Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Int Immunol ; 34(1): 45-52, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687536

RESUMEN

Medullary thymic epithelial cells (mTECs) help shape the thymic microenvironment for T-cell development by expressing a variety of peripheral tissue-restricted antigens (TRAs). The self-tolerance of T cells is established by negative selection of autoreactive T cells that bind to TRAs. To increase the diversity of TRAs, a fraction of mTECs terminally differentiates into distinct subsets resembling atypical types of epithelial cells in specific peripheral tissues. As such, thymic tuft cells that express peripheral tuft cell genes have recently emerged. Here, we show that the transcription factor SRY-box transcription factor 4 (Sox4) is highly expressed in mTECs and is essential for the development of thymic tuft cells. Mice lacking Sox4 specifically in TECs had a significantly reduced number of thymic tuft cells with no effect on the differentiation of other mTEC subsets, including autoimmune regulator (Aire)+ and Ccl21a+ mTECs. Furthermore, Sox4 expression was diminished in mice deficient in TEC-specific lymphotoxin ß receptor (LTßR), indicating a role for the LTßR-Sox4 axis in the differentiation of thymic tuft cells. Given that Sox4 promotes differentiation of peripheral tuft cells, our findings suggest that mTECs employ the same transcriptional program as peripheral epithelial cells. This mechanism may explain how mTECs diversify peripheral antigen expression to project an immunological self within the thymic medulla.


Asunto(s)
Receptor beta de Linfotoxina/genética , Factores de Transcripción SOXC/genética , Timo/inmunología , Animales , Diferenciación Celular/inmunología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción SOXC/inmunología , Transducción de Señal/genética , Timo/citología
2.
J Exp Med ; 218(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33724364

RESUMEN

The spleen contains a myriad of conventional dendritic cell (cDC) subsets that protect against systemic pathogen dissemination by bridging antigen detection to the induction of adaptive immunity. How cDC subsets differentiate in the splenic environment is poorly understood. Here, we report that LTα1ß2-expressing Rorgt+ ILC3s, together with B cells, control the splenic cDC niche size and the terminal differentiation of Sirpα+CD4+Esam+ cDC2s, independently of the microbiota and of bone marrow pre-cDC output. Whereas the size of the splenic cDC niche depended on lymphotoxin signaling only during a restricted time frame, the homeostasis of Sirpα+CD4+Esam+ cDC2s required continuous lymphotoxin input. This latter property made Sirpα+CD4+Esam+ cDC2s uniquely susceptible to pharmacological interventions with LTßR agonists and antagonists and to ILC reconstitution strategies. Together, our findings demonstrate that LTα1ß2-expressing Rorgt+ ILC3s drive splenic cDC differentiation and highlight the critical role of ILC3s as perpetual regulators of lymphoid tissue homeostasis.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata , Tejido Linfoide/inmunología , Linfotoxina-alfa/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Dendríticas/metabolismo , Femenino , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal/genética , Bazo/citología , Bazo/metabolismo
3.
Immunohorizons ; 4(11): 688-700, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154038

RESUMEN

Autoimmune pancreatitis (AIP) is a rare form of chronic pancreatitis, for which treatment options, especially the long-term management, are limited. The only therapy that has been established and accepted so far is corticosteroids, but the relapse rate is significant. In the current study, we discern the effector mechanisms of targeted LTßR pathway inhibition using LTßR-Ig. Furthermore, the efficacy of LTßR-Ig therapy is compared with the depletion of immune cell subsets (CD4+ and CD20+), which are suggested to play a pathological role in AIP development. Three well-established mouse models of AIP were used to examine treatment efficacies and mechanisms. Tg(Ela1-Lta,b) mice represent a genetic model, in which AIP develops spontaneously. In MRL/Mp and IL-10-/- mice, AIP is induced by repeated polyinosinic:polycytidylic acid injection. Mice with AIP were treated with anti-CD20, anti-CD4 mAbs, or targeted LTßR-Ig. LTßR-Ig and anti-CD20 treatment led to significant improvement of AIP, including a decrease in autoantibody production and pancreatic inflammation in Tg(Ela1-Lta,b) and IL-10-/- mice. The molecular mechanism of this beneficial effect possibly involves the downregulation of Stat3 and noncanonical NF-κb activation. Anti-CD4 treatment reduced Th1 and Th2 signature but did not alleviate AIP. Additionally, in contrast to anti-CD20 or anti-CD4 treatments, blocking LTßR signaling disrupted tertiary lymphoid organs in all three models. We demonstrate that treatment with LTßR-Ig or anti-CD20 Ab alleviated murine AIP. LTßR-Ig treatment for AIP was effective in both lymphotoxin-dependent and lymphotoxin-independent AIP models, possibly because of its dual anti-inflammatory and antiautoimmune mechanisms.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Pancreatitis Autoinmune/tratamiento farmacológico , Inmunoglobulina G/farmacología , Interleucina-10/metabolismo , Receptor beta de Linfotoxina/efectos de los fármacos , Animales , Antígenos CD20/inmunología , Pancreatitis Autoinmune/inducido químicamente , Pancreatitis Autoinmune/patología , Antígenos CD4/inmunología , Modelos Animales de Enfermedad , Femenino , Interleucina-10/genética , Receptor beta de Linfotoxina/inmunología , Masculino , Ratones , Ratones Transgénicos , Poli I-C/administración & dosificación , Transducción de Señal/inmunología
4.
Nat Commun ; 11(1): 2198, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366944

RESUMEN

The thymus supports multiple αß T cell lineages that are functionally distinct, but mechanisms that control this multifaceted development are poorly understood. Here we examine medullary thymic epithelial cell (mTEC) heterogeneity and its influence on CD1d-restricted iNKT cells. We find three distinct mTEClow subsets distinguished by surface, intracellular and secreted molecules, and identify LTßR as a cell-autonomous controller of their development. Importantly, this mTEC heterogeneity enables the thymus to differentially control iNKT sublineages possessing distinct effector properties. mTEC expression of LTßR is essential for the development thymic tuft cells which regulate NKT2 via IL-25, while LTßR controls CD104+CCL21+ mTEClow that are capable of IL-15-transpresentation for regulating NKT1 and NKT17. Finally, mTECs regulate both iNKT-mediated activation of thymic dendritic cells, and iNKT availability in extrathymic sites. In conclusion, mTEC specialization controls intrathymic iNKT cell development and function, and determines iNKT pool size in peripheral tissues.


Asunto(s)
Diferenciación Celular/inmunología , Células Epiteliales/inmunología , Células T Asesinas Naturales/inmunología , Timocitos/inmunología , Timo/inmunología , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Antígenos CD1d/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proliferación Celular/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos/inmunología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/metabolismo
5.
J Immunol ; 204(5): 1085-1090, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969387

RESUMEN

Lymphotoxin ß receptor (LTßR) signaling is crucial for lymphoid tissue organogenesis and immune homeostasis. To identify novel regulatory mechanisms for signaling, we implemented a two-step screen that uses coexpression analysis of human fibroblasts undergoing LTßR stimulation and affinity-purification mass spectrometry for the LTßR signaling protein TNFR-associated factor 3 (TRAF3). We identify Ewing sarcoma (EWS) protein as a novel LTßR signaling component that associates with TRAF3 but not with TNFR-associated factor 2 (TRAF2). The EWS:TRAF3 complex forms under unligated conditions that are disrupted following activation of the LTßR. We conclude that EWS limits expression of proinflammatory molecules, GM-CSF, and ERK-2, promoting immune homeostasis.


Asunto(s)
Receptor beta de Linfotoxina/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Complejos Multiproteicos/inmunología , Proteína EWS de Unión a ARN/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Células HEK293 , Humanos , Receptor beta de Linfotoxina/genética , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Complejos Multiproteicos/genética , Proteína EWS de Unión a ARN/genética , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología
6.
Sci Immunol ; 4(42)2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31862865

RESUMEN

Redundant mechanisms support immunoglobulin A (IgA) responses to intestinal antigens. These include multiple priming sites [mesenteric lymph nodes (MLNs), Peyer's patches, and isolated lymphoid follicles] and various cytokines that promote class switch to IgA, even in the absence of T cells. Despite these backup mechanisms, vaccination against enteric pathogens such as rotavirus has limited success in some populations. Genetic and environmental signals experienced during early life are known to influence mucosal immunity, yet the mechanisms for how these exposures operate remain unclear. Here, we used rotavirus infection to follow antigen-specific IgA responses through time and in different gut compartments. Using genetic and pharmacological approaches, we tested the role of the lymphotoxin (LT) pathway-known to support IgA responses-at different developmental stages. We found that LT-ß receptor (LTßR) signaling in early life programs intestinal IgA responses in adulthood by affecting antibody class switch recombination to IgA and subsequent generation of IgA antibody-secreting cells within an intact MLN. In addition, early-life LTßR signaling dictates the phenotype and function of MLN stromal cells to support IgA responses in the adult. Collectively, our studies uncover new mechanistic insights into how early-life LTßR signaling affects mucosal immune responses during adulthood.


Asunto(s)
Inmunoglobulina A/inmunología , Ganglios Linfáticos/inmunología , Receptor beta de Linfotoxina/inmunología , Linfotoxina-alfa/inmunología , Mesenterio/inmunología , Células del Estroma/inmunología , Animales , Heces/microbiología , Femenino , Inmunidad Mucosa , Ganglios Linfáticos/citología , Receptor beta de Linfotoxina/genética , Linfotoxina-alfa/genética , Masculino , Mesenterio/citología , Ratones Endogámicos C57BL , Ratones Noqueados
7.
J Exp Med ; 216(10): 2242-2252, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31324739

RESUMEN

The induction of adaptive immunity is dependent on the structural organization of LNs, which is in turn governed by the stromal cells that underpin LN architecture. Using a novel fate-mapping mouse model, we trace the developmental origin of mesenchymal LN stromal cells (mLNSCs) to a previously undescribed embryonic fibroblast activation protein-α (FAP)+ progenitor. FAP+ cells of the LN anlagen express lymphotoxin ß receptor (LTßR) and vascular cell adhesion molecule (VCAM), but not intercellular adhesion molecule (ICAM), suggesting they are early mesenchymal lymphoid tissue organizer (mLTo) cells. Clonal labeling shows that FAP+ progenitors locally differentiate into mLNSCs. This process is also coopted in nonlymphoid tissues in response to infection to facilitate the development of tertiary lymphoid structures, thereby mimicking the process of LN ontogeny in response to infection.


Asunto(s)
Embrión de Mamíferos/inmunología , Gelatinasas/inmunología , Ganglios Linfáticos/inmunología , Proteínas de la Membrana/inmunología , Células Madre Mesenquimatosas/inmunología , Modelos Inmunológicos , Serina Endopeptidasas/inmunología , Animales , Embrión de Mamíferos/citología , Endopeptidasas , Gelatinasas/genética , Ganglios Linfáticos/citología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Serina Endopeptidasas/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
8.
J Immunol ; 202(10): 2999-3007, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30952816

RESUMEN

The lymphatic vasculature is an important route for dendritic cell (DC) or tumor cell migration from peripheral tissues to draining lymph nodes (DLNs). However, the underlying molecular and cellular mechanisms remain poorly understood. In this study, using conventional bone marrow chimeric mice and additional UVB radiation, we found that deficiency of LIGHT but not lymphotoxin (LT) α1ß2, likely on radioresistant Langerhans cells (LCs), resulted in impaired skin DC migration to DLNs during LPS-induced inflammation. In addition, LT ß receptor (LTßR), but not herpes virus entry mediator, was found to be the receptor of LIGHT controlling DC migration. Furthermore, conditional deficiency of LTßR in Tie2 cre or Lyve1 cre mice, but not in LTßR-deficient bone marrow chimeric mice, impaired DC migration, suggesting an important role of LTßR in radioresistant lymphatic endothelial cells (LECs), although the role of LTßR in blood endothelial cells remains intriguing. Mechanistically, the gene expression of both CCL21 and CCL19 was found to be reduced in skin LECs isolated from LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice compared with their controls upon LPS stimulation. Soluble recombinant LIGHT was able to upregulate CCL21 and CCL19 gene expression on SVEC4-10 endothelial cells. Doxycycline, an inhibitor of soluble LIGHT release in the inflamed skin, impaired skin CCL21 and CCL19 expression and DC migration. In addition, melanoma cell metastasis to DLNs was also inhibited in LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice. Together, our data suggest, to our knowledge, a previously unrecognized scenario in which LCs activate LECs via the LIGHT-LTßR signaling axis to promote DC migration or tumor cell metastasis.


Asunto(s)
Células Endoteliales/inmunología , Células de Langerhans/inmunología , Vasos Linfáticos/inmunología , Receptor beta de Linfotoxina/inmunología , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Células Endoteliales/patología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Células de Langerhans/patología , Lipopolisacáridos/toxicidad , Vasos Linfáticos/patología , Receptor beta de Linfotoxina/genética , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
9.
Sci Rep ; 8(1): 17727, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30531962

RESUMEN

Though lymphotoxin (LT) is highly expressed by type I helper T (Th1) cells, its contribution to CD4+ T cell differentiation during infections and diseases remains a mystery. In HSV-1 infection, we observed that LTßR signaling is required to limit the Th1 response. Using bone marrow chimeric mice, mixed-T-cell chimeric mice, and LTßR in vivo blockades, we unexpectedly observed that LT, especially T cell-derived LT, played an indispensable role in limiting the Th1 response. The LTßR-Ig blockade promoted the Th1 response by increasing infiltration of monocytes and monocyte-derived DCs and up-regulating IL-12 secretion in the lymphoid environment. Our findings identified a novel role for T cell-derived LT in manipulating Th1 differentiation.


Asunto(s)
Herpes Simple/inmunología , Herpesvirus Humano 1/inmunología , Linfotoxina-alfa/inmunología , Células TH1/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Interleucina-12/inmunología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/inmunología , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología
10.
J Exp Med ; 215(12): 2984-2993, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30425120

RESUMEN

The emigration of mature thymocytes from the thymus is critical for establishing peripheral T cell compartments. However, the pathways controlling this process and the timing of egress in relation to postselection developmental stages are poorly defined. Here, we reexamine thymocyte egress and test current and opposing models in relation to the requirement for LTßR, a regulator of thymic microenvironments and thymocyte emigration. Using cell-specific gene targeting, we show that the requirement for LTßR in thymocyte egress is distinct from its control of thymic epithelium and instead maps to expression by endothelial cells. By separating emigration into sequential phases of perivascular space (PVS) entry and transendothelial migration, we reveal a developmentally ordered program of egress where LTßR operates to rate limit access to the PVS. Collectively, we show the process of thymic emigration ensures only the most mature thymocytes leave the thymus and demonstrate a role for LTßR in the initiation of thymus emigration that segregates from its control of medulla organization.


Asunto(s)
Movimiento Celular/inmunología , Células Endoteliales/inmunología , Receptor beta de Linfotoxina/inmunología , Timocitos/inmunología , Timo/inmunología , Animales , Movimiento Celular/genética , Células Endoteliales/citología , Receptor beta de Linfotoxina/genética , Ratones , Ratones Noqueados , Timocitos/citología , Timo/citología
11.
Arthritis Rheumatol ; 70(9): 1470-1480, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29604186

RESUMEN

OBJECTIVE: To evaluate the clinical efficacy and safety of baminercept, a lymphotoxin ß receptor IgG fusion protein (LTßR-Ig), for the treatment of primary Sjögren's syndrome (SS), and to explore the possible mechanisms of action of this treatment. METHODS: In this multicenter trial, 52 patients with primary SS were randomized in a 2:1 ratio to receive subcutaneous injections of 100 mg of baminercept every week for 24 weeks or matching placebo. The primary end point was the change between screening and week 24 in the stimulated whole salivary flow (SWSF) rate. Secondary end points included the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI), as well as measurements of select chemokines and cytokines and enumeration of peripheral blood B and T cell subsets. RESULTS: The change from baseline to week 24 in the SWSF rate was not significantly different between the baminercept and placebo treatment groups (baseline-adjusted mean change -0.01 versus 0.07 ml/minute; P = 0.332). The change in the ESSDAI during treatment was also not significantly different between the treatment groups (baseline-adjusted mean change -1.23 versus -0.15; P = 0.104). Although the incidence of adverse events was similar between the treatment groups, baminercept therapy was associated with a higher incidence of liver toxicity, including 2 serious adverse events. Baminercept also produced a significant decrease in plasma levels of CXCL13 and significant changes in the number of circulating B and T cells, consistent with its known inhibitory effects on LTßR signaling. CONCLUSION: In this trial, treatment with baminercept failed to significantly improve glandular and extraglandular disease in patients with primary SS, despite evidence from mechanistic studies showing that it blocks LTßR signaling.


Asunto(s)
Proteínas Recombinantes de Fusión/uso terapéutico , Síndrome de Sjögren/tratamiento farmacológico , Adulto , Anciano , Linfocitos B/efectos de los fármacos , Quimiocina CXCL13/sangre , Método Doble Ciego , Femenino , Humanos , Receptor beta de Linfotoxina/inmunología , Masculino , Persona de Mediana Edad , Proteínas Recombinantes de Fusión/inmunología , Síndrome de Sjögren/sangre , Síndrome de Sjögren/inmunología , Linfocitos T/efectos de los fármacos , Resultado del Tratamiento
12.
Nat Immunol ; 19(4): 342-353, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29507355

RESUMEN

Pathogens have co-evolved with mosquitoes to optimize transmission to hosts. Mosquito salivary-gland extract is known to modulate host immune responses and facilitate pathogen transmission, but the underlying molecular mechanisms of this have remained unknown. In this study, we identified and characterized a prominent 15-kilodalton protein, LTRIN, obtained from the salivary glands of the mosquito Aedes aegypti. LTRIN expression was upregulated in blood-fed mosquitoes, and LTRIN facilitated the transmission of Zika virus (ZIKV) and exacerbated its pathogenicity by interfering with signaling through the lymphotoxin-ß receptor (LTßR). Mechanically, LTRIN bound to LTßR and 'preferentially' inhibited signaling via the transcription factor NF-κB and the production of inflammatory cytokines by interfering with the dimerization of LTßR during infection with ZIKV. Furthermore, treatment with antibody to LTRIN inhibited mosquito-mediated infection with ZIKV, and abolishing LTßR potentiated the infectivity of ZIKV both in vitro and in vivo. This study provides deeper insight into the transmission of mosquito-borne diseases in nature and supports the therapeutic potential of inhibiting the action of LTRIN to disrupt ZIKV transmission.


Asunto(s)
Aedes/virología , Proteínas de Insectos/metabolismo , Saliva/metabolismo , Infección por el Virus Zika/transmisión , Virus Zika/patogenicidad , Animales , Humanos , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Ratones , Mosquitos Vectores/química , Mosquitos Vectores/inmunología , Mosquitos Vectores/metabolismo , Saliva/química
13.
Cytokine ; 101: 39-47, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27623349

RESUMEN

Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.


Asunto(s)
Antivirales/inmunología , Tejido Linfoide/fisiología , Linfotoxina-alfa/inmunología , Virosis/inmunología , Animales , Autoinmunidad , Homeostasis/inmunología , Humanos , Inflamación , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Tejido Linfoide/inmunología , Receptor beta de Linfotoxina/inmunología , Linfotoxina-alfa/efectos de los fármacos , Linfotoxina-alfa/genética , Ratones , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Virosis/tratamiento farmacológico , Virosis/fisiopatología
14.
Cell Mol Immunol ; 15(7): 697-709, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28579615

RESUMEN

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.


Asunto(s)
Inmunidad Innata , Linfocitos/inmunología , Heterotrímero de Linfotoxina alfa1 y beta2/inmunología , Receptor beta de Linfotoxina/inmunología , Microbiota/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Linfocitos/patología , Heterotrímero de Linfotoxina alfa1 y beta2/genética , Receptor beta de Linfotoxina/genética , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/genética
15.
Cancer Immunol Res ; 5(11): 1005-1015, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28947544

RESUMEN

T-cell infiltration into tumors represents a critical bottleneck for immune-mediated control of cancer. We previously showed that this bottleneck can be overcome by depleting immunosuppressive Foxp3+ regulatory T cells (Tregs), a process that can increase frequencies of tumor-infiltrating lymphocytes through promoting the development of specialized portals for lymphocyte entry, namely high endothelial venules (HEVs). In this paper, we used a carcinogen-induced tumor model that allows for coevolution of the tumor microenvironment and the immune response to demonstrate that Treg depletion not only results in widespread disruption to HEV networks in lymph nodes (LNs) but also activates CD8+ T cells, which then drive intratumoral HEV development. Formation of these vessels contrasts with ontogenic HEV development in LNs in that the process is dependent on the TNF receptor and independent of lymphotoxin ß receptor-mediated signaling. These intratumoral HEVs do not express the chemokine CCL21, revealing a previously undescribed intratumoral blood vessel phenotype. We propose a model where Treg depletion enables a self-amplifying loop of T-cell activation, which promotes HEV development, T-cell infiltration, and ultimately, tumor destruction. The findings point to a need to test for HEV development as part of ongoing clinical studies in patients with cancer. Cancer Immunol Res; 5(11); 1005-15. ©2017 AACR.


Asunto(s)
Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Dendríticas/inmunología , Endotelio Vascular/inmunología , Depleción Linfocítica , Linfocitos Infiltrantes de Tumor/inmunología , Receptor beta de Linfotoxina/inmunología , Metilcolantreno , Ratones , Neoplasias/inducido químicamente , Receptores del Factor de Necrosis Tumoral/inmunología
16.
J Exp Med ; 214(11): 3183-3195, 2017 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-28830910

RESUMEN

During αßT cell development, the thymus medulla represents an essential microenvironment for T cell tolerance. This functional specialization is attributed to its typical organized topology consisting of a branching structure that contains medullary thymic epithelial cell (mTEC) networks to support negative selection and Foxp3+ T-regulatory cell (T-reg) development. Here, by performing TEC-specific deletion of the thymus medulla regulator lymphotoxin ß receptor (LTßR), we show that thymic tolerance mechanisms operate independently of LTßR-mediated mTEC development and organization. Consistent with this, mTECs continue to express Fezf2 and Aire, regulators of intrathymic self-antigens, and support T-reg development despite loss of LTßR-mediated medulla organogenesis. Moreover, we demonstrate that LTßR controls thymic tolerance by regulating the frequency and makeup of intrathymic dendritic cells (DCs) required for effective thymocyte negative selection. In all, our study demonstrates that thymus medulla specialization for thymic tolerance segregates from medulla organogenesis and instead involves LTßR-mediated regulation of the thymic DC pool.


Asunto(s)
Tolerancia Central/inmunología , Células Epiteliales/inmunología , Receptor beta de Linfotoxina/inmunología , Timo/inmunología , Animales , Autoantígenos/inmunología , Tolerancia Central/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Timo/embriología , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo , Proteína AIRE
17.
J Immunol ; 197(7): 2665-72, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27549174

RESUMEN

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


Asunto(s)
Movimiento Celular , Receptor beta de Linfotoxina/inmunología , Células Madre/citología , Linfocitos T/citología , Timo/citología , Animales , Receptor beta de Linfotoxina/deficiencia , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre/inmunología , Linfocitos T/inmunología , Timo/inmunología
18.
Nat Commun ; 7: 12021, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27323847

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Rechazo de Injerto/inmunología , Trasplante de Islotes Pancreáticos , Heterotrímero de Linfotoxina alfa1 y beta2/inmunología , Receptor beta de Linfotoxina/inmunología , Linfocitos T Reguladores/inmunología , Migración Transendotelial y Transepitelial/inmunología , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/mortalidad , Células Endoteliales/citología , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Supervivencia de Injerto/genética , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/cirugía , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Vasos Linfáticos/citología , Vasos Linfáticos/inmunología , Heterotrímero de Linfotoxina alfa1 y beta2/genética , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Transducción de Señal , Análisis de Supervivencia , Linfocitos T Reguladores/citología , Trasplante Homólogo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
19.
Science ; 352(6287): aaf4822, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-27174992

RESUMEN

Immunoglobulin A (IgA) induction primarily occurs in intestinal Peyer's patches (PPs). However, the cellular interactions necessary for IgA class switching are poorly defined. Here we show that in mice, activated B cells use the chemokine receptor CCR6 to access the subepithelial dome (SED) of PPs. There, B cells undergo prolonged interactions with SED dendritic cells (DCs). PP IgA class switching requires innate lymphoid cells, which promote lymphotoxin-ß receptor (LTßR)-dependent maintenance of DCs. PP DCs augment IgA production by integrin αvß8-mediated activation of transforming growth factor-ß (TGFß). In mice where B cells cannot access the SED, IgA responses against oral antigen and gut commensals are impaired. These studies establish the PP SED as a niche supporting DC-B cell interactions needed for TGFß activation and induction of mucosal IgA responses.


Asunto(s)
Linfocitos B/inmunología , Células Dendríticas/inmunología , Inmunoglobulina A Secretora/biosíntesis , Cambio de Clase de Inmunoglobulina , Ganglios Linfáticos Agregados/inmunología , Receptores CCR6/inmunología , Animales , Comunicación Celular/inmunología , Movimiento Celular/inmunología , Inmunoglobulina A Secretora/genética , Integrinas/inmunología , Mucosa Intestinal/inmunología , Activación de Linfocitos , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Mutantes , Receptores CCR6/genética
20.
Cancer Cell ; 29(3): 285-296, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26977880

RESUMEN

Immune checkpoint blockade therapies fail to induce responses in the majority of cancer patients, so how to increase the objective response rate becomes an urgent challenge. Here, we demonstrate that sufficient T cell infiltration in tumor tissues is a prerequisite for response to PD-L1 blockade. Targeting tumors with tumor necrosis factor superfamily member LIGHT activates lymphotoxin ß-receptor signaling, leading to the production of chemokines that recruit massive numbers of T cells. Furthermore, targeting non-T cell-inflamed tumor tissues by antibody-guided LIGHT creates a T cell-inflamed microenvironment and overcomes tumor resistance to checkpoint blockade. Our data indicate that targeting LIGHT might be a potent strategy to increase the responses to checkpoint blockades and other immunotherapies in non-T cell-inflamed tumors.


Asunto(s)
Antineoplásicos/inmunología , Antígeno B7-H1/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Secuencia de Aminoácidos , Animales , Resistencia a Antineoplásicos/inmunología , Humanos , Inmunoterapia/métodos , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA