Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Nat Immunol ; 13(10): 947-53, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22922363

RESUMEN

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.


Asunto(s)
Inmunidad Mucosa , Receptor beta de Linfotoxina/fisiología , Linfotoxina-alfa/fisiología , Obesidad , Animales , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Ciego/microbiología , Ciego/trasplante , Dieta , Metabolismo Energético , Vida Libre de Gérmenes , Interleucina-23/deficiencia , Interleucina-23/fisiología , Interleucinas/deficiencia , Interleucinas/fisiología , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/deficiencia , Linfotoxina-alfa/genética , Metagenoma , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/inmunología , Obesidad/metabolismo , Aumento de Peso/inmunología , Interleucina-22
2.
Am J Pathol ; 190(1): 252-269, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31585070

RESUMEN

The mouse lymph node (LN) can provide a niche to grow metanephric kidney to maturity. Here, we show that signaling through the lymphotoxin-ß receptor (LTßR) is critical for kidney organogenesis both in the LN and the omentum. By transplanting kidney rudiments either in the LNs of mice undergoing LTßR antagonist treatment or in the omenta of Ltbr knockout (Ltbr-/-) mice, the host LTßR signals were found to be crucial for obtaining a well-vascularized kidney graft. Indeed, defective LTßR signaling correlated with decreased expression of endothelial and angiogenic markers in kidney grafts as well as structural alterations. Because the number of glomerular endothelial cells expressing the LTßR target nuclear factor κB-inducing kinase (NIK) decreased in the absence of a functional LTßR, it was speculated that an LTßR/NIK axis mediated the angiogenetic signals required for successful ectopic kidney organogenesis, given the established role of NIK in neovascularization. However, the transplantation of kidney rudiments in omenta of Nik-/- mice revealed that NIK is dispensable for ectopic kidney vascular integration and maturation. Finally, defective LTßR signaling impaired compensatory glomerular adaptation to renal mass reduction, indicating that kidney regeneration approaches, besides whole kidney reconstruction, might benefit from the presence of LTßR signals.


Asunto(s)
Glomérulos Renales/trasplante , Tejido Linfoide/citología , Receptor beta de Linfotoxina/fisiología , Neovascularización Fisiológica , Organogénesis , Animales , Células Endoteliales/citología , Glomérulos Renales/citología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/fisiología , Regeneración , Transducción de Señal , Quinasa de Factor Nuclear kappa B
3.
J Virol ; 92(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29743364

RESUMEN

B cell-derived lymphotoxin (LT) is required for the development of follicular dendritic cell clusters for the formation of primary and secondary lymphoid follicles, but the role of T cell-derived LT in antibody response has not been well demonstrated. We observed that lymphotoxin ß-receptor (LTßR) signaling is essential for optimal humoral immune response and protection against an acute herpes simplex virus 1 (HSV-1) infection. Blocking the LTßR pathway caused poor maintenance of germinal center B (GC-B) cells and follicular helper T (Tfh) cells. Using bone marrow chimeric mice and adoptive transplantation, we determined that T cell-derived LT played an indispensable role in the humoral immune response to HSV-1. Upregulation of gamma interferon by the LTßR-Ig blockade impairs the sustainability of Tfh-like cells, leading to an impaired humoral immune response. Our findings have identified a novel role of T cell-derived LT in the humoral immune response against HSV-1 infection.IMPORTANCE Immunocompromised people are susceptible to HSV-1 infection and lethal recurrence, which could be inhibited by anti-HSV-1 humoral immune response in the host. This study sought to explore the role of T cell-derived LT in the anti-HSV-1 humoral immune response using LT-LTßR signaling-deficient mice and the LTßR-Ig blockade. The data indicate that the T cell-derived LT may play an essential role in sustaining Tfh-like cells and ensure Tfh-like cells' migration into primary or secondary follicles for further maturation. This study provides insights for vaccine development against infectious diseases.


Asunto(s)
Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Inmunidad Humoral/inmunología , Receptor beta de Linfotoxina/fisiología , Linfotoxina-alfa/metabolismo , Linfocitos T/metabolismo , Animales , Centro Germinal , Herpes Simple/metabolismo , Herpes Simple/virología , Ratones , Ratones Noqueados , Transducción de Señal
5.
Biochim Biophys Acta ; 1865(2): 204-19, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26923876

RESUMEN

The LTα1ß2 and LIGHT TNF superfamily cytokines exert pleiotropic physiological functions through the activation of their cognate lymphotoxin-ß receptor (LTßR). Interestingly, since the discovery of these proteins, accumulating evidence has pinpointed a role for LTßR signaling in carcinogenesis. Early studies have shown a potential anti-tumoral role in a subset of solid cancers either by triggering apoptosis in malignant cells or by eliciting an anti-tumor immune response. However, more recent studies provided robust evidence that LTßR signaling is also involved in diverse cell-intrinsic and microenvironment-dependent pro-oncogenic mechanisms, affecting several solid and hematological malignancies. Consequently, the usefulness of LTßR signaling axis blockade has been investigated as a potential therapeutic approach for cancer. Considering the seemingly opposite roles of LTßR signaling in diverse cancer types and their key implications for therapy, we here extensively review the different mechanisms by which LTßR activation affects carcinogenesis, focusing on the diverse contexts and different models assessed.


Asunto(s)
Receptor beta de Linfotoxina/fisiología , Neoplasias/etiología , Transducción de Señal/fisiología , Animales , Humanos , Inflamación/etiología , FN-kappa B/fisiología , Microambiente Tumoral , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
6.
Br J Haematol ; 171(5): 736-51, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26456771

RESUMEN

Lymphotoxin-mediated activation of the lymphotoxin-ß receptor (LTßR; LTBR) has been implicated in cancer, but its role in T-cell acute lymphoblastic leukaemia (T-ALL) has remained elusive. Here we show that the genes encoding lymphotoxin (LT)-α and LTß (LTA, LTB) are expressed in T-ALL patient samples, mostly of the TAL/LMO molecular subtype, and in the TEL-JAK2 transgenic mouse model of cortical/mature T-ALL (Lta, Ltb). In these mice, expression of Lta and Ltb is elevated in early stage T-ALL. Surface LTα1 ß2 protein is expressed in primary mouse T-ALL cells, but only in the absence of microenvironmental LTßR interaction. Indeed, surface LT expression is suppressed in leukaemic cells contacting Ltbr-expressing but not Ltbr-deficient stromal cells, both in vitro and in vivo, thus indicating that dynamic surface LT expression in leukaemic cells depends on interaction with its receptor. Supporting the notion that LT signalling plays a role in T-ALL, inactivation of Ltbr results in a significant delay in TEL-JAK2-induced leukaemia onset. Moreover, young asymptomatic TEL-JAK2;Ltbr(-/-) mice present markedly less leukaemic thymocytes than age-matched TEL-JAK2;Ltbr(+/+) mice and interference with LTßR function at this early stage delayed T-ALL development. We conclude that LT expression by T-ALL cells activates LTßR signalling in thymic stromal cells, thus promoting leukaemogenesis.


Asunto(s)
Receptor beta de Linfotoxina/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Expresión Génica/genética , Humanos , Inmunofenotipificación , Janus Quinasa 2/genética , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Transducción de Señal , Microambiente Tumoral/genética
7.
J Immunol ; 186(3): 1486-94, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21187446

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , Envejecimiento/metabolismo , Feto , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Bazo/inmunología , Bazo/metabolismo , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Compartimento Celular/inmunología , Femenino , Feto/anatomía & histología , Feto/inmunología , Feto/metabolismo , Receptor beta de Linfotoxina/deficiencia , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/inmunología , Bazo/citología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Mediators Inflamm ; 2013: 484378, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453421

RESUMEN

Tumor necrosis factor (TNF) and TNF receptors (TNFR) superfamily members are engaged in diverse cellular phenomena such as cellular proliferation, morphogenesis, apoptosis, inflammation, and immune regulation. Their role in regulating viral infections has been well documented. Viruses have evolved with numerous strategies to interfere with TNF-mediated signaling indicating the importance of TNF and TNFR superfamily in viral pathogenesis. Recent research reports suggest that TNF and TNFRs play an important role in the pathogenesis of HIV. TNFR signaling modulates HIV replication and HIV proteins interfere with TNF/TNFR pathways. Since immune activation and inflammation are the hallmark of HIV infection, the use of TNF inhibitors can have significant impact on HIV disease progression. In this review, we will describe how HIV infection is modulated by signaling mediated through members of TNF and TNFR superfamily and in turn how these latter could be targeted by HIV proteins. Finally, we will discuss the emerging therapeutics options based on modulation of TNF activity that could ultimately lead to the cure of HIV-infected patients.


Asunto(s)
Infecciones por VIH/inmunología , Receptores del Factor de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Apoptosis , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/etiología , Humanos , Receptor beta de Linfotoxina/fisiología , Receptores OX40/fisiología , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Transducción de Señal , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
9.
J Reprod Immunol ; 153: 103693, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35987137

RESUMEN

Intrauterine adhesion (IUA) is a fibrotic disease, with complex and multifactorial process, causing menstrual disorders, pregnancy loss or infertility. LIGHT (also named TNFSF14), mainly expressed by immune cells, has been reported to be associated with tissue fibrosis. However, the features of immunocyte subsets, the expression and roles of LIGHT and its receptor HVEM (herpes virus entry mediator) and LTßR (lymphotoxin beta receptor) in IUA remain largely unknown. Compared with the control group, we observed increased ratios of CD45+ cells, neutrophils, T cells, macrophages and decreased natural killer cells proportion, and high LIGHT expression on CD4+ T cells and macrophages in IUA endometrium. Further analysis showed there was a positive correlation between upregulated profibrotic factors (e.g., ɑ-smooth muscle actin, transforming growth factor ß1) and HVEM in IUA endometrial tissue. More importantly, recombinant human LIGHT protein directly up-regulated the expression of HVEM, LTßR, profibrotic and proinflammatory factors expression in human endometrial stromal cells. These findings reveal abnormal changes of immune cell subsets proportion and the overexpression of LIGHT-HVEM/LTßR axis in IUA endometrium, should contribute to inflammation and fibrosis formation of IUA.


Asunto(s)
Receptor beta de Linfotoxina , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Enfermedades Uterinas , Actinas , Femenino , Fibrosis/genética , Humanos , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/fisiología , Embarazo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Enfermedades Uterinas/genética , Enfermedades Uterinas/patología
10.
Ann Surg ; 253(5): 996-1003, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21368655

RESUMEN

OBJECTIVE: To determine effects of (1) parenteral nutrition (PN), (2) exogenous Lymphotoxin ß receptor (LTßR) stimulation in PN animals, and (3) exogenous LTßR blockade in chow animals on NF-κB activation pathways and products: MAdCAM-1, chemokine (C-C motif) Ligand (CCL) 19, CCL20, CCL25, interleukin (IL)-4, and IL-10. BACKGROUND: LT stimulates LTßR in Peyer's patches (PP) to activate NF-κB via the noncanonical pathway. The p100/RelB precursor yields p52/RelB producing MAdCAM-1, cytokines, and chemokines important in cell trafficking. TNFα, IL-1ß, and bacterial products stimulate the inflammatory canonical NF-κB pathway producing p65/p50 and c-Rel/p50. PN decreases LTßR, MAdCAM-1, and chemokines in PP and lowers small intestinal IgA compared with chow. METHODS: Canonical (p50 and p65) and noncanonical (p52 and Rel B) NF-κB proteins in PP were analyzed by TransAM NF-κB kit after 5 days of chow or PN, 2 days of LTßR stimulation or 3 days of LTßR blockade. MAdCAM-1, chemokines, and cytokines in PP were measured by ELISA after LTßR stimulation or blockade. RESULTS: PN significantly reduced all NF-κB proteins in PP compared with chow. Exogenous LTßR stimulation during PN increased p50, p52, Rel B, MAdCAM-1, IL-4, and IL-10 in PP, but not p65, CCL19, CCL20, or CCL25 compared with PN. LTßR blockade reduced noncanonical products (p52 and Rel B), MAdCAM-1, CCL19, CCL20, CCL25, IL-4, and IL-10 but had no effect on the inflammatory pathway (p50 and p65) compared with chow. CONCLUSION: Lack of enteral stimulation during PN decreases both canonical and noncanonical NF-κB pathways in PP. LTßR stimulation during PN feeding completely restores PP noncanonical NF-κB activity, MAdCAM-1, IL-4, IL-10, and partly the canonical pathway. LTßR blockade decreases the noncanonical NF-κB activity, MAdCAM-1, chemokines, and cytokines without effect on the canonical NF-κB activity in PP.


Asunto(s)
Receptor beta de Linfotoxina/metabolismo , FN-kappa B/metabolismo , Nutrición Parenteral/efectos adversos , Transducción de Señal , Análisis de Varianza , Animales , Moléculas de Adhesión Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunoglobulinas/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Mucosa Intestinal/metabolismo , Receptor beta de Linfotoxina/fisiología , Masculino , Ratones , Ratones Endogámicos , Mucoproteínas/metabolismo , Nutrición Parenteral/métodos , Distribución Aleatoria , Sensibilidad y Especificidad
11.
Arterioscler Thromb Vasc Biol ; 30(3): 395-402, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20139367

RESUMEN

OBJECTIVE: Mouse aorta smooth muscle cells (SMC) express tumor necrosis factor receptor superfamily member 1A (TNFR-1) and lymphotoxin beta-receptor (LTbetaR). Circumstantial evidence has linked the SMC LTbetaR to tertiary lymphoid organogenesis in hyperlipidemic mice. Here, we explored TNFR-1 and LTbetaR signaling in cultured SMC. METHODS AND RESULTS: TNFR-1 signaling activated the classical RelA NF-kappaB pathway, whereas LTbetaR signaling activated the classical RelA and alternative RelB NF-kappaB pathways, and both signaling pathways synergized to enhance p100 inhibitor processing to the p52 subunit of NF-kappaB. Microarrays showed that simultaneous TNFR-1/LTbetaR activation resulted in elevated mRNA encoding leukocyte homeostatic chemokines CCL2, CCL5, CXCL1, and CX3CL1. Importantly, SMC acquired features of lymphoid tissue organizers, which control tertiary lymphoid organogenesis in autoimmune diseases through hyperinduction of CCL7, CCL9, CXCL13, CCL19, CXCL16, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1. TNFR-1/LTbetaR cross-talk resulted in augmented secretion of lymphorganogenic chemokine proteins. Supernatants of TNFR-1/LTbetaR-activated SMC markedly supported migration of splenic T cells, B cells, and macrophages/dendritic cells. Experiments with ltbr(-/-) SMC indicated that LTbetaR-RelB activation was obligatory to generate the lymphoid tissue organizer phenotype. CONCLUSIONS: SMC may participate in the formation of tertiary lymphoid tissue in atherosclerosis by upregulation of lymphorganogenic chemokines involved in T-lymphocyte, B-lymphocyte, and macrophage/dendritic cell attraction.


Asunto(s)
Diferenciación Celular/fisiología , Tejido Linfoide/citología , Receptor beta de Linfotoxina/fisiología , Miocitos del Músculo Liso/citología , FN-kappa B/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Aorta/citología , Aorta/efectos de los fármacos , Aorta/fisiología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Tejido Linfoide/fisiología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Factor de Necrosis Tumoral alfa/farmacología
12.
J Immunol ; 182(9): 5439-45, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380791

RESUMEN

The formation of lymph nodes is a complex process crucially controlled through triggering of LTbetaR on mesenchymal cells by LTalpha(1)beta(2) expressing lymphoid tissue inducer (LTi) cells. This leads to the induction of chemokines to attract more hematopoietic cells and adhesion molecules to retain them. In this study, we show that the extravasation of the first hematopoietic cells at future lymph node locations occurs independently of LTalpha and that these cells, expressing TNF-related activation-induced cytokine (TRANCE), are the earliest LTi cells. By paracrine signaling the first expression of LTalpha(1)beta(2) is induced. Subsequent LTbetaR triggering on mesenchymal cells leads to their differentiation to stromal organizers, which now also start to express TRANCE, IL-7, as well as VEGF-C, in addition to the induced adhesion molecules and chemokines. Both TRANCE and IL-7 will further induce the expression of LTalpha(1)beta(2) on newly arrived immature LTi cells, resulting in more LTbetaR triggering, generating a positive feedback loop. Thus, LTbetaR triggering by LTi cells during lymph node development creates a local environment to which hematopoietic precursors are attracted and where they locally differentiate into fully mature, LTalpha(1)beta(2) expressing, LTi cells. Furthermore, the same signals may regulate lymphangiogenesis to the lymph node through induction of VEGF-C.


Asunto(s)
Proteínas Angiogénicas/biosíntesis , Citocinas/biosíntesis , Ganglios Linfáticos/inmunología , Receptor beta de Linfotoxina/fisiología , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología , Proteínas Angiogénicas/genética , Animales , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Citocinas/genética , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/embriología , Ganglios Linfáticos/metabolismo , Tejido Linfoide/embriología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Heterotrímero de Linfotoxina alfa1 y beta2/biosíntesis , Heterotrímero de Linfotoxina alfa1 y beta2/deficiencia , Heterotrímero de Linfotoxina alfa1 y beta2/genética , Heterotrímero de Linfotoxina alfa1 y beta2/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ligando RANK/biosíntesis , Ligando RANK/genética , Células del Estroma/inmunología , Células del Estroma/metabolismo
13.
Front Immunol ; 12: 675538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054863

RESUMEN

Tertiary lymphoid structures (TLS) are ectopically formed aggregates of organized lymphocytes and antigen-presenting cells that occur in solid tissues as part of a chronic inflammation response. Sharing structural and functional characteristics with conventional secondary lymphoid organs (SLO) including discrete T cell zones, B cell zones, marginal zones with antigen presenting cells, reticular stromal networks, and high endothelial venues (HEV), TLS are prominent centers of antigen presentation and adaptive immune activation within the periphery. TLS share many signaling axes and leukocyte recruitment schemes with SLO regarding their formation and function. In cancer, their presence confers positive prognostic value across a wide spectrum of indications, spurring interest in their artificial induction as either a new form of immunotherapy, or as a means to augment other cell or immunotherapies. Here, we review approaches for inducible (iTLS) that utilize chemokines, inflammatory factors, or cellular analogues vital to TLS formation and that often mirror conventional SLO organogenesis. This review also addresses biomaterials that have been or might be suitable for iTLS, and discusses remaining challenges facing iTLS manufacturing approaches for clinical translation.


Asunto(s)
Inmunoterapia , Estructuras Linfoides Terciarias/inmunología , Colágeno/metabolismo , Reacción a Cuerpo Extraño/prevención & control , Humanos , Hidrogeles , Receptor beta de Linfotoxina/fisiología , Nanopartículas , Neoplasias/inmunología , Neoplasias/terapia , Estructuras Linfoides Terciarias/fisiopatología
14.
Front Immunol ; 12: 712632, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335629

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina/fisiología , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Antibacterianos/inmunología , Autoinmunidad , Moléculas de Adhesión Celular/metabolismo , Quimiocinas/metabolismo , Citrobacter rodentium/inmunología , Cruzamientos Genéticos , Regulación del Desarrollo de la Expresión Génica , Homeostasis/inmunología , Inmunoglobulina A/biosíntesis , Inmunoglobulina A/inmunología , Inflamación , Células Asesinas Naturales/inmunología , Tejido Linfoide/citología , Receptor beta de Linfotoxina/biosíntesis , Receptor beta de Linfotoxina/deficiencia , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos MRL lpr , Ratones Transgénicos , Neutrófilos/inmunología , Eliminación de Secuencia , Organismos Libres de Patógenos Específicos , Esplenomegalia/inmunología
15.
Eur J Immunol ; 39(10): 2765-78, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19757439

RESUMEN

Nose-associated lymphoid tissue (NALT) in the rodent upper respiratory tract develops postnatally and is considered to be independent of several factors known to be involved in the organogenesis of LN and Peyer's patches (PP). In this study we demonstrate that at least two different pathways result in NALT development. Following NALT anlage formation the intrinsic pathway relies on a signaling cascade including those mediated through the chemokine receptor CXCR5 and the lymphotoxin beta receptor (LTbetaR). This allows for the formation of high endothelial venules and thereby the recruitment of lymphocytes into NALT. Alternatively, high endothelial venule formation and lymphocyte recruitment can be induced in the NALT anlage by environmental signals, which are independent of LT-betaR and chemokine receptor CXCR5 signaling but in part rely on CD40 ligand. Thus, our study identifies a novel mechanism that facilitates the rescue of NALT development at late stages in adult life independent of the canonical LTbetaR-CXCR5 signaling axis.


Asunto(s)
Antígenos/inmunología , Tejido Linfoide/crecimiento & desarrollo , Receptor beta de Linfotoxina/fisiología , Mucosa Nasal/crecimiento & desarrollo , Receptores CXCR5/fisiología , Transducción de Señal/inmunología , Traslado Adoptivo , Envejecimiento/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos de Superficie/metabolismo , Linfocitos B/inmunología , Ligando de CD40/inmunología , Moléculas de Adhesión Celular/metabolismo , Recuento de Células , Movimiento Celular/inmunología , Vida Libre de Gérmenes/inmunología , Ganglios Linfáticos/citología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfocitos/citología , Tejido Linfoide/irrigación sanguínea , Tejido Linfoide/patología , Linfotoxina-alfa/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mucoproteínas , Mucosa Nasal/irrigación sanguínea , Mucosa Nasal/patología , Propionibacterium acnes/inmunología , Bazo/citología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Vénulas/crecimiento & desarrollo , Vénulas/metabolismo , Vénulas/patología
16.
Hepatology ; 49(1): 227-39, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19111021

RESUMEN

UNLABELLED: Lymphotoxin-beta (LTbeta) is a proinflammatory cytokine and a member of the tumor necrosis factor (TNF) superfamily known for its role in mediating lymph node development and homeostasis. Our recent studies suggest a role for LTbeta in mediating the pathogenesis of human chronic liver disease. We hypothesize that LTbeta co-ordinates the wound healing response in liver injury via direct effects on hepatic stellate cells. This study used the choline-deficient, ethionine-supplemented (CDE) dietary model of chronic liver injury, which induces inflammation, liver progenitor cell proliferation, and portal fibrosis, to assess (1) the cellular expression of LTbeta, and (2) the role of LTbeta receptor (LTbetaR) in mediating wound healing, in LTbetaR(-/-) versus wild-type mice. In addition, primary isolates of hepatic stellate cells were treated with LTbetaR-ligands LTbeta and LTbeta-related inducible ligand competing for glycoprotein D binding to herpesvirus entry mediator on T cells (LIGHT), and mediators of hepatic stellate cell function and fibrogenesis were assessed. LTbeta was localized to progenitor cells immediately adjacent to activated hepatic stellate cells in the periportal region of the liver in wild-type mice fed the CDE diet. LTbetaR(-/-) mice fed the CDE diet showed significantly reduced fibrosis and a dysregulated immune response. LTbetaR was demonstrated on isolated hepatic stellate cells, which when stimulated by LTbeta and LIGHT, activated the nuclear factor kappa B (NF-kappaB) signaling pathway. Neither LTbeta nor LIGHT had any effect on alpha-smooth muscle actin, tissue inhibitor of metalloproteinase 1, transforming growth factor beta, or procollagen alpha(1)(I) expression; however, leukocyte recruitment-associated factors intercellular adhesion molecule 1 and regulated upon activation T cells expressed and secreted (RANTES) were markedly up-regulated. RANTES caused the chemotaxis of a liver progenitor cell line expressing CCR5. CONCLUSION: This study suggests that LTbetaR on hepatic stellate cells may be involved in paracrine signaling with nearby LTbeta-expressing liver progenitor cells mediating recruitment of progenitor cells, hepatic stellate cells, and leukocytes required for wound healing and regeneration during chronic liver injury.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Hígado/lesiones , Heterotrímero de Linfotoxina alfa1 y beta2/fisiología , Receptor beta de Linfotoxina/fisiología , Cicatrización de Heridas/fisiología , Animales , Línea Celular , Proliferación Celular , Células Cultivadas , Deficiencia de Colina/fisiopatología , Colágeno Tipo I/biosíntesis , Modelos Animales de Enfermedad , Etionina/fisiología , Regeneración Hepática/fisiología , Heterotrímero de Linfotoxina alfa1 y beta2/biosíntesis , Masculino , Ratones , Ratas
17.
J Immunol ; 181(9): 6189-200, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941209

RESUMEN

Mesenchymal stromal cells are crucial components of secondary lymphoid organs (SLOs). Organogenesis of SLOs involves specialized stromal cells, designated lymphoid tissue organizer (LTo) in the embryonic anlagen; in the adult, several distinct stromal lineages construct elaborate tissue architecture and regulate lymphocyte compartmentalization. The relationship between the LTo and adult stromal cells, however, remains unclear, as does the precise number of stromal cell types that constitute mature SLOs are unclear. From mouse lymph nodes, we established a VCAM-1(+)ICAM-1(+)MAdCAM-1(+) reticular cell line that can produce CXCL13 upon LTbetaR stimulation and support primary B cell adhesion and migration in vitro. A similar stromal population sharing many characteristics with the LTo, designated marginal reticular cells (MRCs), was found in the outer follicular region immediately underneath the subcapsular sinus of lymph nodes. Moreover, MRCs were commonly observed at particular sites in various SLOs even in Rag2(-/-) mice, but were not found in ectopic lymphoid tissues, suggesting that MRCs are a developmentally determined element. These findings lead to a comprehensive view of the stromal composition and architecture of SLOs.


Asunto(s)
Tejido Linfoide/citología , Tejido Linfoide/inmunología , Mesodermo/citología , Mesodermo/inmunología , Envejecimiento/inmunología , Animales , Animales Recién Nacidos , Linfocitos B/citología , Linfocitos B/inmunología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Quimiocina CXCL13/biosíntesis , Fibroblastos/inmunología , Fibroblastos/metabolismo , Humanos , Tejido Linfoide/embriología , Tejido Linfoide/metabolismo , Receptor beta de Linfotoxina/fisiología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/inmunología , Células del Estroma/citología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Quinasa de Factor Nuclear kappa B
18.
Exp Parasitol ; 126(3): 326-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20433831

RESUMEN

Tumor necrosis factor (TNF) has long been recognized to promote malaria parasite killing, but also to contribute to the development of severe malaria disease. The precise molecular mechanisms that influence these different outcomes in malaria patients are not well understood, but the virulence and drug-resistance phenotype of malaria parasites and the genetic background and age of patients are likely to be important determinants. In the past few years, important roles for other TNF family members in host immune responses to malaria parasites and the induction of disease pathology have been discovered. In this review, we will summarize these more recent findings and highlight major gaps in our current knowledge. We will also discuss future research strategies that may allow us to better understand the sometimes subtle and intricate effects of TNF family molecules during malaria infection.


Asunto(s)
Malaria/inmunología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Evolución Biológica , Humanos , Receptor beta de Linfotoxina/fisiología , Malaria/genética , Malaria Cerebral/etiología , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Selección Genética , Transducción de Señal , Linfocitos T Reguladores/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
19.
Dev Comp Immunol ; 32(8): 980-91, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18329710

RESUMEN

Gut-associated lymphoid tissue (GALT) development requires interaction with the intestinal microbiota. Because murine secondary lymphoid tissue development is driven by positive feedback interactions between B cells and stromal cells, we used in situ hybridization to determine whether intestinal commensals influence such interactions during rabbit appendix development. The features of positive feedback interactions we examined (CXCL13 mRNA expression, B cell accumulation and FDC differentiation) increased during early follicle development, but stalled in the absence of intestinal commensals. These features were reinitiated by commensals that stimulated follicle development and intrafollicular B cell proliferation. Our results suggest that rabbit appendix follicles develop in two phases: an initial phase of B cell recruitment to nascent follicles, possibly through positive feedback interactions, and a subsequent phase of intrafollicular B cell proliferation stimulated by intestinal commensals. In addition, we found that intestinal commensals stimulate appendix CCL21 mRNA expression and T cell area formation.


Asunto(s)
Apéndice/inmunología , Linfocitos B/fisiología , Intestinos/microbiología , Linfocitos T/fisiología , Animales , Apéndice/microbiología , Quimiocina CCL21/genética , Quimiocina CXCL13/genética , Activación de Linfocitos , Receptor beta de Linfotoxina/fisiología , ARN Mensajero/análisis , Conejos , Receptores de Complemento 3b/análisis , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Transducción de Señal
20.
Clin Cancer Res ; 13(17): 5202-10, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17785576

RESUMEN

PURPOSE: One of the impediments of immunotherapy against cancer is the suppression of tumor-specific CTLs in the tumor microenvironment, partly due to the selective inhibition of the perforin pathway and the emergence of Fas-resistant tumors. Therefore, we sought to identify perforin- and Fas-independent cytotoxic pathways and explored the potential of targeting LTbetaR with tumor-specific CTLs to induce tumor rejection in vivo. EXPERIMENTAL DESIGN: Fas-resistant tumors were examined for their susceptibility to perforin-deficient (pfp) CTLs via CTL adoptive transfer in mouse models of experimental lung metastasis. The specificity of LTbetaR, a cell surface death receptor, in causing tumor rejection by CTLs was analyzed by LTbetaR-specific neutralizing monoclonal antibody in vitro. The specificity and efficacy of LTbetaR in the suppression of established tumors was further investigated by silencing LTbetaR in tumor cells in vivo. RESULTS: pfp CTLs exhibited significant cytotoxicity against Fas-resistant tumors in vivo. The perforin- and Fas-independent cytotoxicity was directly mediated, at least in part, by the adoptively transferred CTLs. It was observed that LTbetaR was expressed on the tumor cell surface, and LTalpha, LTbeta, and LIGHT, all of which are ligands for LTbetaR, were either constitutively expressed or activated in the tumor-specific CTLs and primary CD8(+) T cells. Blocking LTbetaR with LTbetaR-specific neutralizing monoclonal antibody decreased CTL cytotoxicity in vitro. Silencing LTbetaR using LTbetaR-specific short hairpin RNA reduced the ability of pfp CTLs to induce tumor rejection in vivo. CONCLUSION: LTbetaR directly mediates CTL-directed tumor rejection in vivo. Targeting LTbetaR with tumor-specific CTLs is a potential therapeutic approach.


Asunto(s)
Inmunoterapia Adoptiva , Linfotoxina beta/inmunología , Neoplasias/terapia , Linfocitos T Citotóxicos/inmunología , Animales , Citotoxicidad Inmunológica , Femenino , Rechazo de Injerto , Interferón gamma/farmacología , Receptor beta de Linfotoxina/análisis , Receptor beta de Linfotoxina/fisiología , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Perforina , Proteínas Citotóxicas Formadoras de Poros/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Receptor fas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA