Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 11: 625630, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552090

RESUMO

Ectopic lymphoid tissues (eLTs) characterized by B cell aggregation contribute to the local immunoglobulin production in nasal polyps (NPs). B cell-activating factor (BAFF) is vital for B cell survival, proliferation, and maturation. The purpose of this study is to investigate whether BAFF is involved in the B cell survival and eLT formation in NPs. The mRNA and protein levels of BAFF in NP tissues with and without eLTs were detected by PCR and ELISA assay, respectively. The cellular sources of BAFF and active caspase-3-positive B cells in NPs were studied by immunofluorescence staining. B cells purified from NP tissues were stimulated with BAFF and were analyzed by flow cytometry. Stromal cells purified from NP tissues were stimulated with lymphotoxin (LT) α1ß2, and BAFF levels in culture supernatants were analyzed by ELISA. Compared with those in control tissues and NPs without eLTs, the BAFF levels were elevated in NPs with eLTs. Abundant BAFF-positive cells and few active caspase-3-positive apoptotic B cells were found in NPs with eLTs, in contrast to those in NPs without eLTs. There was a negative correlation between the numbers of BAFF-positive cells and frequencies of apoptotic B cells in total B cells in NP tissues. BAFF protected nasal polyp B cells from apoptosis in vitro. Stromal cells were an important cellular source of BAFF in NPs with eLTs. LTα1ß2 induced BAFF production from nasal stromal cells in vitro. We propose that BAFF contribute to eLT formation in NPs by promoting B cell survival.


Assuntos
Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Pólipos Nasais/imunologia , Estruturas Linfoides Terciárias/imunologia , Apoptose/imunologia , Linfócitos B/patologia , Caspase 3/imunologia , Sobrevivência Celular , Feminino , Humanos , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Masculino , Pólipos Nasais/patologia , Estruturas Linfoides Terciárias/patologia
2.
Cell Mol Immunol ; 15(7): 697-709, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28579615

RESUMO

Splenomegaly is a well-known phenomenon typically associated with inflammation. However, the underlying cause of this phenotype has not been well characterized. Furthermore, the splenomegaly phenotype seen in lymphotoxin (LT) signaling-deficient mice is characterized by increased numbers of splenocytes and splenic neutrophils. Splenomegaly, as well as the related phenotype of increased lymphocyte counts in non-lymphoid tissues, is thought to result from the absence of secondary lymphoid tissues in LT-deficient mice. We now present evidence that mice deficient in LTα1ß2 or LTßR develop splenomegaly and increased numbers of lymphocytes in non-lymphoid tissues in a microbiota-dependent manner. Antibiotic administration to LTα1ß2- or LTßR-deficient mice reduces splenomegaly. Furthermore, re-derived germ-free Ltbr-/- mice do not exhibit splenomegaly or increased inflammation in non-lymphoid tissues compared to specific pathogen-free Ltbr-/- mice. By using various LTß- and LTßR-conditional knockout mice, we demonstrate that retinoic acid-related orphan receptor γT-positive type 3 innate lymphoid cells provide the required active LT signaling to prevent the development of splenomegaly. Thus, this study demonstrates the importance of LT-mediated immune responses for the prevention of splenomegaly and systemic inflammation induced by microbiota.


Assuntos
Imunidade Inata , Linfócitos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Microbiota/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Animais , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Linfócitos/patologia , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T gama-delta/genética
3.
J Immunol ; 197(5): 1957-67, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27474071

RESUMO

Lymphangiogenesis associated with tertiary lymphoid structure (TLS) has been reported in numerous studies. However, the kinetics and dynamic changes occurring to the lymphatic vascular network during TLS development have not been studied. Using a viral-induced, resolving model of TLS formation in the salivary glands of adult mice we demonstrate that the expansion of the lymphatic vascular network is tightly regulated. Lymphatic vessel expansion occurs in two distinct phases. The first wave of expansion is dependent on IL-7. The second phase, responsible for leukocyte exit from the glands, is regulated by lymphotoxin (LT)ßR signaling. These findings, while highlighting the tight regulation of the lymphatic response to inflammation, suggest that targeting the LTα1ß2/LTßR pathway in TLS-associated pathologies might impair a natural proresolving mechanism for lymphocyte exit from the tissues and account for the failure of therapeutic strategies that target these molecules in diseases such as rheumatoid arthritis.


Assuntos
Interleucina-7/metabolismo , Linfangiogênese , Vasos Linfáticos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Estruturas Linfoides Terciárias/imunologia , Animais , Regulação da Expressão Gênica , Inflamação , Interleucina-7/genética , Interleucina-7/imunologia , Vasos Linfáticos/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Camundongos , Glândulas Salivares/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Estruturas Linfoides Terciárias/patologia
4.
Nat Commun ; 7: 12021, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27323847

RESUMO

Regulatory T cells (Tregs) are essential to suppress unwanted immunity or inflammation. After islet allo-transplant Tregs must migrate from blood to allograft, then via afferent lymphatics to draining LN to protect allografts. Here we show that Tregs but not non-Treg T cells use lymphotoxin (LT) during migration from allograft to draining LN, and that LT deficiency or blockade prevents normal migration and allograft protection. Treg LTαß rapidly modulates cytoskeletal and membrane structure of lymphatic endothelial cells; dependent on VCAM-1 and non-canonical NFκB signalling via LTßR. These results demonstrate a form of T-cell migration used only by Treg in tissues that serves an important role in their suppressive function and is a unique therapeutic focus for modulating suppression.


Assuntos
Diabetes Mellitus Experimental/terapia , Rejeição de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Linfócitos T Reguladores/imunologia , Migração Transendotelial e Transepitelial/imunologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/mortalidade , Células Endoteliais/citologia , Células Endoteliais/imunologia , Regulação da Expressão Gênica , Rejeição de Enxerto/genética , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/genética , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/cirurgia , Linfonodos/citologia , Linfonodos/imunologia , Vasos Linfáticos/citologia , Vasos Linfáticos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais , Análise de Sobrevida , Linfócitos T Reguladores/citologia , Transplante Homólogo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
5.
J Immunol ; 194(6): 2616-23, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25662995

RESUMO

Secondary lymphoid organs (SLO) provide the structural framework for coconcentration of Ag and Ag-specific lymphocytes required for an efficient adaptive immune system. The spleen is the primordial SLO, and evolved concurrently with Ig/TCR:pMHC-based adaptive immunity. The earliest cellular/histological event in the ontogeny of the spleen's lymphoid architecture, the white pulp (WP), is the accumulation of B cells around splenic vasculature, an evolutionarily conserved feature since the spleen's emergence in early jawed vertebrates such as sharks. In mammals, B cells are indispensable for both formation and maintenance of SLO microarchitecture; their expression of lymphotoxin α1ß2 (LTα1ß2) is required for the LTα1ß2:CXCL13 positive feedback loop without which SLO cannot properly form. Despite the spleen's central role in the evolution of adaptive immunity, neither the initiating event nor the B cell subset necessary for WP formation has been identified. We therefore sought to identify both in mouse. We detected CXCL13 protein in late embryonic splenic vasculature, and its expression was TNF-α and RAG-2 independent. A substantial influx of CXCR5(+) transitional B cells into the spleen occurred 18 h before birth. However, these late embryonic B cells were unresponsive to CXCL13 (although responsive to CXCL12) and phenotypically indistinguishable from blood-derived B cells. Only after birth did B cells acquire CXCL13 responsiveness, accumulate around splenic vasculature, and establish the uniquely splenic B cell compartment, enriched for CXCL13-responsive late transitional cells. Thus, CXCL13 is the initiating component of the CXCL13:LTα1ß2 positive feedback loop required for WP ontogeny, and CXCL13-responsive late transitional B cells are the initiating subset.


Assuntos
Linfócitos B/imunologia , Quimiocina CXCL13/imunologia , Receptores CXCR5/imunologia , Baço/imunologia , Animais , Animais Recém-Nascidos , Linfócitos B/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Retroalimentação Fisiológica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/irrigação sanguínea , Baço/embriologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
6.
J Exp Med ; 211(7): 1421-31, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-24913234

RESUMO

Natural killer (NK) cell development relies on signals provided from the bone marrow (BM) microenvironment. It is thought that lymphotoxin (LT) α1ß2 expressed by the NK cell lineage interacts with BM stromal cells to promote NK cell development. However, we now report that a small number of RORγt(+) innate lymphoid cells (ILCs), and not CD3(-)NK1.1(+) cells, express LT to drive NK development. Similar to LT(-/-) or RORγt(-/-) mice, the mice conditionally lacking LTα1ß2 on RORγt(+) ILCs experience a developmental arrest at the immature NK stages, between stages of NK development to the mature NK cell stage. This developmental block results in a functional deficiency in the clearance of NK-sensitive tumor cells. Reconstitution of Thy1(+) ILCs from BM or purified RORγt(+) ILCs from lamina propria lymphocytes into LT-deficient RORγt(+) BM cultures rescues NK cell development. These data highlight a previously undiscovered role of RORγt(+) ILCs for NK cell development and define LT from ILCs as an essential molecule for the stromal microenvironment supporting NK cell development.


Assuntos
Imunidade Inata/fisiologia , Células Matadoras Naturais/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Linfotoxina-alfa/imunologia , Animais , Células Matadoras Naturais/citologia , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Linfotoxina-alfa/genética , Camundongos , Camundongos Knockout , Mucosa/citologia , Mucosa/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Antígenos Thy-1/genética , Antígenos Thy-1/imunologia
7.
J Biotechnol ; 172: 73-6, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24384233

RESUMO

The tumor necrosis factor superfamily (TNFSF) consists of more than 20 members that can modulate cellular and immunological functions, including cell survival and the stimulation of an inflammatory response. Many TNF superfamily members display potent anticancer activity when used as recombinant proteins in vitro and in vivo. While TNF, TRAIL and FasL have already been used as payloads in antibody-based pharmacodelivery strategies, most TNF superfamily members have not yet been investigated as antibody payloads. Here, we report the cloning, production and characterization of eight novel antibody fusion proteins based on CD40L, FasL, TRAIL, LiGHT, VEGI, lymphotoxin alpha, lymphotoxin beta and lymphotoxin alpha1/beta2. The monoclonal antibody F8 was chosen as fusion partner of proven tumor targeting performance, which recognizes the alternatively-spliced EDA domain of fibronectin, a marker of angiogenesis. A quantitative biodistribution analysis performed with radioiodinated protein preparations in tumor-bearing mice revealed that TRAIL and lymphotoxin alpha1/beta2 were able to selectively accumulate at the tumor site, while all other members of the TNF superfamily abrogated the selective tumor targeting performance of the parental antibody or accumulated also in healthy tissues. The study indicates that even cytokines, which are closely related in terms of structure and function, may have a substantially different impact on the biodistribution and functional properties of the corresponding fusions with disease-homing antibodies.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais Humanizados , Ligante de CD40/genética , Ligante de CD40/imunologia , Células CHO , Linhagem Celular , Cricetulus , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Feminino , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Camundongos , Camundongos da Linhagem 129 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/metabolismo
8.
J Immunol ; 191(9): 4611-8, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24078690

RESUMO

Innate lymphoid cells encompass a diverse array of lymphocyte subsets with unique phenotype that initiate inflammation and provide host defenses in specific microenvironments. In this study, we identify a rare human CD4(+)CD3(-) innate-like lymphoid population with high TNF expression that is enriched in blood from patients with rheumatoid arthritis. These CD4(+)CD3(-) cells belong to the T cell lineage, but the lack of AgR at the cell surface renders them nonresponsive to TCR-directed stimuli. By developing a culture system that sustains survival, we show that CD4(+)CD3(-) innate-like T cells display IL-7-dependent induction of surface lymphotoxin-αß, demonstrating their potential to modify tissue microenvironments. Furthermore, expression of CCR6 on the CD4(+)CD3(-) population defines a CD127(high) subset that is highly responsive to IL-7. This CD4(+)CD3(-) population is enriched in the peripheral blood from rheumatoid arthritis patients, suggesting a link to their involvement in chronic inflammatory disease.


Assuntos
Artrite Reumatoide/metabolismo , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Fatores de Necrose Tumoral/metabolismo , Adulto , Idoso , Animais , Artrite Reumatoide/imunologia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Feminino , Células HEK293 , Proteínas de Homeodomínio/genética , Humanos , Inflamação , Interleucina-7/metabolismo , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Receptores CCR6/metabolismo
9.
Eur J Immunol ; 43(2): 348-59, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23112125

RESUMO

CD19-deficient mice were used as a model to study follicular dendritic cell (FDC) activation because these mice have normal numbers of FDC-containing primary follicles, but lack the ability to activate FDCs or form GCs. It was hypothesized that CD19 expression is necessary for B-cell activation and upregulation of membrane lymphotoxin (mLT) expression, which promotes FDC activation. Using VCAM-1 and FcγRII/III as FDC activation markers, it was determined that the adoptive transfer of CD19(+) wild-type B cells into CD19-deficient hosts rescued GC formation and FDC activation, demonstrating that CD19 expression on B cells is required for FDC activation. In contrast, CD19(+) donor B cells lacking mLT were unable to induce VCAM-1 expression on FDCs, furthermore FcγRII/III upregulation was impaired in FDCs stimulated with mLT-deficient B cells. VCAM-1 expression on FDCs, but not FcγRII/III, was rescued when CD19-deficient B cells expressing transgenic mLT were cotransferred into recipient mice with CD19(+) , mLT-deficient B cells, suggesting that FDC activation requires the CD19-dependent upregulation of mLT on activated B cells. Collectively, these data demonstrate that activated B cells are responsible for the initiation of FDC activation resulting in a microenvironment supportive of GC development and maintenance.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/biossíntese , Animais , Antígenos CD19/biossíntese , Antígenos CD19/genética , Antígenos CD19/imunologia , Ativação Linfocitária , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de IgG/biossíntese , Receptores de IgG/genética , Receptores de IgG/imunologia , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
10.
Cytokine Growth Factor Rev ; 22(5-6): 301-10, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22152226

RESUMO

This review focuses on the biological functions and signalling pathways activated by Lymphotoxin α (LTα)/Lymphotoxin ß (LTß) and their receptor LTßR. Genetic mouse models shed light on crucial roles for LT/LTßR to build and to maintain the architecture of lymphoid organs and to ensure an adapted immune response against invading pathogens. However, chronic inflammation, autoimmunity, cell death or cancer development are disorders that occur when the LT/LTßR system is twisted. Biological inhibitors, such as antagonist antibodies or decoy receptors, have been developed and used in clinical trials for diseases associated to the LT/LTßR system. Recent progress in the understanding of cellular trafficking and NF-κB signalling pathways downstream of LTα/LTß may bring new opportunities to develop therapeutics that target the pathological functions of these cytokines.


Assuntos
Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Linfotoxina-alfa/imunologia , Linfotoxina-beta/imunologia , Animais , Morte Celular , Expressão Gênica , Humanos , Heterotrímero de Linfotoxina alfa1 e beta2/química , Receptor beta de Linfotoxina/química , Linfotoxina-alfa/química , Linfotoxina-alfa/genética , Linfotoxina-beta/química , Linfotoxina-beta/genética , NF-kappa B/imunologia , Estrutura Terciária de Proteína , Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Transdução de Sinais
11.
Immunol Rev ; 244(1): 75-84, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22017432

RESUMO

Lymph nodes (LNs) maintain active homeostasis at steady state. However, in response to changes in the local environment, such as local infection, cancer, vaccination, and autoimmune disease, dramatic remodeling of LN occurs. This remodeling includes changes in size, lymph and blood flow, immune cell trafficking and cellularity, lymphatic and blood vessel growth and activation, as well as microarchitecture. Therefore, inflammatory conditions often lead to enlarged nodes; after local inflammation resolves, LNs actively regress in size and return to steady state. Remodeling of lymphatic vessels (LVs) and blood vessels (BVs) during both the expansion and regression phases are key steps in controlling LN size as well as function. The cells, membrane-associated molecules, and soluble cytokines that are essential for LV and BV homeostasis as well as dynamic changes in the expansion and regression phases have not been well defined. Understanding the underlying cellular and molecular mechanisms behind LN remodeling would help us to better control undesired immune responses (e.g. inflammation and autoimmune diseases) or promote desired responses (e.g. antitumor immunity and vaccination). In this review, we focus on how the closely related tumor necrosis factor (TNF) members: LIGHT (TNFSF14), lymphotoxin-αß, and TNF-α contribute to the remodeling of LNs at various stages of inflammation.


Assuntos
Vasos Sanguíneos/imunologia , Imunidade Inata , Linfonodos/imunologia , Vasos Linfáticos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Transdução de Sinais/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Movimento Celular , Microambiente Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Expressão Gênica , Homeostase/imunologia , Humanos , Inflamação , Linfonodos/irrigação sanguínea , Linfonodos/citologia , Linfonodos/metabolismo , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Camundongos , Camundongos Knockout , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
12.
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
13.
Immunity ; 30(3): 408-20, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19303389

RESUMO

The lymphotoxin LTalpha(1)beta(2) supports the development and maintenance of several aspects of spleen structure, but its significance for marginal sinus (MS) vascular organization is unclear. We showed here that, in early postnatal lymphotoxin-deficient mice, the developing Flk-1+ white pulp vessels failed to organize or upregulate MAdCAM-1, leading to altered spatial rearrangement of both the white pulp endothelial cells and the smooth muscle actin-expressing cells. In vitro, MAdCAM-1 directed the reorganization of LTbeta receptor+ endothelial cells grown on Matrigel. LTalpha(1)beta(2) also regulated the maintenance of both MAdCAM-1 expression and mature MS structure in adult mice, contributing importantly to normal trafficking of CD11b+ cells in response to bacterial antigens. Together, our studies demonstrate that LTalpha(1)beta(2) and LTbeta receptor signals control proper development and maintenance of the mature MS structure and implicate MAdCAM-1 in the structuring of the MS endothelial cells that is important for the movement of immune cells within the spleen.


Assuntos
Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Baço/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos CD/metabolismo , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Células Cultivadas , Células Endoteliais/metabolismo , Ensaio de Imunoadsorção Enzimática , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucoproteínas , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Baço/citologia
14.
Immunol Rev ; 223: 186-201, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18613837

RESUMO

SUMMARY: Cytokines mediate key communication pathways essential for regulation of immune responses. Full activation of antigen-responding lymphocytes requires cooperating signals from the tumor necrosis factor (TNF)-related cytokines and their specific receptors. LIGHT, a lymphotoxin-beta (LTbeta)-related TNF family member, modulates T-cell activation through two receptors, the herpesvirus entry mediator (HVEM) and indirectly through the LT-beta receptor. An unexpected finding revealed a non-canonical binding site on HVEM for the immunoglobulin superfamily member, B and T lymphocyte attenuator (BTLA), and an inhibitory signaling protein suppressing T-cell activation. Thus, HVEM can act as a molecular switch between proinflammatory and inhibitory signaling. The non-canonical HVEM-BTLA pathway also acts to counter LTbetaR signaling that promotes the proliferation of antigen-presenting dendritic cells (DCs) within lymphoid tissue microenvironments. These results indicate LTbeta receptor and HVEM-BTLA pathways form an integrated signaling circuit. Targeting these cytokine pathways with specific antagonists (antibody or decoy receptor) can alter lymphocyte differentiation and activation. Alternately, agonists directed at their cell surface receptors can restore homeostasis and potentially reset immune and inflammatory processes, which may be useful in treating autoimmune and infectious diseases and cancer.


Assuntos
Doenças Autoimunes/imunologia , Ativação Linfocitária/efeitos dos fármacos , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/farmacologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Doenças Autoimunes/terapia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Retroalimentação Fisiológica , Humanos , Receptor beta de Linfotoxina/imunologia , Camundongos , Receptor Cross-Talk , Receptores Imunológicos , Membro 14 de Receptores do Fator de Necrose Tumoral , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
15.
Immunol Rev ; 223: 202-20, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18613838

RESUMO

SUMMARY: The lymphotoxin (LT) system is part of the tumor necrosis factor family and is required for lymph node development. It has provided a wonderful tool for the dissection of processes critical not only for lymphoid organ development but also the maintenance of the adult immune architecture and the formation of ectopic organized lymphoid tissues in chronically inflamed sites. A soluble lymphotoxin-beta receptor-immunoglobulin (LTbetaR-Ig) fusion protein can block this pathway and is currently being tested in the treatment of autoimmune disease. This review focuses on the immunological consequences of combined LT and LIGHT inhibition with LTbetaR-Ig administration as distinct from the developmental biology.


Assuntos
Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Transdução de Sinais/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Quimiocinas/imunologia , Ensaios Clínicos como Assunto , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/antagonistas & inibidores , Modelos Imunológicos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/imunologia , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia
16.
J Immunol ; 178(4): 2272-7, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277132

RESUMO

Activation of the noncanonical pathway through the interaction of lymphotoxin (LT)-alpha(1)beta(2) and LT-betaR is essential for the development of secondary lymphoid organs including lymph nodes (LN) and Peyer's patches (PP). Although TNFR-associated factor (TRAF) 2 and TRAF5 were identified as signal transducers for the LT-betaR, roles for TRAF2 and TRAF5 in the development of secondary lymphoid organs remain obscure. In this study, we show that PP but not mesenteric LN development is severely impaired in traf2(-/-) and traf2(-/-)traf5(-/-) mice. Development of VCAM-1(+) and ICAM-1(+) mesenchymal cells and expression of CXCL13, a crucial chemokine for the development of PP, are severely impaired in PP anlagen in the intestines of traf2(-/-) mice. Surprisingly, TNF-alpha stimulation potently up-regulates cxcl13 mRNA expression in wild-type murine embryonic fibroblasts, which is impaired in traf2(-/-) and relA(-/-) murine embryonic fibroblasts. Moreover, RelA is recruited to the promoter of cxcl13 gene upon TNF-alpha stimulation and PP development is impaired in TNFR type 1 (tnfr1)(-/-) mice. These results underscore a crucial role for the TNFR1-TRAF2-RelA-dependent canonical pathway in the development of PP through up-regulation of cxcl13 mRNA.


Assuntos
Quimiocinas CXC/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Nódulos Linfáticos Agregados/embriologia , Fator 2 Associado a Receptor de TNF/imunologia , Fator 5 Associado a Receptor de TNF/imunologia , Fator de Transcrição RelA/imunologia , Animais , Quimiocina CXCL13 , Quimiocinas CXC/biossíntese , Fibroblastos/citologia , Fibroblastos/imunologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/imunologia , Intestino Delgado/citologia , Intestino Delgado/embriologia , Intestino Delgado/imunologia , Linfonodos/embriologia , Linfonodos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Mesoderma/citologia , Mesoderma/imunologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Fator 2 Associado a Receptor de TNF/deficiência , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 5 Associado a Receptor de TNF/deficiência , Fator 5 Associado a Receptor de TNF/metabolismo , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA