Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Nat Immunol ; 22(8): 1042-1051, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34267375

RESUMO

Pathogens and vaccines that produce persisting antigens can generate expanded pools of effector memory CD8+ T cells, described as memory inflation. While properties of inflating memory CD8+ T cells have been characterized, the specific cell types and tissue factors responsible for their maintenance remain elusive. Here, we show that clinically applied adenovirus vectors preferentially target fibroblastic stromal cells in cultured human tissues. Moreover, we used cell-type-specific antigen targeting to define critical cells and molecules that sustain long-term antigen presentation and T cell activity after adenovirus vector immunization in mice. While antigen targeting to myeloid cells was insufficient to activate antigen-specific CD8+ T cells, genetic activation of antigen expression in Ccl19-cre-expressing fibroblastic stromal cells induced inflating CD8+ T cells. Local ablation of vector-targeted cells revealed that lung fibroblasts support the protective function and metabolic fitness of inflating memory CD8+ T cells in an interleukin (IL)-33-dependent manner. Collectively, these data define a critical fibroblastic niche that underpins robust protective immunity operating in a clinically important vaccine platform.


Assuntos
Adenoviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Interleucina-33/imunologia , Ativação Linfocitária/imunologia , Células Estromais/imunologia , Adenoviridae/genética , Animais , Linhagem Celular Tumoral , Quimiocina CCL19/metabolismo , Quimera/genética , Epitopos de Linfócito T/imunologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Vetores Genéticos/imunologia , Humanos , Pulmão/citologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinação
2.
Nat Immunol ; 17(12): 1388-1396, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27798617

RESUMO

Fibroblastic reticular cells (FRCs) of secondary lymphoid organs form distinct niches for interaction with hematopoietic cells. We found here that production of the cytokine IL-15 by FRCs was essential for the maintenance of group 1 innate lymphoid cells (ILCs) in Peyer's patches and mesenteric lymph nodes. Moreover, FRC-specific ablation of the innate immunological sensing adaptor MyD88 unleashed IL-15 production by FRCs during infection with an enteropathogenic virus, which led to hyperactivation of group 1 ILCs and substantially altered the differentiation of helper T cells. Accelerated clearance of virus by group 1 ILCs precipitated severe intestinal inflammatory disease with commensal dysbiosis, loss of intestinal barrier function and diminished resistance to colonization. In sum, FRCs act as an 'on-demand' immunological 'rheostat' by restraining activation of group 1 ILCs and thereby preventing immunopathological damage in the intestine.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Coronavirus/imunologia , Infecções por Enterobacteriaceae/imunologia , Fibroblastos/imunologia , Interleucina-15/metabolismo , Linfócitos/imunologia , Vírus da Hepatite Murina/imunologia , Animais , Células Cultivadas , Imunidade Inata , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Nódulos Linfáticos Agregados/patologia , Células Th1/imunologia , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo
3.
Immunity ; 48(1): 120-132.e8, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29343433

RESUMO

Group 3 innate lymphoid cells (ILC3s) sense environmental signals and are critical for tissue integrity in the intestine. Yet, which signals are sensed and what receptors control ILC3 function remain poorly understood. Here, we show that ILC3s with a lymphoid-tissue-inducer (LTi) phenotype expressed G-protein-coupled receptor 183 (GPR183) and migrated to its oxysterol ligand 7α,25-hydroxycholesterol (7α,25-OHC). In mice lacking Gpr183 or 7α,25-OHC, ILC3s failed to localize to cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Gpr183 deficiency in ILC3s caused a defect in CP and ILF formation in the colon, but not in the small intestine. Localized oxysterol production by fibroblastic stromal cells provided an essential signal for colonic lymphoid tissue development, and inflammation-induced increased oxysterol production caused colitis through GPR183-mediated cell recruitment. Our findings show that GPR183 promotes lymphoid organ development and indicate that oxysterol-GPR183-dependent positioning within tissues controls ILC3 activity and intestinal homeostasis.


Assuntos
Colite/metabolismo , Linfócitos/metabolismo , Tecido Linfoide/metabolismo , Oxisteróis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Movimento Celular/genética , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Citocinas/metabolismo , Citometria de Fluxo , Imunofluorescência , Ligantes , Linfócitos/patologia , Tecido Linfoide/patologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
4.
Immunity ; 47(1): 80-92.e4, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28709801

RESUMO

Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.


Assuntos
Células Endoteliais/fisiologia , Linfonodos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Organogênese , Animais , Diferenciação Celular , Células Cultivadas , Coristoma , Embrião de Mamíferos , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais
5.
Immunity ; 38(5): 1013-24, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23623380

RESUMO

The stromal scaffold of the lymph node (LN) paracortex is built by fibroblastic reticular cells (FRCs). Conditional ablation of lymphotoxin-ß receptor (LTßR) expression in LN FRCs and their mesenchymal progenitors in developing LNs revealed that LTßR-signaling in these cells was not essential for the formation of LNs. Although T cell zone reticular cells had lost podoplanin expression, they still formed a functional conduit system and showed enhanced expression of myofibroblastic markers. However, essential immune functions of FRCs, including homeostatic chemokine and interleukin-7 expression, were impaired. These changes in T cell zone reticular cell function were associated with increased susceptibility to viral infection. Thus, myofibroblasic FRC precursors are able to generate the basic T cell zone infrastructure, whereas LTßR-dependent maturation of FRCs guarantees full immunocompetence and hence optimal LN function during infection.


Assuntos
Infecções por Coronavirus/imunologia , Linfonodos/citologia , Linfonodos/metabolismo , Miofibroblastos/fisiologia , Linfócitos T/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/imunologia , Interleucina-7/biossíntese , Linfonodos/imunologia , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-beta/biossíntese , Linfotoxina-beta/metabolismo , Glicoproteínas de Membrana/biossíntese , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vírus da Hepatite Murina/imunologia , Miofibroblastos/citologia , Transdução de Sinais
6.
Nat Immunol ; 9(6): 667-75, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18425132

RESUMO

The generation of lymphoid microenvironments in early life depends on the interaction of lymphoid tissue-inducer cells with stromal lymphoid tissue-organizer cells. Whether this cellular interface stays operational in adult secondary lymphoid organs has remained elusive. We show here that during acute infection with lymphocytic choriomeningitis virus, antiviral cytotoxic T cells destroyed infected T cell zone stromal cells, which led to profound disruption of secondary lymphoid organ integrity. Furthermore, the ability of the host to respond to secondary antigens was lost. Restoration of the lymphoid microanatomy was dependent on the proliferative accumulation of lymphoid tissue-inducer cells in secondary lymphoid organs during the acute phase of infection and lymphotoxin alpha(1)beta(2) signaling. Thus, crosstalk between lymphoid tissue-inducer cells and stromal cells is reactivated in adults to maintain secondary lymphoid organ integrity and thereby contributes to the preservation of immunocompetence.


Assuntos
Infecções por Arenaviridae/imunologia , Sistema Linfático/fisiologia , Vírus da Coriomeningite Linfocítica/imunologia , Células Estromais/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Infecções por Arenaviridae/sangue , Infecções por Arenaviridae/virologia , Ativação Linfocitária/imunologia , Tecido Linfoide/imunologia , Especificidade de Órgãos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores
7.
PLoS Biol ; 14(7): e1002515, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27415420

RESUMO

Fibroblastic reticular cells (FRCs) form the cellular scaffold of lymph nodes (LNs) and establish distinct microenvironmental niches to provide key molecules that drive innate and adaptive immune responses and control immune regulatory processes. Here, we have used a graph theory-based systems biology approach to determine topological properties and robustness of the LN FRC network in mice. We found that the FRC network exhibits an imprinted small-world topology that is fully regenerated within 4 wk after complete FRC ablation. Moreover, in silico perturbation analysis and in vivo validation revealed that LNs can tolerate a loss of approximately 50% of their FRCs without substantial impairment of immune cell recruitment, intranodal T cell migration, and dendritic cell-mediated activation of antiviral CD8+ T cells. Overall, our study reveals the high topological robustness of the FRC network and the critical role of the network integrity for the activation of adaptive immune responses.


Assuntos
Comunicação Celular/imunologia , Movimento Celular/imunologia , Fibroblastos/imunologia , Linfonodos/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Contagem de Células , Movimento Celular/genética , Quimiocina CCL19/genética , Quimiocina CCL19/imunologia , Quimiocina CCL19/metabolismo , Células Dendríticas/citologia , Células Dendríticas/imunologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Linfonodos/citologia , Linfonodos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo
8.
Eur J Immunol ; 45(8): 2218-31, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-25973789

RESUMO

The thymic epithelium forms specialized niches to enable thymocyte differentiation. While the common epithelial progenitor of medullary and cortical thymic epithelial cells (mTECs and cTECs) is well defined, early stages of mTEC lineage specification have remained elusive. Here, we utilized in vivo targeting of mTECs to resolve their differentiation pathways and to determine whether mTEC progenitors participate in thymocyte education. We found that mTECs descend from a lineage committed, podoplanin (PDPN)-expressing progenitor located at the cortico-medullary junction. PDPN(+) junctional TECs (jTECs) represent a distinct TEC population that builds the thymic medulla, but only partially supports negative selection and thymocyte differentiation. Moreover, conditional gene targeting revealed that abrogation of alternative NF-κB pathway signaling in the jTEC stage completely blocked mTEC development. Taken together, this study identifies jTECs as lineage-committed mTEC progenitors and shows that NF-κB-dependent progression of jTECs to mTECs is critical to secure central tolerance.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Glicoproteínas de Membrana/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Células-Tronco/imunologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/citologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , NF-kappa B/genética , Transdução de Sinais/genética , Células-Tronco/citologia , Timo/citologia
9.
J Immunol ; 192(11): 5192-200, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24778443

RESUMO

Abs play a significant role in protection against the intracellular bacterium Salmonella Typhi. In this article, we investigated how long-term protective IgM responses can be elicited by a S. Typhi outer-membrane protein C- and F-based subunit vaccine (porins). We found that repeated Ag exposure promoted a CD4(+) T cell-dependent germinal center reaction that generated mutated IgM-producing B cells and was accompanied by a strong expansion of IFN-γ-secreting T follicular helper cells. Genetic ablation of individual cytokine receptors revealed that both IFN-γ and IL-17 are required for optimal germinal center reactions and production of porin-specific memory IgM(+) B cells. However, more profound reduction of porin-specific IgM B cell responses in the absence of IFN-γR signaling indicated that this cytokine plays a dominant role. Importantly, mutated IgM mAbs against porins exhibited bactericidal capacity and efficiently augmented S. Typhi clearance. In conclusion, repeated vaccination with S. Typhi porins programs type I T follicular helper cell responses that contribute to the diversification of B cell memory and promote the generation of protective IgM Abs.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Centro Germinativo/imunologia , Imunoglobulina M/imunologia , Memória Imunológica , Interferon gama/imunologia , Salmonella typhi/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Feminino , Centro Germinativo/patologia , Humanos , Interferon gama/genética , Masculino , Camundongos , Camundongos Knockout , Vacinas contra Salmonella/genética , Vacinas contra Salmonella/imunologia , Febre Tifoide/genética , Febre Tifoide/imunologia , Febre Tifoide/patologia , Febre Tifoide/prevenção & controle
10.
Blood ; 121(20): 4101-9, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-23558016

RESUMO

It is not known how naive B cells compute divergent chemoattractant signals of the T-cell area and B-cell follicles during in vivo migration. Here, we used two-photon microscopy of peripheral lymph nodes (PLNs) to analyze the prototype G-protein-coupled receptors (GPCRs) CXCR4, CXCR5, and CCR7 during B-cell migration, as well as the integrin LFA-1 for stromal guidance. CXCR4 and CCR7 did not influence parenchymal B-cell motility and distribution, despite their role during B-cell arrest in venules. In contrast, CXCR5 played a nonredundant role in B-cell motility in follicles and in the T-cell area. B-cell migration in the T-cell area followed a random guided walk model, arguing against directed migration in vivo. LFA-1, but not α4 integrins, contributed to B-cell motility in PLNs. However, stromal network guidance was LFA-1 independent, uncoupling integrin-dependent migration from stromal attachment. Finally, we observed that despite a 20-fold reduction of chemokine expression in virus-challenged PLNs, CXCR5 remained essential for B-cell screening of antigen-presenting cells. Our data provide an overview of the contribution of prototype GPCRs and integrins during naive B-cell migration and shed light on the local chemokine availability that these cells compute.


Assuntos
Linfócitos B/fisiologia , Comunicação Celular/fisiologia , Quimiocinas/fisiologia , Quimiotaxia de Leucócito/genética , Antígeno-1 Associado à Função Linfocitária/metabolismo , Receptores CCR7/fisiologia , Receptores CXCR4/fisiologia , Receptores CXCR5/fisiologia , Células Estromais/fisiologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/fisiologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Comunicação Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Quimiocinas/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Feminino , Deleção de Genes , Antígeno-1 Associado à Função Linfocitária/fisiologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores CCR7/genética , Receptores CCR7/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Células Estromais/metabolismo
11.
Eur J Immunol ; 43(12): 3233-43, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23963995

RESUMO

Attenuation of T cell-mediated damage of blood endothelial cells (BECs) in transplanted organs is important to prevent transplant vasculopathy (TV) and chronic rejection. Here, we assessed the importance of minor histocompatibility antigen (mHA) distribution and different coinhibitory molecules for T cell-BEC interaction. A transgenic mHA was directed specifically to BECs using the Tie2 promoter and cellular interactions were assessed in graft-versus-host disease-like and heterotopic heart transplantation settings. We found that cognate CD4(+) T-cell help was critical for the activation of BEC-specific CD8(+) T cells. However, systemic mHA expression on BECs efficiently attenuated adoptively transferred, BEC-specific CD4(+) and CD8(+) T cells and hence prevented tissue damage, whereas restriction of mHA expression to heart BECs precipitated the development of TV. Importantly, the lack of the coinhibitory molecules programmed death-1 (PD-1) and B and T lymphocyte attenuator fostered the initial activation of BEC-specific CD4(+) T cells, but did not affect development of TV. In contrast, TV was significantly augmented in the absence of PD-1 on BEC-specific CD8(+) T cells. Taken together, these results indicate that antigen distribution in the vascular bed determines the impact of coinhibition and, as a consequence, critically impinges on T cell-mediated vascular immunopathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração , Antígenos de Histocompatibilidade Menor/imunologia , Doenças Vasculares/imunologia , Aloenxertos , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Rejeição de Enxerto/genética , Rejeição de Enxerto/metabolismo , Rejeição de Enxerto/patologia , Camundongos , Camundongos Knockout , Antígenos de Histocompatibilidade Menor/biossíntese , Antígenos de Histocompatibilidade Menor/genética , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
12.
Blood ; 120(24): 4675-83, 2012 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22955921

RESUMO

Nonhematopoietic stromal cells of secondary lymphoid organs form important scaffold and fluid transport structures, such as lymph node (LN) trabeculae, lymph vessels, and conduits. Furthermore, through the production of chemokines and cytokines, these cells generate a particular microenvironment that determines lymphocyte positioning and supports lymphocyte homeostasis. IL-7 is an important stromal cell-derived cytokine that has been considered to be derived mainly from T-cell zone fibroblastic reticular cells. We show here that lymphatic endothelial cells (LECs) are a prominent source of IL-7 both in human and murine LNs. Using bacterial artificial chromosome transgenic IL-7-Cre mice, we found that fibroblastic reticular cells and LECs strongly up-regulated IL-7 expression during LN remodeling after viral infection and LN reconstruction after avascular transplantation. Furthermore, IL-7-producing stromal cells contributed to de novo formation of LyveI-positive lymphatic structures connecting reconstructed LNs with the surrounding tissue. Importantly, diphtheria toxin-mediated depletion of IL-7-producing stromal cells completely abolished LN reconstruction. Taken together, this study identifies LN LECs as a major source of IL-7 and shows that IL-7-producing stromal cells are critical for reconstruction and remodeling of the distinct LN microenvironment.


Assuntos
Células Endoteliais/metabolismo , Interleucina-7/metabolismo , Linfonodos/metabolismo , Células Estromais/metabolismo , Adulto , Animais , Linhagem Celular , Proliferação de Células , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Imuno-Histoquímica , Interleucina-7/genética , Rim/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Linfonodos/embriologia , Linfonodos/transplante , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Immunol ; 189(2): 841-9, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22696443

RESUMO

Abs confer protection from secondary infection with Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. In this study, we demonstrate that Ab-mediated protection is effective across L. pneumophila serogroups, suggesting that Abs specific for conserved protein Ags are sufficient to mediate this protective effect. We used two independent methods to identify immunogenic L. pneumophila protein Ags, namely, the screening of a λ phage library representing the complete L. pneumophila genome and two-dimensional gel electrophoresis combined with Western blot analysis and protein spot identification by mass spectrometry. A total of 30 novel L. pneumophila B cell Ags were identified, the majority of which are located in or associated with the bacterial membrane, where they are accessible for Abs and, therefore, likely to be relevant for Ab-mediated protection against L. pneumophila. Selected B cell Ags were recombinantly expressed and tested in a vaccination protocol. Mice immunized with either single-protein Ags or an Ag combination showed reduced bacterial titers in bronchoalveolar lavage and lung after L. pneumophila challenge. To determine the clinical relevance of these findings, we tested Legionnaires' disease patient sera for reactivity with the identified L. pneumophila Ags. The recognized Ags were indeed conserved across host species, because Abs specific for all three selected Ags could be detected in patient sera, rendering the identified protein Ags potential vaccine candidates.


Assuntos
Antígenos de Bactérias/isolamento & purificação , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/microbiologia , Legionella pneumophila/imunologia , Doença dos Legionários/imunologia , Administração Intranasal , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/uso terapêutico , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Subpopulações de Linfócitos B/metabolismo , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/síntese química , Vacinas Bacterianas/imunologia , Bacteriófago lambda/genética , Bacteriófago lambda/imunologia , Sequência Conservada/imunologia , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Legionella pneumophila/patogenicidade , Doença dos Legionários/sangue , Doença dos Legionários/prevenção & controle , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
14.
Eur J Immunol ; 42(9): 2311-21, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22730043

RESUMO

Myocarditis is a potentially lethal inflammatory heart disease of children and young adults that frequently leads to dilated cardiomyopathy (DCM). Since diagnostic procedures and efficient therapies are lacking, it is important to characterize the critical immune effector pathways underlying the initial cardiac inflammation and the transition from myocarditis to DCM. We describe here a T-cell receptor (TCR) transgenic mouse model with spontaneously developing autoimmune myocarditis that progresses to lethal DCM. Cardiac magnetic resonance imaging revealed early inflammation-associated changes in the ventricle wall including transient thickening of the left ventricle wall. Furthermore, we found that IFN-γ was a major effector cytokine driving the initial inflammatory process and that the cooperation of IFN-γ and IL-17A was essential for the development of the progressive disease. This novel TCR transgenic mouse model permits the identification of the central pathophysiological and immunological processes involved in the transition from autoimmune myocarditis to DCM.


Assuntos
Doenças Autoimunes/imunologia , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/patologia , Miocardite/imunologia , Miocardite/patologia , Células Th1/imunologia , Células Th17/imunologia , Animais , Autoantígenos/imunologia , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Inflamação/imunologia , Inflamação/patologia , Interferon gama/imunologia , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/imunologia , Remodelação Ventricular/imunologia
15.
Blood ; 115(23): 4725-33, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20185585

RESUMO

Adaptive immune responses are characterized by substantial restructuring of secondary lymphoid organs. The molecular and cellular factors responsible for virus-induced lymphoid remodeling are not well known to date. Here we applied optical projection tomography, a mesoscopic imaging technique, for a global analysis of the entire 3-dimensional structure of mouse peripheral lymph nodes (PLNs), focusing on B-cell areas and high endothelial venule (HEV) networks. Structural homeostasis of PLNs was characterized by a strict correlation between total PLN volume, B-cell volume, B-cell follicle number, and HEV length. After infection with lymphocytic choriomeningitis virus, we observed a substantial, lymphotoxin (LT) beta-receptor-dependent reorganization of the PLN microarchitecture, in which an initial B-cell influx was followed by 3-fold increases in PLN volume and HEV network length on day 8 after infection. Adoptive transfer experiments revealed that virus-induced PLN and HEV network remodeling required LTalpha(1)beta(2)-expressing B cells, whereas the inhibition of vascular endothelial growth factor-A signaling pathways had no significant effect on PLN expansion. In summary, lymphocytic choriomeningitis virus-induced PLN growth depends on a vascular endothelial growth factor-A-independent, LT- and B cell-dependent morphogenic pathway, as revealed by an in-depth mesoscopic analysis of the global PLN structure.


Assuntos
Imunidade Adaptativa , Linfócitos B/imunologia , Linfonodos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Animais , Linfócitos B/patologia , Linfócitos B/virologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Homeostase/genética , Homeostase/imunologia , Linfonodos/patologia , Linfonodos/virologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/biossíntese , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Receptor beta de Linfotoxina/biossíntese , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Urol Int ; 88(3): 343-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22378292

RESUMO

INTRODUCTION: Few published in vitro studies have shown antitumor drug action or possible synergistic effects of fluoroquinolones. To assess the potential role of combination therapy, cytotoxic effects of ciprofloxacin and epirubicin alone and in combination were determined. MATERIAL AND METHODS: Human urothelial cancer cell lines HT1197 and HT1376 were exposed in vitro for 1 h to different concentrations of epirubicin (0.02-2 mg/ml) and for 72 h to ciprofloxacin (0.004-0.8 mg/ml). Cytotoxicity was determined using the microculture tetrazolium assay and flow cytometry. Synergistic cytotoxic effects were determined by calculating combination indices. RESULTS: Median effect concentrations of epirubicin for HT1376 and HT1197 cells were as low as 124 and 117 µg/ml, respectively. Ciprofloxacin-treated cells exhibited profound cytotoxic effects at concentrations of 50-100 µg/ml, which is far below the intravesical concentration reached by standard oral application. In addition, a pronounced synergistic effect was found when the two treatments were combined. CONCLUSIONS: This study provides evidence that ciprofloxacin and epirubicin exhibit synergistic cytotoxic effects in vitro. After confirmatory animal experiments, future clinical studies of adjuvant chemotherapy after transurethral bladder resection may include treatment arms with combinations of fluoroquinolones based on the observed synergistic effects to reduce both side effects and costs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Urológicas/patologia , Urotélio/efeitos dos fármacos , Adulto , Linhagem Celular Tumoral , Ciprofloxacina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Epirubicina/farmacologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Urotélio/patologia
17.
Nat Commun ; 13(1): 2027, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35440118

RESUMO

Innate lymphoid cells (ILCs) govern immune cell homeostasis in the intestine and protect the host against microbial pathogens. Various cell-intrinsic pathways have been identified that determine ILC development and differentiation. However, the cellular components that regulate ILC sustenance and function in the intestinal lamina propria are less known. Using single-cell transcriptomic analysis of lamina propria fibroblasts, we identify fibroblastic reticular cells (FRCs) that underpin cryptopatches (CPs) and isolated lymphoid follicles (ILFs). Genetic ablation of lymphotoxin-ß receptor expression in Ccl19-expressing FRCs blocks the maturation of CPs into mature ILFs. Interactome analysis shows the major niche factors and processes underlying FRC-ILC crosstalk. In vivo validation confirms that a sustained lymphotoxin-driven feedforward loop of FRC activation including IL-7 generation is critical for the maintenance of functional ILC populations. In sum, our study indicates critical fibroblastic niches within the intestinal lamina propria that control ILC homeostasis and functionality and thereby secure protective gut immunity.


Assuntos
Imunidade Inata , Linfócitos , Fibroblastos , Homeostase , Intestinos
18.
Nat Commun ; 12(1): 4734, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34354077

RESUMO

The tumor microenvironment (TME) is a complex amalgam of tumor cells, immune cells, endothelial cells and fibroblastic stromal cells (FSC). Cancer-associated fibroblasts are generally seen as tumor-promoting entity. However, it is conceivable that particular FSC populations within the TME contribute to immune-mediated tumor control. Here, we show that intratumoral treatment of mice with a recombinant lymphocytic choriomeningitis virus-based vaccine vector expressing a melanocyte differentiation antigen resulted in T cell-dependent long-term control of melanomas. Using single-cell RNA-seq analysis, we demonstrate that viral vector-mediated transduction reprogrammed and activated a Cxcl13-expressing FSC subset that show a pronounced immunostimulatory signature and increased expression of the inflammatory cytokine IL-33. Ablation of Il33 gene expression in Cxcl13-Cre-positive FSCs reduces the functionality of intratumoral T cells and unleashes tumor growth. Thus, reprogramming of FSCs by a self-antigen-expressing viral vector in the TME is critical for curative melanoma treatment by locally sustaining the activity of tumor-specific T cells.


Assuntos
Melanoma Experimental/terapia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/patologia , Técnicas de Reprogramação Celular/métodos , Quimiocina CXCL13/genética , Quimiocina CXCL13/imunologia , Feminino , Vetores Genéticos , Interleucina-33/deficiência , Interleucina-33/genética , Interleucina-33/imunologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Vírus da Coriomeningite Linfocítica/genética , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Estromais/imunologia , Células Estromais/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Microambiente Tumoral/imunologia
19.
Blood ; 111(9): 4588-95, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18195091

RESUMO

Endothelial cells (ECs) presenting minor histocompatibility antigen (mhAg) are major target cells for alloreactive effector CD8(+) T cells during chronic transplant rejection and graft-versus-host disease (GVHD). The contribution of ECs to T-cell activation, however, is still a controversial issue. In this study, we have assessed the antigen-presenting capacity of ECs in vivo using a transgenic mouse model with beta-galactosidase (beta-gal) expression confined to the vascular endothelium (Tie2-LacZ mice). In a GVHD-like setting with adoptive transfer of beta-gal-specific T-cell receptor-transgenic T cells, beta-gal expression by ECs was not sufficient to either activate or tolerize CD8(+) T cells. Likewise, transplantation of fully vascularized heart or liver grafts from Tie2-LacZ mice into nontransgenic recipients did not suffice to activate beta-gal-specific CD8(+) T cells, indicating that CD8(+) T-cell responses against mhAg cannot be initiated by ECs. Moreover, we could show that spontaneous activation of beta-gal-specific CD8(+) T cells in Tie2-LacZ mice was exclusively dependent on CD11c(+) dendritic cells (DCs), demonstrating that mhAgs presented by ECs remain immunologically ignored unless presentation by DCs is granted.


Assuntos
Células Endoteliais/imunologia , Antígenos de Histocompatibilidade Menor/análise , Imunologia de Transplantes , Animais , Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Endotélio Vascular/citologia , Camundongos , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor/imunologia , Transplante de Órgãos/efeitos adversos
20.
J Immunol ; 181(4): 2522-32, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18684943

RESUMO

Caspase-8, the proximal enzyme in the death-induction pathway of the TNF/nerve growth factor receptor family, is activated upon juxtaposition of its molecules within the receptor complexes and is then self-processed. Caspase-8 also contributes to the regulation of cell survival and growth, but little is known about the similarities or the differences between the mechanisms of these nonapoptotic functions and of the enzyme's apoptotic activity. In this study, we report that in bacterial artificial chromosome-transgenic mice, in which the aspartate residue upstream of the initial self-processing site in caspase-8 (D387) was replaced by alanine, induction of cell death by Fas is compromised. However, in contrast to caspase-8-deficient mice, which die in utero at mid-gestation, the mice mutated at D387 were born alive and seemed to develop normally. Moreover, mice with the D387A mutation showed normal in vitro growth responses of T lymphocytes to stimulation of their Ag receptor as well as of B lymphocytes to stimulation by LPS, normal differentiation of bone marrow macrophage precursors in response to M-CSF, and normal generation of myeloid colonies by the bone marrow hematopoietic progenitors, all of which are compromised in cells deficient in caspase-8. These finding indicated that self-processing of activated caspase-8 is differentially involved in the different functions of this enzyme: it is needed for the induction of cell death through the extrinsic cell death pathway but not for nonapoptotic functions of caspase-8.


Assuntos
Síndrome de Alstrom , Apoptose/genética , Caspase 8/genética , Cromossomos Artificiais Bacterianos/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Mutação , Processamento Pós-Transcricional do RNA/imunologia , Animais , Formação de Anticorpos/genética , Apresentação de Antígeno/genética , Apoptose/imunologia , Caspase 8/metabolismo , Linhagem Celular , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Clonais , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Proteína Ligante Fas/toxicidade , Humanos , Ativação Linfocitária/genética , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA