Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 286
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 13(10): 947-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22922363

RESUMO

Microbiota are essential for weight gain in mouse models of diet-induced obesity (DIO), but the pathways that cause the microbiota to induce weight gain are unknown. We report that mice deficient in lymphotoxin, a key molecule in gut immunity, were resistant to DIO. Ltbr(-/-) mice had different microbial community composition compared to their heterozygous littermates, including an overgrowth of segmented filamentous bacteria (SFB). Furthermore, cecal transplantation conferred leanness to germ-free recipients. Housing Ltbr(-/-) mice with their obese siblings rescued weight gain in Ltbr(-/-) mice, demonstrating the communicability of the obese phenotype. Ltbr(-/-) mice lacked interleukin 23 (IL-23) and IL-22, which can regulate SFB. Mice deficient in these pathways also resisted DIO, demonstrating that intact mucosal immunity guides diet-induced changes to the microbiota to enable obesity.


Assuntos
Imunidade nas Mucosas , Receptor beta de Linfotoxina/fisiologia , Linfotoxina-alfa/fisiologia , Obesidade , Animais , Bactérias/crescimento & desenvolvimento , Bactérias/imunologia , Ceco/microbiologia , Ceco/transplante , Dieta , Metabolismo Energético , Vida Livre de Germes , Interleucina-23/deficiência , Interleucina-23/fisiologia , Interleucinas/deficiência , Interleucinas/fisiologia , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/deficiência , Linfotoxina-alfa/genética , Metagenoma , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo , Aumento de Peso/imunologia , Interleucina 22
2.
Nat Immunol ; 12(10): 941-8, 2011 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-21874025

RESUMO

Colonic patches (CLPs) and isolated lymphoid follicles (ILFs) are two main lymphoid structures in the colon. Lymphoid tissue-inducer cells (LTi cells) are indispensable for the development of ILFs. LTi cells also produce interleukin 17 (IL-17) and IL-22, signature cytokines secreted by IL-17-producing helper T cells. Here we report that IL-22 acted downstream of the lymphotoxin pathway and regulated the organization and maintenance of mature CLPs and ILFs in the colon during infection with Citrobacter rodentium. Lymphotoxin (LTα(1)ß(2)) regulated the production of IL-22 during infection with C. rodentium, but the lymphotoxin-like protein LIGHT did not. IL-22 signaling was sufficient to restore the organization of CLPs and ILFs and host defense against infection with C. rodentium in mice lacking lymphotoxin signals, which suggests that IL-22 connects the lymphotoxin pathway to mucosal epithelial defense mechanisms.


Assuntos
Citrobacter rodentium , Colo/imunologia , Infecções por Enterobacteriaceae/imunologia , Interleucinas/fisiologia , Tecido Linfoide/fisiologia , Linfotoxina-alfa/fisiologia , Animais , Colo/microbiologia , Interleucina-23/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Interleucina 22
4.
J Immunol ; 193(3): 1194-203, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24951816

RESUMO

Spleen is a tissue with regenerative capacity, which allows autotransplantation of human spleen fragments to counteract the effects of splenectomy. We now reveal in a murine model that transplant of neonatal spleen capsule alone leads to the regeneration of full spleen tissue. This finding indicates that graft-derived spleen stromal cells, but not lymphocytes, are essential components of tissue neogenesis, a finding verified by transplant and regeneration of Rag1KO spleen capsules. We further demonstrate that lymphotoxin and lymphoid tissue inducer cells participate in two key elements of spleen neogenesis, bulk tissue regeneration and white pulp organization, identifying a lymphotoxin-dependent pathway for neonatal spleen regeneration that contrasts with previously defined lymphotoxin-independent embryonic spleen organogenesis.


Assuntos
Linfotoxina-alfa/fisiologia , Regeneração/imunologia , Baço/imunologia , Baço/transplante , Animais , Animais Recém-Nascidos , Humanos , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Transdução de Sinais/imunologia , Baço/citologia , Células Estromais/citologia , Células Estromais/imunologia
5.
Blood ; 116(12): 2173-82, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20566898

RESUMO

Lymphotoxin-α (LTα), lymphotoxin-ß (LTß), and tumor necrosis factor-α (TNFα) are inflammatory mediators that play crucial roles in lymphoid organ development. We demonstrate here that LTα also contributes to the function of lymphatic vessels and to lymphangiogenesis during inflammation. LTα(-/-) mice exhibited reduced lymph flow velocities and increased interstitial fluid pressure. Airways of LTß(-/-) mice infected with Mycoplasma pulmonis had significantly more lymphangiogenesis than wild type (WT) or LTα(-/-) mice, as did the skin draining immunization sites of LTß(-/-) mice. Macrophages, B cells, and T cells, known sources of LT and TNFα, were apparent in the skin surrounding the immunization sites as were LTα, LTß, and TNFα mRNAs. Ectopic expression of LTα led to the development of LYVE-1 and Prox1-positive lymphatic vessels within tertiary lymphoid organs (TLOs). Quantification of pancreatic lymphatic vessel density in RIPLTαLTß(-/-) and WT mice revealed that LTα was sufficient for inducing lymphangiogenesis and that LTß was not required for this process. Kidneys of inducible LTα transgenic mice developed lymphatic vessels before the appearance of obvious TLOs. These data indicate that LTα plays a significant role in lymphatic vessel function and in inflammation-associated lymphangiogenesis.


Assuntos
Linfangiogênese , Linfotoxina-alfa/fisiologia , Animais , Imunização , Inflamação , Rim/imunologia , Vasos Linfáticos , Linfotoxina-alfa/deficiência , Linfotoxina-alfa/genética , Linfotoxina-beta , Camundongos , Camundongos Knockout , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis , Pele
6.
Front Immunol ; 13: 1009306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36189308

RESUMO

Tumor-associated tertiary lymphoid structures are ectopic lymphoid aggregates that have considerable morphological, cellular, and molecular similarity to secondary lymphoid organs, particularly lymph nodes. Tumor vessels expressing peripheral node addressin (PNAd) are hallmark features of these structures. Previous work from our laboratory demonstrated that PNAd is displayed on intratumoral vasculature of murine tumors, and its expression is controlled by the engagement of lymphotoxin-α3, secreted by effector CD8 T cells, with tumor necrosis factor receptors (TNFR) on tumor endothelial cells (TEC). The goals of the present work were: 1) to identify differences in expression of genes encoding the scaffolding proteins and glycosyl transferases associated with PNAd biosynthesis in TEC and lymph node blood endothelial cells (LN BEC); and 2) to determine which of these PNAd associated components are regulated by TNFR signaling. We found that the same genes encoding scaffolding proteins and glycosyl transferases were upregulated in PNAd+ LN BEC and PNAd+ TEC relative to their PNAdneg counterparts. The lower level of PNAd expression on TEC vs LN BEC was associated with relatively lower expression of these genes, particularly the carbohydrate sulfotransferase Chst4. Loss of PNAd on TEC in the absence of TNFR signaling was associated with lack of upregulation of these same genes. A small subset of PNAd+ TEC remaining in the absence of TNFR signaling showed normal upregulation of a subset of these genes, but reduced upregulation of genes encoding the scaffolding proteins podocalyxin and nepmucin, and carbohydrate sulfotransferase Chst2. Lastly, we found that checkpoint immunotherapy augmented both the fraction of TEC expressing PNAd and their surface level of this ligand. This work points to strong similarities in the regulation of PNAd expression on TEC by TNFR signaling and on LN BEC by lymphotoxin-ß receptor signaling, and provides a platform for the development of novel strategies that manipulate PNAd expression on tumor vasculature as an element of cancer immunotherapy.


Assuntos
Células Endoteliais , Neoplasias , Animais , Ligantes , Linfotoxina-alfa/fisiologia , Linfotoxina-beta , Camundongos , Neoplasias/genética , Receptores do Fator de Necrose Tumoral , Sulfotransferases
7.
Hepatology ; 52(6): 2158-66, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21064033

RESUMO

The matrix metalloproteinase (MMP) family of proteins degrades extracellular matrix (ECM) components as well as processes cytokines and growth factors. MMPs are involved in regulating ECM homeostasis in both normal physiology and disease pathophysiology. Here we report the critical roles of mmp23b in normal zebrafish liver development. Mmp23b was initially identified as a gene linked to the genomic locus of an enhancer trap transgenic zebrafish line in which green fluorescent protein (GFP) expression was restricted to the developing liver. Follow-up analysis of mmp23b messenger RNA (mRNA) expression confirmed its liver-specific expression pattern. Morpholino knockdown of mmp23b resulted in defective hepatocyte proliferation, causing a reduction in liver size while maintaining relatively normal pancreas and gut development. Genetically, we showed that mmp23b functions through the tumor necrosis factor (TNF) signaling pathway. Antisense knockdown of tnfa or tnfb in zebrafish caused similar reductions of liver size, whereas overexpression of tnfa or tnfb rescued liver defects in mmp23b morphants but not vice versa. Biochemically, MMP23B, the human ortholog of Mmp23b, directly interacts with TNF and mediates its release from the cell membrane in a cell culture system. Because mmp23b/MMP23B is highly conserved, our findings in zebrafish warrant further investigation of its role in regulating liver development in mammals.


Assuntos
Hepatócitos/citologia , Fígado/crescimento & desenvolvimento , Metaloproteinases da Matriz/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Proliferação de Células , Técnicas de Silenciamento de Genes , Hepatócitos/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Linfotoxina-alfa/genética , Linfotoxina-alfa/fisiologia , Metaloproteinases da Matriz/farmacologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia , Peixe-Zebra
8.
Ann Intern Med ; 152(3): 159-66, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20124232

RESUMO

Abnormal and exaggerated deposition of extracellular matrix is the hallmark of many fibrotic diseases, including systemic sclerosis and pulmonary, liver, and kidney fibrosis. The spectrum of affected organs, the usually progressive nature of the fibrotic process, the large number of affected persons, and the absence of effective treatment pose an enormous challenge when treating fibrotic diseases. Delineation of the central role of transforming growth factor-beta (TGF-beta) and identification of the specific cellular receptors, kinases, and other mediators involved in the fibrotic process have provided a sound basis for development of effective therapies. The inhibition of signaling pathways activated by TGF-beta represents a novel therapeutic approach for the fibrotic disorders. One of these TGF-beta pathways results in the activation of the nonreceptor tyrosine kinase cellular Abelson (c-Abl), and c-Abl inhibitors, including imatinib mesylate, diminishing the fibrogenic effects of TGF-beta. Thus, recently acquired basic knowledge about the pathogenesis of the fibrotic process has enabled the development of novel therapeutic agents capable of modifying the deleterious effects of the fibrotic diseases.


Assuntos
Fibrose/tratamento farmacológico , Fibrose/etiologia , Fibrose/patologia , Humanos , Linfotoxina-alfa/fisiologia
9.
J Exp Med ; 185(12): 2177-82, 1997 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-9182689

RESUMO

Immunization of mice with myelin components results in experimental autoimmune encephalomyelitis (EAE), which is mediated by myelin-specific CD4(+) T cells and anti-myelin antibodies. Tumor necrosis factor alpha (TNF-alpha) and lymphotoxin alpha (LT-alpha) are thought to be involved in the events leading to inflammatory demyelination in the central nervous system. To ascertain this hypothesis 129 x C57BL/6 mice with an inactivation of the tnf and lta genes (129 x C57BL/6(-/-)) and SJL/J mice derived from backcrosses of the above mentioned mutant mice (SJL-/-) were immunized with mouse spinal cord homogenate (MSCH) or proteolipid protein. Both 129 x C57BL/6(-/-) mice and SJL-/- mice developed EAE. In SJL-/- mice immunized with MSCH, a very severe form of EAE with weight loss, paralysis of all four limbs, and lethal outcome was observed. The histologic hallmark was an intense perivascular and parenchymal infiltration with predominantly CD4(+) T cells and some CD8(+) T cells associated with demyelination in both brain and spinal cord. These results indicate that TNF-alpha and LT-alpha are not essential for the development of EAE.


Assuntos
Encefalomielite Autoimune Experimental/etiologia , Linfotoxina-alfa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Doença Aguda , Animais , Autoimunidade , Feminino , Camundongos , Camundongos Endogâmicos C57BL
10.
J Exp Med ; 197(9): 1153-63, 2003 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-12732657

RESUMO

Lymph node (LN) function depends on T and B cell compartmentalization, antigen presenting cells, and high endothelial venules (HEVs) expressing mucosal addressin cell adhesion molecule (MAdCAM-1) and peripheral node addressin (PNAd), ligands for naive cell entrance into LNs. Luminal PNAd expression requires a HEV-restricted sulfotransferase (HEC-6ST). To investigate LT alpha beta's activities in lymphoid organogenesis, mice simultaneously expressing LT alpha and LT beta under rat insulin promoter II (RIP) control were compared with RIPLT alpha mice in a model of lymphoid neogenesis and with LT beta-/- mice. RIPLT alpha beta pancreata exhibited massive intra-islet mononuclear infiltrates that differed from the more sparse peri-islet cell accumulations in RIPLT alpha pancreata: separation into T and B cell areas was more distinct with prominent FDC networks, expression of lymphoid chemokines (CCL21, CCL19, and CXCL13) was more intense, and L-selectin+ cells were more frequent. In contrast to the predominant abluminal PNAd pattern of HEV in LT beta-/- MLN and RIPLT alpha pancreatic infiltrates, PNAd was expressed at the luminal and abluminal aspects of HEV in wild-type LN and in RIPLT alpha beta pancreata, coincident with HEC-6ST. These data highlight distinct roles of LT alpha and LT alpha beta in lymphoid organogenesis supporting the notion that HEC-6ST-dependent luminal PNAd is under regulation by LT alpha beta.


Assuntos
Tecido Linfoide/embriologia , Linfotoxina-alfa/fisiologia , Proteínas de Membrana/fisiologia , Sulfotransferases/metabolismo , Animais , Indução Enzimática , Imuno-Histoquímica , Linfotoxina-beta , Camundongos , Camundongos Transgênicos , Sulfotransferases/biossíntese
11.
J Exp Med ; 188(8): 1503-10, 1998 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-9782127

RESUMO

Secondary lymphoid tissue organogenesis requires tumor necrosis factor (TNF) and lymphotoxin alpha (LTalpha). The role of TNF in B cell positioning and formation of follicular structure was studied by comparing the location of newly produced naive recirculating and antigen-stimulated B cells in TNF-/- and TNF/LTalpha-/- mice. By creating radiation bone marrow chimeras from wild-type and TNF-/- mice, formation of normal splenic B cell follicles was shown to depend on TNF production by radiation-sensitive cells of hemopoietic origin. Reciprocal adoptive transfers of mature B cells between wild-type and knockout mice indicated that normal follicular tropism of recirculating naive B cells occurs independently of TNF derived from the recipient spleen. Moreover, soluble TNF receptor-IgG fusion protein administered in vivo failed to prevent B cell localization to the follicle or the germinal center reaction. Normal T zone tropism was observed when antigen-stimulated B cells were transferred into TNF-/- recipients, but not into TNF/LTalpha-/- recipients. This result appeared to account for the defect in isotype switching observed in intact TNF/LTalpha-/- mice because TNF/LTalpha-/- B cells, when stimulated in vitro, switched isotypes normally. Thus, TNF is necessary for creating the permissive environment for B cell movement and function, but is not itself responsible for these processes.


Assuntos
Linfócitos B/fisiologia , Baço/citologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Movimento Celular , Centro Germinativo/fisiologia , Humanos , Linfotoxina-alfa/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Fator de Necrose Tumoral alfa/deficiência
12.
J Exp Med ; 185(12): 2111-20, 1997 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-9182683

RESUMO

LTalpha-deficient (LTalpha-/-) mice show altered splenic microarchitecture. This includes loss of normal B cell-T cell compartmentalization, of follicular dendritic cell (FDC) clusters, and of ability to form germinal centers (GC). LTalpha-/- mice immunized with sheep red blood cells (SRBC) produced high levels of antigen-specific IgM but no IgG in either primary or secondary responses, demonstrating failure of Ig class switching. This inability to switch to IgG could have been due to the altered splenic microarchitecture in these mice. Alternatively, it could have been due directly to a requirement for LTalpha expression by lymphocytes cooperating in the antibody response. To investigate this, we performed reciprocal spleen cell transfers. When irradiated LTalpha-/- mice were reconstituted with wild-type splenocytes and immunized immediately with SRBC, splenic microarchitecture remained disturbed and there was no IgG response. In contrast, when irradiated wild-type animals received splenocytes from LTalpha-/- mice, follicle structure and a strong IgG response were retained. These data indicate that LTalpha-deficient B cells and T cells have no intrinsic defect in ability to generate an IgG response. Rather, the altered microenvironment characteristic of LTalpha-/- mice appears to result in impaired ability to switch to a productive IgG response. To investigate whether prolonged expression of LTalpha could alter the structure and function of spleen follicles, reciprocal bone marrow (BM) transplantation was performed. Six weeks after reconstitution of LTalpha-/- mice with wild-type BM, spleen follicle structure was partially restored, with return of FDC clusters and GC. B cell/T cell compartmentalization remained abnormal and white pulp zones were small. This was accompanied by restoration of IgG response to SRBC. Reconstitution of wild-type mice with LTalpha-/- BM resulted in loss of FDC clusters and GC, and loss of the IgG response, although compartmentalized B cell and T cell zones were largely retained. Thus, defective IgG production is not absolutely associated with abnormal B cell and T cell compartmentalization. Rather, expression of LTalpha supports the maturation of spleen follicle structure, including the development and maintenance of FDC clusters, which supports Ig class switching and an effective IgG response.


Assuntos
Imunoglobulina G/biossíntese , Linfotoxina-alfa/fisiologia , Baço/ultraestrutura , Animais , Transplante de Medula Óssea , Células Dendríticas/ultraestrutura , Eritrócitos/imunologia , Centro Germinativo/ultraestrutura , Imunoglobulina M/biossíntese , Camundongos , Camundongos Endogâmicos C57BL
13.
J Exp Med ; 172(4): 1193-200, 1990 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-2212948

RESUMO

Uncertainty regarding pathogenic mechanisms has been a major impediment to effective prevention and treatment for human neurologic diseases such as multiple sclerosis, tropical spastic paraparesis, and AIDS demyelinating disease. Here, we implicate lymphotoxin (LT) (tumor necrosis factor beta [TNF-beta]) and TNF-alpha in experimental allergic encephalomyelitis (EAE), a murine model of an autoimmune demyelinating disease. In this communication, we report that treatment of recipient mice with an antibody that neutralizes LT and TNF-alpha prevents transfer of clone-mediated EAE. LNC-8, a myelin basic protein-specific T cell line, produces high levels of LT and TNF-alpha after activation by concanavalin A, antibody to the CD-3 epsilon component of the T cell receptor, or myelin basic protein presented in the context of syngeneic spleen cells. LNC-8 cells transfer clinical signs of EAE. When LNC-8 recipient mice were also treated with TN3.19.12, a monoclonal antibody that neutralizes LT and TNF-alpha, the severity of the transferred EAE was reduced, while control antibodies did not alter the disease. The effect of anti-LT/TNF-alpha treatment was long lived and has been sustained for 5 mo. These findings suggest that LT and TNF-alpha and the T cells that produce them play an important role in EAE.


Assuntos
Encefalomielite Autoimune Experimental/etiologia , Linfotoxina-alfa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos/imunologia , Linhagem Celular , Encefalomielite Autoimune Experimental/imunologia , Linfotoxina-alfa/imunologia , Camundongos , RNA Mensageiro/análise , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/imunologia
14.
J Exp Med ; 186(12): 1997-2004, 1997 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-9396768

RESUMO

In mice deficient in either lymphotoxin alpha (LT-alpha) or type I tumor necrosis factor receptor (TNFR-I), organized clusters of follicular dendritic cells (FDC) and germinal centers (GC) are absent from the spleen. We investigated the role of LT-alpha and TNFR-I in the establishment of spleen FDC and GC structure by using reciprocal bone marrow (BM) transfer. When LT-alpha-deficient mice were reconstituted with wild-type BM, FDC organization and the ability to form GC were restored, indicating that the LT-alpha-expressing cells required to establish organized FDC are derived from BM. The role of LT-alpha in establishing organized FDC structure was further investigated by the transfer of complement receptor 1 and 2 (CR1/2)-deficient BM cells into LT-alpha-deficient mice. Organized FDC were identified with both the FDC-M1 and anti-CR1 monoclonal antibodies in these BM-chimeric mice, indicating that these cells were derived from the LT-alpha-deficient recipient. Thus, expression of LT-alpha in the BM-derived cells, but not in the non-BM-derived cells, is required for the maturation of FDC from non-BM precursor cells. In contrast, when TNFR-I-deficient mice were reconstituted with wild-type BM, they showed no detectable FDC clusters or GC formation. This indicates that TNFR-I expression on non-BM-derived cellular components is necessary for the establishment of these lymphoid structures. TNFR-I-deficient BM was able to restore FDC organization and GC formation in LT-alpha-deficient mice, indicating that formation of these structures does not require TNFR-I expression on BM-derived cells. The data in this study demonstrate that FDC organization and GC formation are controlled by both LT-alpha-expressing BM-derived cells and by TNFR-I-expressing non-BM-derived cells.


Assuntos
Células da Medula Óssea/citologia , Células Dendríticas/citologia , Linfotoxina-alfa/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Animais , Transplante de Medula Óssea , Centro Germinativo/citologia , Técnicas Imunoenzimáticas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Baço/citologia
15.
J Exp Med ; 198(11): 1643-52, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14638845

RESUMO

Although elevated levels of IgE in asthmatic patients are strongly associated with lung infiltration by activated T helper (Th) 2 cells, the physiological role of immunoglobulin E (IgE) in the airway remains largely undefined. Lymphotoxin-deficient alpha (LTalpha-/-) mice exhibit increased airway inflammation, paradoxically accompanied by diminished levels of IgE and reduced airway hyperresponsiveness in response to both environmental and induced antigen challenge. The severe lung inflammation in LTalpha-/- mice is Th1 in nature and can be alleviated by IgE reconstitution. Conversely, depletion of IgE in wild-type mice recapitulates the lung pathologies of LTalpha-/- mice. Therefore, this work has revealed that lymphotoxin is essential for IgE production, and a physiological role of IgE in the airway may consist of maintaining the balance of Th1 and Th2 responses to prevent aberrant inflammation.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Imunoglobulina E/sangue , Linfotoxina-alfa/fisiologia , Células Th1/imunologia , Animais , Líquido da Lavagem Broncoalveolar , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunoglobulina E/deficiência , Linfotoxina-alfa/genética , Camundongos , Camundongos Knockout , Mecânica Respiratória
16.
J Exp Med ; 193(11): 1227-38, 2001 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-11390430

RESUMO

Lymph nodes (LNs) are important sentinal organs, populated by circulating lymphocytes and antigen-bearing cells exiting the tissue beds. Although cellular and humoral immune responses are induced in LNs by antigenic challenge, it is not known if LNs are essential for acquired immunity. We examined immune responses in mice that lack LNs due to genetic deletion of lymphotoxin ligands or in utero blockade of membrane lymphotoxin. We report that LNs are absolutely required for generating contact hypersensitivity, a T cell-dependent cellular immune response induced by epicutaneous hapten. We show that the homing of epidermal Langerhans cells in response to hapten application is specifically directed to LNs, providing a cellular basis for this unique LN function. In contrast, the spleen cannot mediate contact hypersensitivity because antigen-bearing epidermal Langerhans cells do not access splenic white pulp. Finally, we formally demonstrate that LNs provide a unique environment essential for generating this acquired immune response by reversing the LN defect in lymphotoxin-alpha(-/)- mice, thereby restoring the capacity for contact hypersensitivity.


Assuntos
Dermatite de Contato/etiologia , Linfonodos/fisiologia , Linfotoxina-alfa/fisiologia , Animais , Movimento Celular , Células Dendríticas/fisiologia , Feminino , Células de Langerhans/fisiologia , Linfotoxina-beta , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H
17.
J Exp Med ; 186(8): 1233-40, 1997 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-9334362

RESUMO

The lymphotoxin (LT)/tumor necrosis factor (TNF) family has been implicated in the neurologic inflammatory diseases multiple sclerosis (MS) and experimental allergic encephalomyelitis (EAE). To determine the role of individual family members in EAE, C57BL/6 mice, LT-alpha-deficient (LT-alpha-/- mice), or LT-beta-deficient (LT-beta-/- mice), and their wild-type (WT) littermates were immunized with rat myelin oligodendrocyte glycoprotein (MOG) peptide 35-55. C57BL/6 and WT mice developed chronic, sustained paralytic disease with average maximum clinical scores of 3.5 and disease indices (a measure of day of onset and sustained disease scores) ranging from 367 to 663 with central nervous system (CNS) inflammation and demyelination. LT-alpha-/- mice were primed so that their splenic lymphocytes proliferated in response to MOG 35-55 and the mice produced anti-MOG antibody. However, LT-alpha-/- mice were quite resistant to EAE with low average clinical scores (<1), an average disease index of 61, and the negligible CNS inflammation and demyelination. WT T cells transferred EAE to LT-alpha-/- recipients. LT-beta-/- mice were susceptible to EAE, though less than WT, with an average maximum clinical score of 1.9 and disease index of 312. These data implicate T cell production of LT-alpha in MOG EAE and support a major role for LT-alpha3, a minor role for the LT-alpha/beta complex, and by inference, no role for TNF-alpha.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Linfotoxina-alfa/fisiologia , Sequência de Aminoácidos , Animais , Encéfalo/patologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/prevenção & controle , Linfotoxina-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Oligodendroglia/imunologia , Medula Espinal/patologia , Vacinação
18.
J Exp Med ; 159(3): 828-43, 1984 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-6421983

RESUMO

Crude preparations of interferon (IFN)-gamma derived from human peripheral blood leukocyte (PBL) cultures induced with 12-O-tetra-decanoylphorbol-13-acetate (TPA) and phytohemagglutinin (PHA) were more cytotoxic to HeLa cells than partially purified nautral or highly purified recombinant human IFN-gamma preparations. Conditioned media from PBL cultures contained, in addition to IFN-gamma, a mixture of cytotoxins, including classic lymphocyte-derived lymphotoxin (LT), and a TPA-induced cytotoxic activity produced by the adherent cell population (presumably monocytes). These two types of cytotoxins, indistinguishable in the mouse L929 cell LT assay, could be differentiated by an antiserum prepared against LT derived from the B lymphoblastoid cell line RPMI 1788. This antiserum neutralized lymphocyte-derived classic LT but failed to neutralize the activity of the monocyte-derived cytotoxin. Processing of conditioned media by sequential chromatography on silicic acid, Con A-Sepharose, and DEAE-Sephacel failed to separate IFN-gamma from the LT activity. However, this procedure did remove the monocyte-derived cytotoxic activity present in the original starting material, leaving predominantly classic LT. This LT showed a slightly basic isoelectric point (pI 7.6) which partially overlapped the more basic pI range of IFN-gamma. The two lymphokine activities also could not be completely separated by fast protein liquid chromatography or molecular sieve chromatography. LT in these partially purified preparations was associated with a protein having an apparent molecular weight of 58,000 on gel filtration. This form dissociated partially into a 20,000 mol wt species after denaturation with 0.1% NaDodSO4. IFN-gamma could be selectively removed from preparations containing both IFN-gamma and LT with the aid of monoclonal antibody to IFN-gamma. The addition of purified LT to purified E. coli-derived recombinant human IFN-gamma resulted in a marked synergistic enhancement of cytotoxicity for HeLa cells.


Assuntos
Interferon gama/fisiologia , Linfotoxina-alfa/fisiologia , Proteínas/fisiologia , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo , Adesão Celular , Precipitação Química , Células Clonais/imunologia , Citotoxicidade Imunológica , Sinergismo Farmacológico , Células HeLa/imunologia , Humanos , Interferon gama/imunologia , Interferon gama/isolamento & purificação , Linfócitos/classificação , Linfócitos/imunologia , Linfotoxina-alfa/imunologia , Linfotoxina-alfa/isolamento & purificação , Monocinas , Proteínas/imunologia , Proteínas/isolamento & purificação
19.
J Exp Med ; 188(4): 745-54, 1998 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-9705956

RESUMO

Lymphotoxin (LT)alpha knockout mice, as well as double LTalpha/tumor necrosis factor (TNF) knockout mice, show a severe splenic disorganization with nonsegregating T/B cell zones and complete absence of primary B cell follicles, follicular dendritic cell (FDC) networks, and germinal centers. In contrast, as shown previously and confirmed in this study, LTbeta-deficient mice show much more conserved T/B cell areas and a reduced but preserved capacity to form germinal centers and FDC networks. We show here that similar to the splenic phenotype of LTbeta-deficient mice, complementation of LTalpha knockout mice with TNF-expressing transgenes leads to a p55 TNF receptor-dependent restoration of B/T cell zone segregation and a partial preservation of primary B cell follicles, FDC networks, and germinal centers. Notably, upon lipopolysaccharide challenge, LTalpha knockout mice fail to produce physiological levels of TNF both in peritoneal macrophage supernatants and in their serum, indicating a coinciding deficiency in TNF expression. These findings suggest that defective TNF expression contributes to the complex phenotype of the LTalpha knockout mice, and uncover a predominant role for TNF and its p55 TNF receptor in supporting, even in the absence of LTalpha, the development and maintenance of splenic B cell follicles, FDC networks, and germinal centers.


Assuntos
Linfócitos B/fisiologia , Linfotoxina-alfa/fisiologia , Baço/fisiologia , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Antígenos CD/genética , Antígenos CD/fisiologia , Centro Germinativo , Humanos , Linfotoxina-alfa/genética , Camundongos , Camundongos Knockout , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral , Baço/citologia , Transgenes , Fator de Necrose Tumoral alfa/genética
20.
J Exp Med ; 187(7): 997-1007, 1998 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-9529316

RESUMO

The transfer of lymphocytes into severe combined immunodeficiency (SCID) mice induces a series of histological changes in the spleen, including the appearance of mature follicular dendritic cells (FDCs). Studies were undertaken to clarify the role of lymphotoxin (LT) in this process. The results show that SCID mice have a small and partially differentiated white pulp containing marginal zone and interdigitating dendritic cells, but lacking FDCs. Transferred spleen cells can segregate into T and B cell areas shortly after their injection to SCID mice. This ability is dependent on signaling through LT-beta receptor (LT-betaR), since blocking ligand-receptor interaction in recipient SCID mice ablates the capacity of the transferred cells to segregate. A week after lymphocyte transfer, host-derived FDCs appeared in the reconstituted SCID mice. This induction of FDCs is dependent on LT-betaR signaling by B cells since LT-alpha-/- B cells are incapable of inducing development of FDCs in SCID mice, even after cotransfer of LT-alpha+/+ T cells. Therefore, LT plays at least two discrete roles in splenic organization. First, it appears that LT induces the differentiation of the white pulp to create sites for lymphocyte segregation. Second, LT expression by B cells drives the maturation of FDCs and the organization of B cell follicles.


Assuntos
Linfócitos B/fisiologia , Células Dendríticas/metabolismo , Linfotoxina-alfa/fisiologia , Baço/fisiologia , Animais , Linfócitos B/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Imuno-Histoquímica , Receptor beta de Linfotoxina , Camundongos , Camundongos SCID , Microscopia Confocal , Fenótipo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Baço/citologia , Linfócitos T/metabolismo , Transplante Isogênico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA