhancing lifespan by 20%, and improving histological signs of disease. Exogenously delivered rhPTX-2 was detected in macrophages but also in tubular epithelial cells, where it counteracted macrophage activation and was cytoprotective for the epithelium. Computational analysis of genes regulated by rhPTX-2 identified the transcriptional regulator c-Jun along with its activator protein-1 (AP-1) binding partners as a central target for the function of rhPTX-2. Accordingly, PTX-2 attenuates c-Jun and AP-1 activity, and reduces expression of AP-1-dependent inflammatory genes in both monocytes and epithelium. Our studies therefore identify rhPTX-2 as a potential therapy for chronic fibrotic disease of the kidney and an important inhibitor of pathological c-Jun signaling in this setting.


Assuntos
Proteína C-Reativa/farmacologia , Rim/patologia , Nefrite Hereditária/terapia , Proteínas do Tecido Nervoso/farmacologia , Proteínas Proto-Oncogênicas c-jun/antagonistas & inibidores , Transdução de Sinais , Fator de Transcrição AP-1/antagonistas & inibidores , Animais , Células Cultivadas , Fibrose , Humanos , Ativação de Macrófagos , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Monócitos , Nefrite Hereditária/patologia , Proteínas Recombinantes/farmacologia
4.
Am J Pathol ; 186(10): 2519-31, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27555112

RESUMO

Pericytes, resident fibroblasts, and mesenchymal stem cells are poorly described cell populations. They have recently been characterized in much greater detail in rodent lungs and have been shown to play important roles in development, homeostasis, response to injury and pathogens, as well as recovery from damage. These closely related mesenchymal cell populations form extensive connections to the lung's internal structure, as well as its internal and external surfaces. They generate and remodel extracellular matrix, coregulate the vasculature, help maintain and restore the epithelium, and act as sentries for the immune system. In this review, we revisit these functions in light of significant advances in characterizing and tracking lung fibroblast populations in rodents. Lineage tracing experiments have mapped the heritage, identified functions that discriminate lung pericytes from resident fibroblasts, identified a subset of mesenchymal stem cells, and shown these populations to be the predominant progenitors of pathological fibroblasts and myofibroblasts in lung diseases. These findings point to the importance of resident lung mesenchymal populations as therapeutic targets in acute lung injury as well as fibrotic and degenerative diseases. Far from being passive and quiescent, pericytes and resident fibroblasts are busily sensing and responding, through diverse mechanisms, to changes in lung health and function.


Assuntos
Fibroblastos/fisiologia , Lesão Pulmonar/terapia , Pulmão/patologia , Células-Tronco Mesenquimais/fisiologia , Pericitos/fisiologia , Fibrose Pulmonar/patologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Modelos Animais de Doenças , Matriz Extracelular/patologia , Humanos , Pulmão/embriologia , Lesão Pulmonar/patologia , Miofibroblastos/fisiologia , Fenótipo , Fibrose Pulmonar/terapia , Roedores
5.
Sci Transl Med ; 8(337): 337ra65, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27147589

RESUMO

Thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 are important initiators of type 2-associated mucosal inflammation and immunity. However, their role in the maintenance of progressive type 2 inflammation and fibrosis is much less clear. Using chronic models of helminth infection and allergic lung inflammation, we show that collective disruption of TSLP, IL-25, and IL-33 signaling suppresses chronic and progressive type 2 cytokine-driven inflammation and fibrosis. In a schistosome lung granuloma model or during chronic Schistosoma mansoni infection in the liver, individual ablation of TSLP, IL-25, or IL-33/ST2 had no impact on the development of IL-4/IL-13-dependent inflammation or fibrosis. However, significant reductions in granuloma-associated eosinophils, hepatic fibrosis, and IL-13-producing type 2 innate lymphoid cells (ILC2s) were observed when signaling of all three mediators was simultaneously disrupted. Combined blockade through monoclonal antibody (mAb) treatment also reduced IL-5 and IL-13 expression during primary and secondary granuloma formation in the lungs. In a model of chronic house dust mite-induced allergic lung inflammation, combined mAb treatment did not decrease established inflammation or fibrosis. TSLP/IL-33 double-knockout mice treated with anti-IL-25 mAb during priming, however, displayed decreased inflammation, mucus production, and lung remodeling in the chronic phase. Together, these studies reveal partially redundant roles for TSLP, IL-25, and IL-33 in the maintenance of type 2 pathology and suggest that in some settings, early combined targeting of these mediators is necessary to ameliorate progressive type 2-driven disease.


Assuntos
Citocinas/metabolismo , Fibrose/imunologia , Inflamação/imunologia , Inflamação/terapia , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Neoplasias Pulmonares/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Citocinas/antagonistas & inibidores , Citocinas/genética , Feminino , Fibrose/tratamento farmacológico , Fibrose/terapia , Granuloma/tratamento farmacológico , Granuloma/imunologia , Granuloma/parasitologia , Granuloma/terapia , Inflamação/tratamento farmacológico , Interleucina-13/antagonistas & inibidores , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-17/antagonistas & inibidores , Interleucina-17/genética , Interleucina-33/antagonistas & inibidores , Interleucina-33/genética , Interleucina-4/antagonistas & inibidores , Interleucina-4/genética , Interleucina-4/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/parasitologia , Neoplasias Pulmonares/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Schistosoma mansoni/imunologia , Schistosoma mansoni/patogenicidade , Linfopoietina do Estroma do Timo
6.
Nat Immunol ; 17(5): 538-44, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27043413

RESUMO

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103(+) dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.


Assuntos
Quitinases/imunologia , Trato Gastrointestinal/imunologia , Imunidade/imunologia , Infecções por Strongylida/imunologia , Animais , Quitinases/genética , Quitinases/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/imunologia , Canais de Cloreto/metabolismo , Citometria de Fluxo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/parasitologia , Expressão Gênica/imunologia , Hormônios Ectópicos/genética , Hormônios Ectópicos/imunologia , Hormônios Ectópicos/metabolismo , Interações Hospedeiro-Parasita/imunologia , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Imunidade/genética , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-13/metabolismo , Lectinas/genética , Lectinas/imunologia , Lectinas/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Nematospiroides dubius/imunologia , Nematospiroides dubius/fisiologia , Nippostrongylus/imunologia , Nippostrongylus/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Infecções por Strongylida/metabolismo , Infecções por Strongylida/parasitologia , beta-N-Acetil-Hexosaminidases/genética , beta-N-Acetil-Hexosaminidases/imunologia , beta-N-Acetil-Hexosaminidases/metabolismo
7.
Oncotarget ; 5(23): 12027-42, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25294815

RESUMO

M2 macrophages promote tumor growth and metastasis, but their interactions with specific tumor cell populations are poorly characterized. Using a mouse model of spontaneous melanoma, we showed that CD34- but not CD34+ tumor-initiating cells (TICs) depend on M2 macrophages for survival and proliferation. Tumor-associated macrophages (TAMs) and macrophage-conditioned media protected CD34- TICs from chemotherapy in vitro. In vivo, while inhibition of CD115 suppressed the macrophage-dependent CD34- TIC population, chemotherapy accelerated its development. The ability of TICs to respond to TAMs was acquired during melanoma progression and immediately preceded a surge in metastatic outgrowth. TAM-derived transforming growth factor-ß (TGFß) and polyamines produced via the Arginase pathway were critical for stimulation of TICs and synergized to promote their growth.


Assuntos
Arginase/metabolismo , Macrófagos/imunologia , Melanoma/imunologia , Melanoma/metabolismo , Células-Tronco Neoplásicas/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Feminino , Masculino , Melanoma/patologia , Camundongos , Camundongos Mutantes , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Transdução de Sinais/imunologia
8.
PLoS Pathog ; 10(9): e1004372, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25211233

RESUMO

Mice expressing a Cre recombinase from the lysozyme M-encoding locus (Lyz2) have been widely used to dissect gene function in macrophages and neutrophils. Here, we show that while naïve resident tissue macrophages from IL-4Rαf(lox/delta)LysM(Cre) mice almost completely lose IL-4Rα function, a large fraction of macrophages elicited by sterile inflammatory stimuli, Schistosoma mansoni eggs, or S. mansoni infection, fail to excise Il4rα. These F4/80(hi)CD11b(hi) macrophages, in contrast to resident tissue macrophages, express lower levels of Lyz2 explaining why this population resists LysM(Cre)-mediated deletion. We show that in response to IL-4 and IL-13, Lyz2(lo)IL-4Rα(+) macrophages differentiate into an arginase 1-expressing alternatively-activated macrophage (AAM) population, which slows the development of lethal fibrosis in schistosomiasis. In contrast, we identified Lyz2(hi)IL-4Rα(+) macrophages as the key subset of AAMs mediating the downmodulation of granulomatous inflammation in chronic schistosomiasis. Our observations reveal a limitation on using a LysMCre mouse model to study gene function in inflammatory settings, but we utilize this limitation as a means to demonstrate that distinct populations of alternatively activated macrophages control inflammation and fibrosis in chronic schistosomiasis.


Assuntos
Fibrose/imunologia , Inflamação/imunologia , Macrófagos Peritoneais/imunologia , Receptores de Superfície Celular/fisiologia , Schistosoma mansoni/patogenicidade , Esquistossomose/imunologia , Animais , Células Cultivadas , Doença Crônica , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose/parasitologia , Fibrose/patologia , Inflamação/parasitologia , Inflamação/patologia , Integrases/metabolismo , Macrófagos Peritoneais/parasitologia , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Neutrófilos/parasitologia , Neutrófilos/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esquistossomose/parasitologia , Esquistossomose/patologia
9.
Immunity ; 40(3): 378-88, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24631153

RESUMO

Innate lymphoid cells (ILCs) are critical in innate immune responses to pathogens and lymphoid organ development. Similar to CD4(+) T helper (Th) cell subsets, ILC subsets positive for interleukin-7 receptor α (IL-7Rα) produce distinct sets of effector cytokines. However, the molecular control of IL-7Rα(+) ILC development and maintenance is unclear. Here, we report that GATA3 was indispensable for the development of all IL-7Rα(+) ILC subsets and T cells but was not required for the development of classical natural killer cells. Conditionally Gata3-deficient mice had no lymph nodes and were susceptible to Citrobactor rodentium infection. After the ILCs had fully developed, GATA3 remained important for the maintenance and functions of ILC2s. Genome-wide gene expression analyses indicated that GATA3 regulated a similar set of cytokines and receptors in Th2 cells and ILC2s, but not in ILC3s. Thus, GATA3 plays parallel roles in regulating the development and functions of CD4(+) T cells and IL-7Rα(+) ILCs.


Assuntos
Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica , Imunidade Inata/genética , Subpopulações de Linfócitos/metabolismo , Receptores de Interleucina-7/genética , Animais , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/imunologia , Fator de Transcrição GATA3/genética , Predisposição Genética para Doença , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptores de Interleucina-7/metabolismo
10.
PLoS Pathog ; 9(10): e1003731, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204274

RESUMO

BACKGROUND: In order to promote infection, the blood-borne parasite Trypanosoma brucei releases factors that upregulate arginase expression and activity in myeloid cells. METHODOLOGY/PRINCIPAL FINDINGS: By screening a cDNA library of T. brucei with an antibody neutralizing the arginase-inducing activity of parasite released factors, we identified a Kinesin Heavy Chain isoform, termed TbKHC1, as responsible for this effect. Following interaction with mouse myeloid cells, natural or recombinant TbKHC1 triggered SIGN-R1 receptor-dependent induction of IL-10 production, resulting in arginase-1 activation concomitant with reduction of nitric oxide (NO) synthase activity. This TbKHC1 activity was IL-4Rα-independent and did not mirror M2 activation of myeloid cells. As compared to wild-type T. brucei, infection by TbKHC1 KO parasites was characterized by strongly reduced parasitaemia and prolonged host survival time. By treating infected mice with ornithine or with NO synthase inhibitor, we observed that during the first wave of parasitaemia the parasite growth-promoting effect of TbKHC1-mediated arginase activation resulted more from increased polyamine production than from reduction of NO synthesis. In late stage infection, TbKHC1-mediated reduction of NO synthesis appeared to contribute to liver damage linked to shortening of host survival time. CONCLUSION: A kinesin heavy chain released by T. brucei induces IL-10 and arginase-1 through SIGN-R1 signaling in myeloid cells, which promotes early trypanosome growth and favors parasite settlement in the host. Moreover, in the late stage of infection, the inhibition of NO synthesis by TbKHC1 contributes to liver pathogenicity.


Assuntos
Arginase/imunologia , Cinesinas/imunologia , Proteínas de Protozoários/imunologia , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Animais , Arginase/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Cinesinas/genética , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Óxido Nítrico/imunologia , Proteínas de Protozoários/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Trypanosoma brucei brucei/genética , Tripanossomíase Africana/genética , Tripanossomíase Africana/patologia
11.
PLoS One ; 8(4): e61961, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637937

RESUMO

Th2-driven lung inflammation increases Arginase 1 (Arg1) expression in alternatively-activated macrophages (AAMs). AAMs modulate T cell and wound healing responses and Arg1 might contribute to asthma pathogenesis by inhibiting nitric oxide production, regulating fibrosis, modulating arginine metabolism and restricting T cell proliferation. We used mice lacking Arg1 in myeloid cells to investigate the contribution of Arg1 to lung inflammation and pathophysiology. In six model systems encompassing acute and chronic Th2-mediated lung inflammation we observed neither a pathogenic nor protective role for myeloid-expressed Arg1. The number and composition of inflammatory cells in the airways and lungs, mucus secretion, collagen deposition, airway hyper-responsiveness, and T cell cytokine production were not altered if AAMs were deficient in Arg1 or simultaneously in both Arg1 and NOS2. Our results argue that Arg1 is a general feature of alternative activation but only selectively regulates Th2 responses. Therefore, attempts to experimentally or therapeutically inhibit arginase activity in the lung should be examined with caution.


Assuntos
Arginase/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Células Th2/imunologia , Animais , Antígenos de Helmintos/imunologia , Arginase/genética , Aspergillus/imunologia , Expressão Gênica , Granuloma/imunologia , Granuloma/metabolismo , Granuloma/patologia , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Células Mieloides/patologia , Ovalbumina/imunologia , Pneumonia/genética , Pneumonia/patologia , Schistosoma mansoni/imunologia
12.
Eur J Immunol ; 41(9): 2509-14, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21952807

RESUMO

Macrophages regulate the initiation, maintenance, and resolution of chronic inflammatory responses and their function depends on their activation status. Studies in mice infected with the helminth parasite Schistosoma mansoni have been particularly helpful in defining the in vivo function of classically and alternatively activated macrophages (AAMϕs). These studies have shown that AAMϕs protect mice from acute and chronic S. mansoni infection through distinct mechanisms, which are discussed in this Viewpoint.


Assuntos
Macrófagos/imunologia , Schistosoma/imunologia , Esquistossomose/imunologia , Doença Aguda , Animais , Doença Crônica , Humanos , Inflamação , Ativação de Macrófagos , Macrófagos/microbiologia , Camundongos
13.
J Exp Med ; 208(5): 893-900, 2011 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-21502330

RESUMO

De novo expression of Muc5ac, a mucin not normally expressed in the intestinal tract, is induced in the cecum of mice resistant to Trichuris muris infection. In this study, we investigated the role of Muc5ac, which is detected shortly before worm expulsion and is associated with the production of interleukin-13 (IL-13), in resistance to this nematode. Muc5ac-deficient mice were incapable of expelling T. muris from the intestine and harbored long-term chronic infections, despite developing strong T(H)2 responses. Muc5ac-deficient mice had elevated levels of IL-13 and, surprisingly, an increase in the T(H)1 cytokine IFN-γ. Because T(H)1 inflammation is thought to favor chronic nematode infection, IFN-γ was neutralized in vivo, resulting in an even stronger T(H)2-type immune response. Nevertheless, despite a more robust T(H)2 effector response, the Muc5ac-deficient mice remained highly susceptible to chronic T. muris infection. Importantly, human MUC5AC had a direct detrimental effect on nematode vitality. Moreover, the absence of Muc5ac caused a significant delay in the expulsion of two other gut-dwelling nematodes (Trichinella spiralis and Nippostrongylus brasiliensis). Thus, for the first time, we identify a single mucin, Muc5ac, as a direct and critical mediator of resistance during intestinal nematode infection.


Assuntos
Enteropatias Parasitárias/imunologia , Mucina-5AC/imunologia , Células Th1/imunologia , Células Th2/imunologia , Tricuríase/imunologia , Trichuris/imunologia , Animais , Ceco/imunologia , Ceco/parasitologia , Doença Crônica , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-13/genética , Interleucina-13/imunologia , Enteropatias Parasitárias/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Mucina-5AC/genética , Mucina-5AC/metabolismo , Tricuríase/genética
14.
Curr Protoc Immunol ; Chapter 14: Unit14.22, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21462164

RESUMO

Macrophages play key roles in wound repair and fibrosis by regulating extracellular matrix turnover. Macrophages can process matrix components themselves, but also recruit and alter the functions of other cell types that directly build or degrade extracellular matrix. Classically activated macrophages (CAM, also called M1) tend to promote tissue injury while alternatively activated macrophages (AAM, also called M2) are often linked with the mechanisms of wound repair and fibrosis. However, rather than promoting collagen deposition, recent studies suggest that arginase-1-expressing AAM suppress chronic inflammation and fibrosis by inhibiting antigen-specific T cell responses. This unit describes methods to measure arginase activity in macrophages and whole tissues as well as assays to quantify the T cell suppressive activity of AAMs. Modified hydroxyproline and soluble collagen assays that can be used to quantify collagen levels in tissues and brochoalveolar lavage fluid are also described. The protocols in this unit should provide the investigator with all the necessary information required to measure arginase activity and to correlate the observed activity with the progression and resolution of fibrosis.


Assuntos
Técnicas Imunológicas , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Cicatrização , Animais , Arginase/metabolismo , Fibrose , Humanos , Macrófagos/metabolismo , Camundongos , Linfócitos T/imunologia
15.
Am J Physiol Gastrointest Liver Physiol ; 300(5): G723-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21292997

RESUMO

Dysregulated wound healing leads to fibrosis, whereby fibroblasts synthesize excess extracellular matrix and scarring impairs proper organ function. Although fibrotic diseases arise from diverse causes and display heterogeneous features, fibrosis commonly associates with chronic inflammation. Recent discoveries reinforce the idea that communication between fibroblasts, macrophages, and CD4 T cells integrates the processes of wound healing and host defense. Signals between macrophages and fibroblasts can exacerbate, suppress, or reverse fibrosis. Fibroblasts and macrophages are activated by T cells, but their activation also engages negative feedback loops that reduce fibrosis by restraining the immune response, particularly when the Th2 cytokine IL-13 contributes to pathology. Thus the interactions among fibroblasts, macrophages, and CD4 T cells likely play general and critical roles in initiating, perpetuating, and resolving fibrosis in both experimental and clinical conditions.


Assuntos
Fibroblastos/patologia , Cirrose Hepática/patologia , Macrófagos/patologia , Células Th17/patologia , Células Th2/patologia , Animais , Fibroblastos/imunologia , Humanos , Cirrose Hepática/imunologia , Macrófagos/imunologia , Células Th17/imunologia , Células Th2/imunologia
16.
J Immunol ; 185(11): 6426-30, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21037099

RESUMO

IL-2 controls the survival of regulatory T cells (Tregs), but it is unclear whether IL-2 also directly affects Treg suppressive capacity in vivo. We have found that eliminating Bim-dependent apoptosis in IL-2- and CD25-deficient mice restored Treg numbers but failed to cure their lethal autoimmune disease, demonstrating that IL-2-dependent survival and suppressive activity can be uncoupled in Tregs. Treatment with IL-2-anti-IL-2-Ab complexes enhanced the numbers and suppressive capacity of IL-2-deprived Tregs with striking increases in CD25, CTLA-4, and CD39/CD73 expression. Although cytokine treatment induced these suppressive mechanisms in both IL-2(-/-) and IL-2(-/-)Bim(-/-) mice, it only reversed autoimmune disease in the latter. Our results suggest that successful IL-2 therapy of established autoimmune diseases will require a threshold quantity of Tregs present at the start of treatment and show that the suppressive capacity of Tregs critically depends on IL-2 even when Treg survival is independent of this cytokine.


Assuntos
Interleucina-2/fisiologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Anemia Hemolítica Autoimune/genética , Anemia Hemolítica Autoimune/imunologia , Anemia Hemolítica Autoimune/terapia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/deficiência , Subunidade alfa de Receptor de Interleucina-2/genética , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Linfócitos T Reguladores/patologia
17.
Semin Liver Dis ; 30(3): 245-57, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20665377

RESUMO

Macrophages are found in close proximity with collagen-producing myofibroblasts and indisputably play a key role in fibrosis. They produce profibrotic mediators that directly activate fibroblasts, including transforming growth factor-beta1 and platelet-derived growth factor, and control extracellular matrix turnover by regulating the balance of various matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases. Macrophages also regulate fibrogenesis by secreting chemokines that recruit fibroblasts and other inflammatory cells. With their potential to act in both a pro- and antifibrotic capacity, as well as their ability to regulate the activation of resident and recruited myofibroblasts, macrophages and the factors they express are integrated into all stages of the fibrotic process. These various, and sometimes opposing, functions may be performed by distinct macrophage subpopulations, the identification of which is a growing focus of fibrosis research. Although collagen-secreting myofibroblasts once were thought of as the master "producers" of fibrosis, this review will illustrate how macrophages function as the master "regulators" of fibrosis.


Assuntos
Matriz Extracelular/metabolismo , Hepatite/imunologia , Mediadores da Inflamação/metabolismo , Cirrose Hepática/imunologia , Fígado/imunologia , Macrófagos/imunologia , Transdução de Sinais , Animais , Arginase/metabolismo , Citocinas/metabolismo , Células Estreladas do Fígado/imunologia , Hepatite/metabolismo , Hepatite/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Ativação de Macrófagos , Macrófagos/metabolismo , Macrófagos/patologia , Fagocitose , Fator de Crescimento Transformador beta1/metabolismo
18.
J Immunol ; 180(5): 2762-6, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292495

RESUMO

Multiple pathways can induce and maintain peripheral T cell tolerance. The goal of this study was to define the contributions of apoptosis and anergy to the maintenance of self-tolerance to a systemic Ag. Upon transfer into mice expressing OVA systemically, OVA-specific DO11 CD4+ T cells are activated transiently, cease responding, and die. Bim is the essential apoptosis-inducing trigger and apoptosis proceeds despite increased expression of Bcl-2 and Bcl-x. However, preventing apoptosis by eliminating Bim does not restore proliferation or cytokine production by DO11 cells. While Foxp3 is transiently induced, anergy is not associated with the stable development of regulatory T cells. Thus, apoptosis is dispensable for tolerance to a systemic self-Ag and cell-intrinsic anergy is sufficient to tolerize T cells.


Assuntos
Apoptose/imunologia , Anergia Clonal/imunologia , Deleção Clonal/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/administração & dosagem , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Cultivadas , Galinhas , Anergia Clonal/genética , Deleção Clonal/genética , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Ovalbumina/genética , Ovalbumina/imunologia , Proteínas Proto-Oncogênicas/administração & dosagem , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Solubilidade , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante
19.
Immunol Rev ; 226: 19-28, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19161413

RESUMO

Interleukin-2 (IL-2) has multiple, sometimes opposing, functions during an inflammatory response. It is a potent inducer of T-cell proliferation and T-helper 1 (Th1) and Th2 effector T-cell differentiation and provides T cells with a long-lasting competitive advantage resulting in the optimal survival and function of memory cells. In a regulatory role, IL-2 is important for the development, survival, and function of regulatory T cells, it enhances Fas-mediated activation-induced cell death, and it inhibits the development of inflammatory Th17 cells. Thus, in its dual and contrasting functions, IL-2 contributes to both the induction and the termination of inflammatory immune responses.


Assuntos
Inflamação/imunologia , Interleucina-2/imunologia , Receptores de Interleucina-2/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Interleucina-2/deficiência , Receptores de Interleucina-2/deficiência , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
20.
J Exp Med ; 199(12): 1725-30, 2004 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-15210748

RESUMO

Adoptive transfer of ovalbumin (OVA)-specific T cells from the DO.11 TCR transgenic mouse on a Rag(-/-) background into mice expressing OVA in pancreatic islet cells induces acute insulitis and diabetes only if endogenous lymphocytes, including regulatory T cells, are removed. When wild-type OVA-specific/Rag(-/-) T cells, which are all CD25(-), are transferred into islet antigen-expressing mice, peripheral immunization with OVA in adjuvant is needed to induce diabetes. In contrast, naive CTLA-4(-/-)/Rag(-/-) OVA-specific T cells (also CD25(-)) develop into Th1 effectors and induce disease upon recognition of the self-antigen alone. These results suggest that CTLA-4 functions to increase the activation threshold of autoreactive T cells, because in its absence self-antigen is sufficient to trigger autoimmunity without peripheral immunization. Further, CTLA-4 and regulatory T cells act cooperatively to maintain tolerance, indicating that the function of CTLA-4 is independent of regulatory cells, and deficiency of both is required to induce pathologic immune responses against the islet self-antigen.


Assuntos
Antígenos de Diferenciação/imunologia , Diabetes Mellitus Tipo 1/imunologia , Tolerância Imunológica/imunologia , Ilhotas Pancreáticas/imunologia , Linfócitos T/imunologia , Animais , Antígenos CD , Autoantígenos/imunologia , Autoimunidade , Antígeno CTLA-4 , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Diabetes Mellitus Tipo 1/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...