Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 712632, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335629

RESUMO

Lymphotoxin beta receptor (LTßR) is a promising therapeutic target in autoimmune and infectious diseases as well as cancer. Mice with genetic inactivation of LTßR display multiple defects in development and organization of lymphoid organs, mucosal immune responses, IgA production and an autoimmune phenotype. As these defects are imprinted in embryogenesis and neonate stages, the impact of LTßR signaling in adulthood remains unclear. Here, to overcome developmental defects, we generated mice with inducible ubiquitous genetic inactivation of LTßR in adult mice (iLTßRΔ/Δ mice) and redefined the role of LTßR signaling in organization of lymphoid organs, immune response to mucosal bacterial pathogen, IgA production and autoimmunity. In spleen, postnatal LTßR signaling is required for development of B cell follicles, follicular dendritic cells (FDCs), recruitment of neutrophils and maintenance of the marginal zone. Lymph nodes of iLTßRΔ/Δ mice were reduced in size, lacked FDCs, and had disorganized subcapsular sinus macrophages. Peyer`s patches were smaller in size and numbers, and displayed reduced FDCs. The number of isolated lymphoid follicles in small intestine and colon were also reduced. In contrast to LTßR-/- mice, iLTßRΔ/Δ mice displayed normal thymus structure and did not develop signs of systemic inflammation and autoimmunity. Further, our results suggest that LTßR signaling in adulthood is required for homeostasis of neutrophils, NK, and iNKT cells, but is dispensable for the maintenance of polyclonal IgA production. However, iLTßRΔ/Δ mice exhibited an increased sensitivity to C. rodentium infection and failed to develop pathogen-specific IgA responses. Collectively, our study uncovers new insights of LTßR signaling in adulthood for the maintenance of lymphoid organs, neutrophils, NK and iNKT cells, and IgA production in response to mucosal bacterial pathogen.


Assuntos
Envelhecimento/imunologia , Tecido Linfoide/imunologia , Receptor beta de Linfotoxina/fisiologia , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/imunologia , Autoimunidade , Moléculas de Adesão Celular/metabolismo , Quimiocinas/metabolismo , Citrobacter rodentium/imunologia , Cruzamentos Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/imunologia , Imunoglobulina A/biossíntese , Imunoglobulina A/imunologia , Inflamação , Células Matadoras Naturais/imunologia , Tecido Linfoide/citologia , Receptor beta de Linfotoxina/biossíntese , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos MRL lpr , Camundongos Transgênicos , Neutrófilos/imunologia , Deleção de Sequência , Organismos Livres de Patógenos Específicos , Esplenomegalia/imunologia
2.
J Immunol ; 197(7): 2665-72, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27549174

RESUMO

The recruitment of lymphoid progenitors to the thymus is essential to sustain T cell production throughout life. Importantly, it also limits T lineage regeneration following bone marrow transplantation, and so contributes to the secondary immunodeficiency that is caused by delayed immune reconstitution. Despite this significance, the mechanisms that control thymus colonization are poorly understood. In this study, we show that in both the steady-state and after bone marrow transplant, lymphotoxin ß receptor (LTßR) controls entry of T cell progenitors to the thymus. We show that this requirement maps to thymic stroma, further underlining the key importance of this TNFR superfamily member in regulation of thymic microenvironments. Importantly, analysis of the requirement for LTßR in relationship to known regulators of thymus seeding suggests that it acts independently of its regulation of thymus-homing chemokines. Rather, we show that LTßR differentially regulates intrathymic expression of adhesion molecules known to play a role in T cell progenitor entry to the thymus. Finally, Ab-mediated in vivo LTßR stimulation following bone marrow transplant enhances initial thymus recovery and boosts donor-derived T cell numbers, which correlates with increased adhesion molecule expression by thymic stroma. Collectively, we reveal a novel link between LTßR and thymic stromal cells in thymus colonization, and highlight its potential as an immunotherapeutic target to boost T cell reconstitution after transplantation.


Assuntos
Movimento Celular , Receptor beta de Linfotoxina/imunologia , Células-Tronco/citologia , Linfócitos T/citologia , Timo/citologia , Animais , Receptor beta de Linfotoxina/deficiência , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/imunologia , Linfócitos T/imunologia , Timo/imunologia
3.
Cell Rep ; 14(10): 2375-88, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947077

RESUMO

Despite the key role of primary and secondary lymphoid organ stroma in immunity, our understanding of the heterogeneity and ontogeny of these cells remains limited. Here, we identify a functionally distinct subset of BP3(-)PDPN(+)PDGFRß(+)/α(+)CD34(+) stromal adventitial cells in both lymph nodes (LNs) and thymus that is located within the vascular niche surrounding PDPN(-)PDGFRß(+)/α(-)Esam-1(+)ITGA7(+) pericytes. CD34(+) adventitial cells developed in late embryonic thymus and in postnatal LNs and in the thymus originated, along with pericytes, from a common anlage-seeding progenitor population. Using lymphoid organ re-aggregate grafts, we demonstrate that adult CD34(+) adventitial cells are capable of differentiating into multiple lymphoid stroma-like subsets including pericyte-, FRC-, MRC-, and FDC-like cells, the development of which was lymphoid environment-dependent. These findings extend the current understanding of lymphoid mesenchymal cell heterogeneity and highlight a role of the CD34(+) adventitia as a potential ubiquitous source of lymphoid stromal precursors in postnatal tissues.


Assuntos
Antígenos CD34/metabolismo , Células Estromais/metabolismo , ADP-Ribosil Ciclase/genética , ADP-Ribosil Ciclase/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Citometria de Fluxo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Imuno-Histoquímica , Linfonodos/citologia , Linfonodos/metabolismo , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pericitos/citologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células Estromais/citologia , Timo/citologia , Timo/metabolismo
4.
J Leukoc Biol ; 100(1): 103-10, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26657790

RESUMO

The lymphotoxin signaling pathway plays an important role in the homeostasis and function of peripheral and mucosal dendritic cells, and dendritic cell-intrinsic lymphotoxin ß receptor expression is required for optimal responses to opportunistic intestinal bacteria. However, it is unknown whether dendritic cell-intrinsic lymphotoxin ß receptor signaling is required for responses to intestinal viral infections. We explored this question by orally administrating murine rotavirus to chimeric mice that lack lymphotoxin ß receptor signaling in the myeloid compartment but retain lymphoid tissues. We found that although clearance of rotavirus was unimpaired in the lymphotoxin ß receptor(-/-) → wild-type chimeric mice compared with wild-type → wild-type chimeric mice, IFN-γ-producing CD8(+) and CD4(+) T cells were significantly increased in the small intestinal lamina propria of lymphotoxin ß receptor(-/-) → wild-type chimeric mice. In contrast, IL-17-producing CD4(+) T cells were reduced in lymphotoxin ß receptor(-/-) → wild-type chimeric mice in the steady state, and this reduction persisted after rotavirus inoculation. In spite of this altered cytokine profile in the small intestinal lamina propria of lymphotoxin ß receptor(-/-) → wild-type chimeric mice, the local production of rotavirus-specific IgA was unperturbed. Collectively, our results demonstrate that lymphotoxin ß receptor signaling in radio-sensitive myeloid cells regulates the balance of IFN-γ and IL-17 cytokine production within the small intestinal lamina propria; however, these perturbations do not affect mucosal antiviral IgA responses.


Assuntos
Citocinas/metabolismo , Células Dendríticas/imunologia , Mucosa Intestinal/imunologia , Receptor beta de Linfotoxina/deficiência , Infecções por Rotavirus/imunologia , Rotavirus/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Imunidade nas Mucosas/imunologia , Imunoglobulina A/imunologia , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Rotavirus/virologia
5.
J Clin Invest ; 125(7): 2877-90, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098211

RESUMO

Systemic lupus erythematosus (SLE) is a severe autoimmune disease that is associated with increased circulating apoptotic cell autoantigens (AC-Ags) as well as increased type I IFN signaling. Here, we describe a pathogenic mechanism in which follicular translocation of marginal zone (MZ) B cells in the spleens of BXD2 lupus mice disrupts marginal zone macrophages (MZMs), which normally clear AC debris and prevent follicular entry of AC-Ags. Phagocytosis of ACs by splenic MZMs required the megakaryoblastic leukemia 1 (MKL1) transcriptional coactivator-mediated mechanosensing pathway, which was maintained by MZ B cells through expression of membrane lymphotoxin-α1ß2 (mLT). Specifically, type I IFN-induced follicular shuttling of mLT-expressing MZ B cells disengaged interactions between these MZ B cells and LTß receptor-expressing MZMs, thereby downregulating MKL1 in MZMs. Loss of MKL1 expression in MZMs led to defective F-actin polymerization, inability to clear ACs, and, eventually, MZM dissipation. Aggregation of plasmacytoid DCs in the splenic perifollicular region, follicular translocation of MZ B cells, and loss of MKL1 and MZMs were also observed in an additional murine lupus model and in the spleens of patients with SLE. Collectively, the results suggest that lupus might be interrupted by strategies that maintain or enhance mechanosensing signaling in the MZM barrier to prevent follicular entry of AC-Ags.


Assuntos
Apoptose/imunologia , Interferon Tipo I/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Mecanotransdução Celular/imunologia , Animais , Autoanticorpos/biossíntese , Linfócitos B/imunologia , Linfócitos B/patologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Macrófagos/imunologia , Macrófagos/patologia , Mecanotransdução Celular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores Imunológicos/metabolismo , Fator de Resposta Sérica/deficiência , Fator de Resposta Sérica/genética , Baço/imunologia , Baço/patologia , Transativadores/deficiência , Transativadores/genética
6.
Circ Res ; 116(8): e57-68, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25740843

RESUMO

RATIONALE: Lymphotoxin ß receptor (LTbR) regulates immune cell trafficking and communication in inflammatory diseases. However, the role of LTbR in atherosclerosis is still unclear. OBJECTIVE: The aim of this study was to elucidate the role of LTbR in atherosclerosis. METHODS AND RESULTS: After 15 weeks of feeding a Western-type diet, mice double-deficient in apolipoprotein E and LTbR (apoE(-/-)/LTbR(-/-)) exhibited lower aortic plaque burden than did apoE(-/-) littermates. Macrophage content at the aortic root and in the aorta was reduced, as determined by immunohistochemistry and flow cytometry. In line with a decrease in plaque inflammation, chemokine (C-C motif) ligand 5 (Ccl5) and other chemokines were transcriptionally downregulated in aortic tissue from apoE(-/-)/LTbR(-/-) mice. Moreover, bone marrow chimeras demonstrated that LTbR deficiency in hematopoietic cells mediated the atheroprotection. Furthermore, during atheroprogression, apoE(-/-) mice exhibited increased concentrations of cytokines, for example, Ccl5, whereas apoE(-/-)/LTbR(-/-) mice did not. Despite this decreased plaque macrophage content, flow cytometric analysis showed that the numbers of circulating lymphocyte antigen 6C (Ly6C)(low) monocytes were markedly elevated in apoE(-/-)/LTbR(-/-) mice. The influx of these cells into atherosclerotic lesions was significantly reduced, whereas apoptosis and macrophage proliferation in atherosclerotic lesions were unaffected. Gene array analysis pointed to chemokine (C-C motif) receptor 5 as the most regulated pathway in isolated CD115(+) cells in apoE(-/-)/LTbR(-/-) mice. Furthermore, stimulating monocytes from apoE(-/-) mice with agonistic anti-LTbR antibody or the natural ligand lymphotoxin-α1ß2, increased Ccl5 mRNA expression. CONCLUSIONS: These findings suggest that LTbR plays a role in macrophage-driven inflammation in atherosclerotic lesions, probably by augmenting the Ccl5-mediated recruitment of monocytes.


Assuntos
Aorta/efeitos dos fármacos , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Receptor beta de Linfotoxina/deficiência , Animais , Antígenos Ly/metabolismo , Aorta/imunologia , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/diagnóstico , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Transplante de Medula Óssea , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiotaxia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Receptor beta de Linfotoxina/genética , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Placa Aterosclerótica , Fatores de Tempo , Transcrição Gênica , Quimeras de Transplante
7.
Mucosal Immunol ; 8(2): 403-13, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25183367

RESUMO

The immune mechanisms regulating epithelial cell repair after injury remain poorly defined. We demonstrate here that lymphotoxin beta receptor (LTßR) signaling in intestinal epithelial cells promotes self-repair after mucosal damage. Using a conditional gene-targeted approach, we demonstrate that LTßR signaling in intestinal epithelial cells is essential for epithelial interleukin-23 (IL-23) production and protection against epithelial injury. We further show that epithelial-derived IL-23 promotes mucosal wound healing by inducing the IL-22-mediated proliferation and survival of epithelial cells and mucus production. Additionally, we identified CD4(-)CCR6(+)T-bet(-) RAR-related orphan receptor gamma t (RORγt)(+) lymphoid tissue inducer cells as the main producers of protective IL-22 after epithelial damage. Thus, our results reveal a novel role for LTßR signaling in epithelial cells in the regulation of intestinal epithelial cell homeostasis to limit mucosal damage.


Assuntos
Interleucina-23/biossíntese , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Receptor beta de Linfotoxina/metabolismo , Transdução de Sinais , Animais , Colite/genética , Colite/imunologia , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Expressão Gênica , Homeostase , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/patologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Knockout , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Cicatrização , Interleucina 22
8.
J Immunol ; 190(10): 5110-7, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23585674

RESUMO

Medullary thymic epithelial cells (mTECs) play a pivotal role in the establishment of self-tolerance in T cells by ectopically expressing various tissue-restricted self-Ags and by chemoattracting developing thymocytes. The nuclear protein Aire expressed by mTECs contributes to the promiscuous expression of self-Ags, whereas CCR7-ligand (CCR7L) chemokines expressed by mTECs are responsible for the attraction of positively selected thymocytes. It is known that lymphotoxin signals from the positively selected thymocytes preferentially promote the expression of CCR7L rather than Aire in postnatal mTECs. However, it is unknown how lymphotoxin signals differentially regulate the expression of CCR7L and Aire in mTECs and whether CCR7L-expressing mTECs and Aire-expressing mTECs are distinct populations. In this study, we show that the majority of postnatal mTECs that express CCL21, a CCR7L chemokine, represent an mTEC subpopulation distinct from the Aire-expressing mTEC subpopulation. Interestingly, the development of CCL21-expressing mTECs, but not Aire-expressing mTECs, is impaired in mice deficient in the lymphotoxin ß receptor. These results indicate that postnatal mTECs consist of heterogeneous subsets that differ in the expression of CCL21 and Aire, and that lymphotoxin ß receptor regulates the development of the CCL21-expressing subset rather than the Aire-expressing subset of postnatal mTECs.


Assuntos
Quimiocina CCL21/biossíntese , Receptor beta de Linfotoxina/metabolismo , Timo/metabolismo , Animais , Diferenciação Celular , Células Epiteliais , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR7 , Transdução de Sinais , Fatores de Transcrição , Proteína AIRE
9.
J Immunol ; 188(7): 3426-33, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22357629

RESUMO

Our previous studies indicated that lymphotoxin ß receptor (LTßR) activation controls and downregulates inflammatory reactions. In this study, we report that LTßR activation on primary mouse macrophages results in induction of tripartite motif containing (TRIM) 30α, which negatively regulates NF-κB activation induced by TLR signaling. LTßR activation results in a downregulation of proinflammatory cytokine and mediator expression upon TLR restimulation, demonstrating that LTßR signaling is involved in the induction of TLR cross-tolerance. Specific knockdown experiments using TRIM30α-specific small interfering RNA abolished the LTßR-dependent induction of TRIM30α and LTßR-mediated TLR cross-tolerance. Concordantly, LTßR activation on bone marrow-derived macrophages induced cross-tolerance to TLR4 and TLR9 ligands in vitro. Furthermore, we have generated cell type-specific LTßR-deficient mice with ablation of LTßR expression on macrophages/neutrophils (LTßR(flox/flox) × LysM-Cre). In bone marrow-derived macrophages derived from these mice LTßR-induced cross-tolerance to TLR4 and TLR9 ligands was impaired. Additionally, mice with a conditional ablation of LTßR expression on macrophages (LTßR(flox/flox) × LysM-Cre) are resistant to LTßR-induced TLR4 tolerance in vivo. Collectively, our data indicate that LTßR activation on macrophages by T cell-derived lymphotoxin α(1)ß(2) controls proinflammatory responses by activation of a TRIM30α-controlled, counterregulatory signaling pathway to protect against exacerbating inflammatory reactions.


Assuntos
Tolerância Imunológica/imunologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Receptor beta de Linfotoxina/imunologia , Macrófagos/imunologia , Receptor 4 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Linhagem Celular/imunologia , Citocinas/biossíntese , Citocinas/genética , Feminino , Regulação da Expressão Gênica/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/imunologia , Receptor 4 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas
10.
Mol Cell Biol ; 31(21): 4319-34, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896778

RESUMO

Several tumor necrosis factor receptor (TNFR) family members activate both the classical and the alternative NF-κB pathways. However, how a single receptor engages these two distinct pathways is still poorly understood. Using lymphotoxin ß receptor (LTßR) as a prototype, we showed that activation of the alternative, but not the classical, NF-κB pathway relied on internalization of the receptor. Further molecular analyses revealed a specific cytosolic region of LTßR essential for its internalization, TRAF3 recruitment, and p100 processing. Interestingly, we found that dynamin-dependent, but clathrin-independent, internalization of LTßR appeared to be required for the activation of the alternative, but not the classical, NF-κB pathway. In vivo, ligand-induced internalization of LTßR in mesenteric lymph node stromal cells correlated with induction of alternative NF-κB target genes. Thus, our data shed light on LTßR cellular trafficking as a process required for specific biological functions of NF-κB.


Assuntos
Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Receptor beta de Linfotoxina/metabolismo , NF-kappa B/metabolismo , Animais , Sequência de Bases , Transporte Biológico Ativo , Cadeias Pesadas de Clatrina/antagonistas & inibidores , Cadeias Pesadas de Clatrina/genética , Cadeias Pesadas de Clatrina/metabolismo , Citosol/metabolismo , Dinamina II/antagonistas & inibidores , Dinamina II/genética , Dinamina II/metabolismo , Células HEK293 , Células HeLa , Humanos , Receptor beta de Linfotoxina/química , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Subunidade p52 de NF-kappa B/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelB/deficiência , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/metabolismo , Quinase Induzida por NF-kappaB
11.
Eur J Immunol ; 41(3): 780-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21341264

RESUMO

The natural cytotoxicity receptor NKp46 is an activating receptor expressed by several distinct innate lymphoid cell (ILC) subsets, including NK cells, some γδ T cells and intestinal RORγt(+) IL-22(+) cells (NCR22 cells, IL-22-producing NKp46(+) cell). NCR22 cells may play a role in mucosal barrier function through IL-22-mediated production of anti-bacterial peptides from intestinal epithelial cells. Previous studies identified a predominant proportion of NCR22 cells in gut cryptopatches (CP), lymphoid structures that are strategically positioned to collect and integrate signals from luminal microbes; however, whether CP or other lymphoid structures condition NCR22 cell differentiation is not known. Programmed and inducible lymphoid tissue development requires cell-surface-expressed lymphotoxin (LT)α(1) ß(2) heterotrimers (provided by lymphoid tissue inducer (LTi) cells) to signal lymphotoxin-ß receptor (LTR)(+) stromal cells. Here, we analyzed NCR22 cells in LTßR-deficient Ncr1(GFP/+) mice that lack organized secondary lymphoid tissues. We found that NCR22 cells develop in the absence of LTßR, become functionally competent and localize to the lamina propria under steady-state conditions. Following infection of LTßR(-/-) mice with the Gram-negative pathogen Citrobacter rodentium, IL-22 production from NCR22 cells was not affected. These results indicate that organized lymphoid tissue structures are not critical for the generation of an intact and fully functional intestinal NCR22 cell compartment.


Assuntos
Antígenos Ly/metabolismo , Interleucinas/biossíntese , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Células Matadoras Naturais/imunologia , Receptor beta de Linfotoxina/imunologia , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Animais , Citrobacter rodentium , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/patologia , Imunidade Inata , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Células Matadoras Naturais/classificação , Células Matadoras Naturais/metabolismo , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais , Interleucina 22
12.
J Immunol ; 186(3): 1486-94, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21187446

RESUMO

Lymphotoxin ß-receptor (LTßR) and TNF receptor-1 (TNFR1) are important for the development of secondary lymphoid organs during embryonic life. The significance of LTßR and TNFR1 for the formation of lymphoid tissue during adult life is not well understood. Immunohistochemistry, morphometry, flow cytometry, and laser microdissection were used to compare wild-type, LTßR(-/-), TNFR1(-/-) spleens with splenic tissue that has been newly formed 8 wk after avascular implantation into adult mice. During ontogeny, LTßR is sufficient to induce formation of the marginal zone, similar-sized T and B cell zones, and a mixed T/B cell zone that completely surrounded the T cell zone. Strikingly, in adult mice, the formation of splenic compartments required both LTßR and TNFR1 expression, demonstrating that the molecular requirements for lymphoid tissue formation are different during embryonic and adult life. Thus, interfering with the TNFR1 pathway offers the possibility to selectively block the formation of ectopic lymphoid tissue and at the same time to spare secondary lymphoid organs such as spleen and lymph nodes. This opens a new perspective for the treatment of autoimmune and inflammatory diseases.


Assuntos
Envelhecimento/imunologia , Envelhecimento/metabolismo , Feto , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Baço/imunologia , Baço/metabolismo , Animais , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Compartimento Celular/imunologia , Feminino , Feto/anatomia & histologia , Feto/imunologia , Feto/metabolismo , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/imunologia , Baço/citologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
13.
PLoS One ; 5(5): e10585, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20498840

RESUMO

BACKGROUND: Infection with adeno-associated virus (AAV) vector with liver tropism leads to persistent expression of foreign antigens in the mouse liver, with no significant liver inflammation or pathology. This provides a model to investigate antigen persistence in the liver and strategies to modulate host immunity to reduce or clear the foreign antigen expressed from AAV vector in the liver. METHODS/PRINCIPAL FINDINGS: We showed that expressing LIGHT with an adenovirus vector (Ad) in mice with established AAV in the liver led to clearance of the AAV. Ad-LIGHT enhanced CD8 effector T cells in the liver, correlated with liver inflammation. LTbetaR-Ig proteins blocked Ad-LIGHT in clearing AAV. Interestingly, in LTbetaR-null mice, Ad-LIGHT still cleared AAV but caused no significant liver inflammation. CONCLUSIONS/SIGNIFICANCE: Our data suggest that LIGHT interaction with the LTbetaR plays a critical role in liver inflammation but is not required for LIGHT-mediated AAV clearance. These findings will shed light on developing novel immuno-therapeutics in treating people chronically infected with hepato-tropic viruses.


Assuntos
Antígenos Virais/imunologia , Dependovirus/imunologia , Inflamação/virologia , Fígado/patologia , Fígado/virologia , Transdução de Sinais/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Dependovirus/genética , Genoma Viral/genética , Inflamação/patologia , Receptor beta de Linfotoxina/deficiência , Camundongos , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia
14.
Immunity ; 32(3): 403-13, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20226692

RESUMO

Epithelial cells provide the first line of defense against mucosal pathogens; however, their coordination with innate and adaptive immune cells is not well understood. Using mice with conditional gene deficiencies, we found that lymphotoxin (LT) from innate cells expressing transcription factor RORgammat, but not from adaptive T and B cells, was essential for the control of mucosal C. rodentium infection. We demonstrate that the LTbetaR signaling was required for the regulation of the early innate response against infection. Furthermore, we have revealed that LTbetaR signals in gut epithelial cells and hematopoietic-derived cells coordinate to protect the host from infection. We further determined that LTbetaR signaling in intestinal epithelial cells was required for recruitment of neutrophils to the infection site early during infection via production of CXCL1 and CXCL2 chemokines. These results support a model wherein LT from RORgammat(+) cells orchestrates the innate immune response against mucosal microbial infection.


Assuntos
Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Células Epiteliais/imunologia , Imunidade Inata , Mucosa Intestinal/imunologia , Receptor beta de Linfotoxina/imunologia , Transdução de Sinais , Imunidade Adaptativa , Animais , Células da Medula Óssea/imunologia , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Infect Immun ; 76(9): 4241-50, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18625738

RESUMO

Lymph nodes (LNs) are important sentinel organs where antigen-presenting cells interact with T cells to induce adaptive immune responses. In cutaneous infection of mice with Leishmania major, resistance depends on the induction of a T-helper-cell-1 (Th1)-mediated cellular immune response in draining, peripheral LNs. We investigated whether draining, peripheral LNs are absolutely required for resistance against L. major infection. We investigated the course of experimental leishmaniasis in wild-type (wt) mice lacking peripheral LNs (pLNs), which we generated by in utero blockade of membrane-bound lymphotoxin, and in mice lacking pLNs or all LNs due to genetic deletion of lymphotoxin ligands or receptors. wt mice of the resistant C57BL/6 strain without local skin-draining LNs were still able to generate specific T-cell responses, but this yielded Th2 cells. This switch to a Th2 response resulted in severe systemic infection. We also confirmed these results with mice lacking pLNs due to genetic depletion of lymphotoxin-beta. The complete absence of LNs due to a genetic depletion of the lymphotoxin-beta receptor also resulted in a marked deterioration of disease and a Th2 response. Thus, in the absence of pLNs, an L. major-specific Th2 response is induced in the remaining secondary lymphoid organs, such as the spleen and non-skin-draining LNs. This indicates a critical requirement for pLNs to induce protective Th1 immunity and suggests that whether Th1 or Th2 priming to the same antigen occurs depends on the site of the primary antigen recognition.


Assuntos
Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Linfonodos/imunologia , Pele/imunologia , Células Th2/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Linfócitos T CD4-Positivos/imunologia , Suscetibilidade a Doenças/imunologia , Feminino , Pé/parasitologia , Humanos , Interferon gama/metabolismo , Interleucina-4/metabolismo , Linfonodos/anormalidades , Receptor beta de Linfotoxina/deficiência , Linfotoxina-alfa/antagonistas & inibidores , Linfotoxina-beta/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença
16.
J Immunol ; 181(1): 400-7, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18566406

RESUMO

Thymic medullary epithelial cells (mTECs) play a major role in central tolerance induction by expressing tissue-specific Ags (TSAs). The expression of a subset of TSAs in mTECs is under the control of Aire (autoimmune regulator). Humans defective for AIRE develop a syndrome characterized by autoimmune disease in several endocrine glands. Aire has been proposed to be regulated by lymphotoxin beta receptor (Ltbetar) signaling and there is evidence that, additionally, Aire-independent transcripts may be regulated by this pathway. Given the potential clinical importance of Aire regulation in mTECs for the control of autoimmunity, we investigated the relation between Ltbetar signaling and TSA expression by whole genome transcriptome analysis. In this study, we show that the absence of Ltbetar has no effect on the expression of Aire and Aire-dependent TSAs. Also, the lack of Ltbetar signaling does not disturb regulatory T cells or the distribution of dendritic cells in the thymus. However, mTECs in Ltbetar-deficient mice show an aberrant distribution within the thymic medulla with disruption of their three-dimensional architecture. This is predicted to impair the interaction between mTECs and thymocytes as shown by the reduced surface uptake of MHCII by mature thymocytes in Ltbetar-deficient mice. We propose that the physiological medullary architecture ensures negative-selection by supporting lympho-epithelial interaction through a large epithelial cell surface distributed evenly across the medulla.


Assuntos
Células Epiteliais/metabolismo , Receptor beta de Linfotoxina/metabolismo , Transdução de Sinais , Timo/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Animais , Autoimunidade/imunologia , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Proteína AIRE
17.
J Immunol ; 180(4): 2284-93, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250437

RESUMO

Germinal center (GC) responses to T-dependent Ags require effective collaboration between Th cells, activated B cells, and follicular dendritic cells within a highly organized microenvironment. Studies using gene-targeted mice have highlighted nonredundant molecules that are key for initiating and maintaining the GC niche, including the molecules of the ICOS, CD40, and lymphotoxin (LT) pathways. Signaling through ICOS has multiple consequences, including cytokine production, expression of CD40L on Th cells, and differentiation into CXCR5(+) follicular Th cells, all of which are important in the GC reaction. We have therefore taken advantage of ICOS(-/-) mice to dissect which downstream elements are required to initiate the formation of GC. In the context of a T-dependent immune response, we found that GC B cells from ICOS(-/-) mice express lower levels of LTalphabeta compared with wild-type GC B cells in vivo, and stimulation of ICOS on T cells induces LTalphabeta on B cells in vitro. Administration of agonistic anti-LTbeta receptor Ab was unable to restore the GC response in ICOS(-/-) mice, suggesting that additional input from another pathway is required for optimal GC generation. In contrast, treatment with agonistic anti-CD40 Ab in vivo recovered GC networks and restored LTalphabeta expression on GC B cells in ICOS(-/-) mice, and this effect was dependent on LTbeta receptor signaling. Collectively, these data demonstrate that ICOS activation is a prerequisite for the up-regulation of LTalphabeta on GC B cells in vivo and provide a model for cooperation between ICOS, CD40, and LT pathways in the context of the GC response.


Assuntos
Antígenos de Diferenciação de Linfócitos T/fisiologia , Antígenos CD40/fisiologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Receptor beta de Linfotoxina/fisiologia , Transdução de Sinais/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Galinhas , Centro Germinativo/patologia , Proteína Coestimuladora de Linfócitos T Induzíveis , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/biossíntese , Linfotoxina-alfa/genética , Linfotoxina-alfa/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrofenóis/administração & dosagem , Nitrofenóis/imunologia , Fenilacetatos , gama-Globulinas/administração & dosagem , gama-Globulinas/imunologia
18.
J Anat ; 212(2): 114-24, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18194204

RESUMO

The reciprocal influences of thymic lymphocyte and nonlymphocyte populations, i.e. thymic cross-talk, are necessary for the proper maturation of thymocytes and the development/maintenance of thymic stromal microenvironments. Although the molecular influences exerted by thymic stromal cells on maturing thymocytes have been extensively studied, the identity of signalling molecules used by thymocytes to influence the thymic stromal cells is still largely unknown. Our study provides the first ultrastructural evidence that the functional lymphotoxin-beta receptor (LTbetaR) signalling pathway is engaged in the cross-talk between thymocytes and the thymic stromal cell population. We show that LTbetaR signalling is of the utmost significance for the preservation of the subcellular integrity of all thymic epithelial cells. In the absence of LTbetaR there is (1) hypertrophy and activation of cortical thymic epithelial cells, (2) the complete loss of fully differentiated medullary thymic epithelial cells, and (3) the inhibited differentiation of remaining medullary thymic epithelial cells with the appearance of prominent intercellular cysts in the thymic medulla.


Assuntos
Células Epiteliais/diagnóstico por imagem , Receptor beta de Linfotoxina/deficiência , Microscopia Eletrônica/métodos , Timo/ultraestrutura , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ultrassonografia
19.
Mol Immunol ; 45(1): 34-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17590442

RESUMO

The lymphotoxin beta receptor (LTbetaR) signalling pathway is involved in the development of secondary lymphoid organs and the maintenance of organized lymphoid tissues. Additionally, previous studies clearly demonstrated the involvement of the LTbetaR interaction with its ligands in promoting intestinal inflammation. In order to dissect the role of LTbetaR activation in the mouse model of acute DSS-induced colitis we treated mice with a functional inhibitor of LTbetaR activation (LTbetaR:Ig) and compared it to disease in LTbetaR-deficient and LTalphabeta-deficient mice. All these modes of LTbetaR signalling ablation resulted in significant aggravation of the disease and in release of inflammatory cytokines such as TNF, IL-6, and IFNgamma. Finally, using mice with conditionally ablated expression of membrane bound LTbeta on T or B cells, respectively, distinct and opposite contributions of surface LTbeta expressed on T or B cells was found. Thus, activation of LTbetaR by LTalphabeta mainly expressed on T lymphocytes is crucial for the down regulation of the inflammatory response in this experimental model.


Assuntos
Linfócitos B/metabolismo , Colite/metabolismo , Receptor beta de Linfotoxina/antagonistas & inibidores , Linfotoxina-beta/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Doença Aguda , Animais , Linfócitos B/efeitos dos fármacos , Colite/induzido quimicamente , Colite/enzimologia , Colite/patologia , Modelos Animais de Doenças , Feminino , Imunoglobulinas/farmacologia , Inflamação/metabolismo , Ligantes , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Peroxidase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Redução de Peso/efeitos dos fármacos
20.
J Immunol ; 179(9): 5693-700, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17947641

RESUMO

The lymphotoxin pathway is critical for the development and maintenance of peripheral lymphoid organs. Mice with deficiencies in members of this pathway lack lymph nodes and Peyer's patches and have abnormal spleen architecture. These animals also develop autoantibodies to and lymphocytic infiltrates of multiple organs, provoking speculation that the lymphotoxin pathway may play a role in central tolerance induction. Indeed, a series of reports has claimed that lymphotoxin signals control the expression of Aire, a transcriptional regulator that is expressed in medullary epithelial cells of the thymus, mediates ectopic transcription of genes encoding a variety of peripheral tissue Ags, and promotes clonal deletion of self-reactive thymocytes. However, one report argued that lymphotoxin signals regulate the composition and organization of the thymus, particularly of the medullary epithelial compartment. Herein, we resolve this controversy in favor of the latter view. The expression and function of Aire were unaffected in medullary epithelial cells of mice lacking either lymphotoxin beta receptor or the lymphotoxin alpha-chain, and there was minimal overlap between the sets of genes controlled by Aire and lymphotoxin. Instead, both knockout lines showed abnormal medullary epithelial cell organization, and the line lacking the beta receptor had significantly fewer medullary epithelial cells. In short, the lymphotoxin pathway drives the developmental rather than selectional properties of thymic stromal cells.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Linfotoxina-alfa/imunologia , Transdução de Sinais/imunologia , Timo/citologia , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células , Células Epiteliais/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Receptor beta de Linfotoxina/deficiência , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Camundongos , Camundongos Knockout , Timo/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Proteína AIRE
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...