Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
J Autoimmun ; 144: 103177, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38368767

RESUMEN

Psoriasis (PS) and atopic dermatitis (AD) are common skin inflammatory diseases characterized by hyper-responsive keratinocytes. Although, some cytokines have been suggested to be specific for each disease, other cytokines might be central to both diseases. Here, we show that Tumor necrosis factor superfamily member 14 (TNFSF14), known as LIGHT, is required for experimental PS, similar to its requirement in experimental AD. Mice devoid of LIGHT, or deletion of either of its receptors, lymphotoxin ß receptor (LTßR) and herpesvirus entry mediator (HVEM), in keratinocytes, were protected from developing imiquimod-induced psoriatic features, including epidermal thickening and hyperplasia, and expression of PS-related genes. Correspondingly, in single cell RNA-seq analysis of PS patient biopsies, LTßR transcripts were found strongly expressed with HVEM in keratinocytes, and LIGHT was upregulated in T cells. Similar transcript expression profiles were also seen in AD biopsies, and LTßR deletion in keratinocytes also protected mice from allergen-induced AD features. Moreover, in vitro, LIGHT upregulated a broad spectrum of genes in human keratinocytes that are clinical features of both PS and AD skin lesions. Our data suggest that agents blocking LIGHT activity might be useful for therapeutic intervention in PS as well as in AD.


Asunto(s)
Dermatitis Atópica , Psoriasis , Humanos , Ratones , Animales , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Dermatitis Atópica/genética , Dermatitis Atópica/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Queratinocitos/metabolismo , Citocinas/metabolismo , Psoriasis/genética , Psoriasis/metabolismo , Inflamación/metabolismo
2.
Elife ; 122023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36912771

RESUMEN

Acute lymphoblastic and myeloblastic leukemias (ALL and AML) have been known to modify the bone marrow microenvironment and disrupt non-malignant hematopoiesis. However, the molecular mechanisms driving these alterations remain poorly defined. Using mouse models of ALL and AML, here we show that leukemic cells turn off lymphopoiesis and erythropoiesis shortly after colonizing the bone marrow. ALL and AML cells express lymphotoxin α1ß2 and activate lymphotoxin beta receptor (LTßR) signaling in mesenchymal stem cells (MSCs), which turns off IL7 production and prevents non-malignant lymphopoiesis. We show that the DNA damage response pathway and CXCR4 signaling promote lymphotoxin α1ß2 expression in leukemic cells. Genetic or pharmacological disruption of LTßR signaling in MSCs restores lymphopoiesis but not erythropoiesis, reduces leukemic cell growth, and significantly extends the survival of transplant recipients. Similarly, CXCR4 blocking also prevents leukemia-induced IL7 downregulation and inhibits leukemia growth. These studies demonstrate that acute leukemias exploit physiological mechanisms governing hematopoietic output as a strategy for gaining competitive advantage.


Asunto(s)
Leucemia Mieloide Aguda , Células Madre Mesenquimatosas , Animales , Ratones , Leucemia Mieloide Aguda/patología , Receptor beta de Linfotoxina/metabolismo , Interleucina-7/metabolismo , Linfopoyesis , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microambiente Tumoral
3.
EMBO Rep ; 24(3): e54228, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36633157

RESUMEN

Estrogen is a disease-modifying factor in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) via estrogen receptor alpha (ERα). However, the mechanisms by which ERα signaling contributes to changes in disease pathogenesis have not been completely elucidated. Here, we demonstrate that ERα deletion in dendritic cells (DCs) of mice induces severe neurodegeneration in the central nervous system in a mouse EAE model and resistance to interferon beta (IFNß), a first-line MS treatment. Estrogen synthesized by extragonadal sources is crucial for controlling disease phenotypes. Mechanistically, activated ERα directly interacts with TRAF3, a TLR4 downstream signaling molecule, to degrade TRAF3 via ubiquitination, resulting in reduced IRF3 nuclear translocation and transcription of membrane lymphotoxin (mLT) and IFNß components. Diminished ERα signaling in DCs generates neurotoxic effector CD4+ T cells via mLT-lymphotoxin beta receptor (LTßR) signaling. Lymphotoxin beta receptor antagonist abolished EAE disease symptoms in the DC-specific ERα-deficient mice. These findings indicate that estrogen derived from extragonadal sources, such as lymph nodes, controls TRAF3-mediated cytokine production in DCs to modulate the EAE disease phenotype.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Receptor alfa de Estrógeno , Ratones , Animales , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Estrógenos/farmacología , Fenotipo , Células Dendríticas/metabolismo , Ratones Endogámicos C57BL
4.
Clin Investig Arterioscler ; 35(1): 1-11, 2023.
Artículo en Inglés, Español | MEDLINE | ID: mdl-35738949

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (VSMCs) undergo a phenotypic-switching process during the generation of unstable atheroma plaques. In this investigation, the potential implication of the tumor necrosis factor superfamily (TNFSF) ligands, in the gene expression signature associated with VSMC plasticity was studied. MATERIAL AND METHODS: Human aortic (ha)VSMCs were obtained commercially and treated with the cytokine TNFSF14, also called LIGHT, the lymphotoxin alpha (LTα), the heterotrimer LTα1ß2 or with vehicle for 72h. The effect of the different treatments on gene expression was analyzed by quantitative PCR and included the study of genes associated with myofibroblast-like cell function, osteochondrogenesis, pluripotency, lymphorganogenesis and macrophage-like cell function. RESULTS: HaVSMCs displayed a change in myofibroblast-like cell genes which consisted in reduced COL1A1 and TGFB1 mRNA levels when treated with LTα or LIGHT and with augmented MMP9 expression levels when treated with LTα. LTα and LIGHT treatments also diminished the expression of genes associated with osteochondrogenesis and pluripotency SOX9, CKIT, and KLF4. By contrary, all the above genes were no affected by the treatment with the trimer LTα1ß2. In addition, haVSMC treatment with LTα, LTα1ß2 and LIGHT altered lymphorganogenic cytokine gene expression which consisted of augmented CCL20 and CCL21 mRNA levels by LTα and a reduction in the gene expression of CCL21 and CXCL13 by LIGHT and LTα1ß2 respectively. Neither, LTα or LIGHT or LTα1ß2 treatments affected the expression of macrophage-like cell markers in haVSMC. CONCLUSIONS: Altogether, indicates that the TNFSF ligands through their interconnected network of signaling, are important in the preservation of VSMC identity against the acquisition of a genetic expression signature compatible with functional cellular plasticity.


Asunto(s)
Receptor beta de Linfotoxina , Músculo Liso Vascular , Humanos , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Músculo Liso Vascular/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfotoxina-alfa/genética , Linfotoxina-alfa/metabolismo , Citocinas , ARN Mensajero/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(40): e2208436119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161919

RESUMEN

Engineered regulatory T cell (Treg cell) therapy is a promising strategy to treat patients suffering from inflammatory diseases, autoimmunity, and transplant rejection. However, in many cases, disease-related antigens that can be targeted by Treg cells are not available. In this study, we introduce a class of synthetic biosensors, named artificial immune receptors (AIRs), for murine and human Treg cells. AIRs consist of three domains: (a) extracellular binding domain of a tumor necrosis factor (TNF)-receptor superfamily member, (b) intracellular costimulatory signaling domain of CD28, and (c) T cell receptor signaling domain of CD3-ζ chain. These AIR receptors equip Treg cells with an inflammation-sensing machinery and translate this environmental information into a CD3-ζ chain-dependent TCR-activation program. Different AIRs were generated, recognizing the inflammatory ligands of the TNF-receptor superfamily, including LIGHT, TNFα, and TNF-like ligand 1A (TL1A), leading to activation, differentiation, and proliferation of AIR-Treg cells. In a graft-versus-host disease model, Treg cells expressing lymphotoxin ß receptor-AIR, which can be activated by the ligand LIGHT, protect significantly better than control Treg cells. Expression and signaling of the corresponding human AIR in human Treg cells prove that this concept can be translated. Engineering Treg cells that target inflammatory ligands leading to TCR signaling and activation might be used as a Treg cell-based therapy approach for a broad range of inflammation-driven diseases.


Asunto(s)
Técnicas Biosensibles , Ingeniería Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Inflamación , Linfocitos T Reguladores , Animales , Antígenos CD28/metabolismo , Humanos , Inflamación/terapia , Ligandos , Receptor beta de Linfotoxina/metabolismo , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/trasplante , Factor de Necrosis Tumoral alfa
6.
FEBS Lett ; 596(20): 2659-2667, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35953458

RESUMEN

Follicular dendritic cells (FDCs) play a crucial role in generating high-affinity antibody-producing B cells during the germinal center (GC) reaction. Herein, we analysed the altered gene expression profile of a mouse FDC line, FL-Y, following lymphotoxin ß receptor stimulation, and observed increased Slam-family member 8 (Slamf8) mRNA expression. Forced Slamf8 expression and SLAMF8-Fc addition enhanced the ability of FL-Y cells to induce FDC-induced monocytic cell (FDMC) differentiation. FDMCs accelerated GC-phenotype proliferation in cultured B cells, suggesting that they are capable of promoting GC responses. Furthermore, a pulldown assay showed that SLAMF8-Fc could bind to SLAMF8-His. Overall, the homophilic interaction of SLAMF8 promotes FDMC differentiation and SLAMF8 might act as a novel regulator of GC responses by regulating FDMC differentiation.


Asunto(s)
Células Dendríticas Foliculares , Receptor beta de Linfotoxina , Ratones , Animales , Células Dendríticas Foliculares/metabolismo , Receptor beta de Linfotoxina/metabolismo , Centro Germinal/metabolismo , Linfocitos B/metabolismo , Diferenciación Celular/genética , ARN Mensajero/metabolismo , Células Dendríticas
7.
PLoS One ; 17(8): e0270907, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35925983

RESUMEN

Multiple myeloma (MM), a malignancy of plasma cells (PCs), has diverse genetic underpinnings and in rare cases these include amplification of the lymphotoxin b receptor (Ltbr) locus. LTßR has well defined roles in supporting lymphoid tissue development and function through actions in stromal and myeloid cells, but whether it is functional in PCs is unknown. Here we showed that Ltbr mRNA was upregulated in mouse PCs compared to follicular B cells, but deficiency in the receptor did not cause a reduction in PC responses to a T-dependent or T-independent immunogen. However, LTßR overexpression (OE) enhanced PC formation in vitro after LPS or anti-CD40 stimulation. In vivo, LTßR OE led to increased antigen-specific splenic and bone marrow (BM) plasma cells responses. LTßR OE PCs had increased expression of Nfkb2 and of the NF-kB target genes Bcl2 and Mcl1, factors involved in the formation of long-lived BM PCs. Our findings suggest a pathway by which Ltbr gene amplifications may contribute to MM development through increased NF-kB activity and induction of an anti-apoptotic transcriptional program.


Asunto(s)
FN-kappa B , Células Plasmáticas , Animales , Linfocitos B/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Células Plasmáticas/metabolismo , Bazo/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35031565

RESUMEN

CD169+ macrophages reside in lymph node (LN) and spleen and play an important role in the immune defense against pathogens. As resident macrophages, they are responsive to environmental cues to shape their tissue-specific identity. We have previously shown that LN CD169+ macrophages require RANKL for formation of their niche and their differentiation. Here, we demonstrate that they are also dependent on direct lymphotoxin beta (LTß) receptor (R) signaling. In the absence or the reduced expression of either RANK or LTßR, their differentiation is perturbed, generating myeloid cells expressing SIGN-R1 in LNs. Conditions of combined haploinsufficiencies of RANK and LTßR revealed that both receptors contribute equally to LN CD169+ macrophage differentiation. In the spleen, the Cd169-directed ablation of either receptor results in a selective loss of marginal metallophilic macrophages (MMMs). Using a RANKL reporter mouse, we identify splenic marginal zone stromal cells as a source of RANKL and demonstrate that it participates in MMM differentiation. The loss of MMMs had no effect on the splenic B cell compartments but compromised viral capture and the expansion of virus-specific CD8+ T cells. Taken together, the data provide evidence that CD169+ macrophage differentiation in LN and spleen requires dual signals from LTßR and RANK with implications for the immune response.


Asunto(s)
Ganglios Linfáticos/inmunología , Receptor beta de Linfotoxina/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal , Bazo/inmunología , Linfocitos B/inmunología , Ligando RANK/metabolismo , Células del Estroma/metabolismo
9.
Mucosal Immunol ; 15(2): 327-337, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34903876

RESUMEN

Fibroblasts mediate tissue remodeling in eosinophilic esophagitis (EoE), a chronic allergen-driven inflammatory pathology. Diverse fibroblast subtypes with homeostasis-regulating or inflammatory profiles have been recognized in various tissues, but which mediators induce these alternate differentiation states remain largely unknown. We recently identified that TNFSF14/LIGHT promotes an inflammatory esophageal fibroblast in vitro. Herein we used esophageal biopsies and primary fibroblasts to investigate the role of the LIGHT receptors, herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR), and their downstream activated pathways, in EoE. In addition to promoting inflammatory gene expression, LIGHT down-regulated homeostatic factors including WNTs, BMPs and type 3 semaphorins. In vivo, WNT2B+ fibroblasts were decreased while ICAM-1+ and IL-34+ fibroblasts were expanded in EoE, suggesting that a LIGHT-driven gene signature was imprinted in EoE versus normal esophageal fibroblasts. HVEM and LTßR overexpression and deficiency experiments demonstrated that HVEM regulates a limited subset of LIGHT targets, whereas LTßR controls all transcriptional effects. Pharmacologic blockade of the non-canonical NIK/p100/p52-mediated NF-κB pathway potently silenced LIGHT's transcriptional effects, with a lesser role found for p65 canonical NF-κB. Collectively, our results show that LIGHT promotes differentiation of esophageal fibroblasts toward an inflammatory phenotype and represses homeostatic gene expression via a LTßR-NIK-p52 NF-κB dominant pathway.


Asunto(s)
Esófago , Inflamación , Transcriptoma , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Esófago/metabolismo , Fibroblastos/metabolismo , Homeostasis , Humanos , Inflamación/genética , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
10.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638990

RESUMEN

Obesity is one of the most prevalent metabolic diseases in the Western world and correlates directly with glucose intolerance and insulin resistance, often culminating in Type 2 Diabetes (T2D). Importantly, our team has recently shown that the TNF superfamily (TNFSF) member protein, TNFSF14, has been reported to protect against high fat diet induced obesity and pre-diabetes. We hypothesized that mimics of TNFSF14 may therefore be valuable as anti-diabetic agents. In this study, we use in silico approaches to identify key regions of TNFSF14 responsible for binding to the Herpes virus entry mediator and Lymphotoxin ß receptor. In vitro evaluation of a selection of optimised peptides identified six potentially therapeutic TNFSF14 peptides. We report that these peptides increased insulin and fatty acid oxidation signalling in skeletal muscle cells. We then selected one of these promising peptides to determine the efficacy to promote metabolic benefits in vivo. Importantly, the TNFSF14 peptide 7 reduced high fat diet-induced glucose intolerance, insulin resistance and hyperinsulinemia in a mouse model of obesity. In addition, we highlight that the TNFSF14 peptide 7 resulted in a marked reduction in liver steatosis and a concomitant increase in phospho-AMPK signalling. We conclude that TNFSF14-derived molecules positively regulate glucose homeostasis and lipid metabolism and may therefore open a completely novel therapeutic pathway for treating obesity and T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Péptidos/administración & dosificación , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/administración & dosificación , Animales , Sitios de Unión , Glucemia/metabolismo , Simulación por Computador , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hiperinsulinismo/tratamiento farmacológico , Hiperinsulinismo/metabolismo , Hipoglucemiantes/síntesis química , Resistencia a la Insulina , Receptor beta de Linfotoxina/química , Receptor beta de Linfotoxina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Péptidos/síntesis química , Miembro 14 de Receptores del Factor de Necrosis Tumoral/química , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/química , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
11.
Front Immunol ; 12: 707404, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276703

RESUMEN

Thymic blood vessels at the perivascular space (PVS) are the critical site for both homing of hematopoietic progenitor cells (HPCs) and egress of mature thymocytes. It has been intriguing how different opposite migrations can happen in the same place. A subset of specialized thymic portal endothelial cells (TPECs) associated with PVS has been identified to function as the entry site for HPCs. However, the cellular basis and mechanism underlying egress of mature thymocytes has not been well defined. In this study, using various conventional and conditional gene-deficient mouse models, we first confirmed the role of endothelial lymphotoxin beta receptor (LTßR) for thymic egress and ruled out the role of LTßR from epithelial cells or dendritic cells. In addition, we found that T cell-derived ligands lymphotoxin (LT) and LIGHT are required for thymic egress, suggesting a crosstalk between T cells and endothelial cells (ECs) for thymic egress control. Furthermore, immunofluorescence staining analysis interestingly showed that TPECs are also the exit site for mature thymocytes. Single-cell transcriptomic analysis of thymic endothelial cells suggested that TPECs are heterogeneous and can be further divided into two subsets depending on BST-1 expression level. Importantly, BST-1hi population is associated with thymic egressing thymocytes while BST-1lo/- population is associated with HPC settling. Thus, we have defined a LT/LIGHT-LTßR signaling-mediated cellular crosstalk regulating thymic egress and uncovered distinct subsets of TPECs controlling thymic homing and egress, respectively.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Receptor beta de Linfotoxina/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Animales , Linfotoxina-alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Timo/citología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
12.
Cell Rep ; 36(3): 109422, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289373

RESUMEN

Tumor-associated tertiary lymphoid structures (TA-TLS) are associated with enhanced patient survival and responsiveness to cancer therapies, but the mechanisms underlying their development are unknown. We show here that TA-TLS development in murine melanoma is orchestrated by cancer-associated fibroblasts (CAF) with characteristics of lymphoid tissue organizer cells that are induced by tumor necrosis factor receptor signaling. CAF organization into reticular networks is mediated by CD8 T cells, while CAF accumulation and TA-TLS expansion depend on CXCL13-mediated recruitment of B cells expressing lymphotoxin-α1ß2. Some of these elements are also overrepresented in human TA-TLS. Additionally, we demonstrate that immunotherapy induces more and larger TA-TLS that are more often organized with discrete T and B cell zones, and that TA-TLS presence, number, and size are correlated with reduced tumor size and overall response to checkpoint immunotherapy. This work provides a platform for manipulating TA-TLS development as a cancer immunotherapy strategy.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Neoplasias/inmunología , Neoplasias/patología , Estructuras Linfoides Terciarias/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Proliferación Celular , Humanos , Inmunoterapia , Activación de Linfocitos/inmunología , Receptor beta de Linfotoxina/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Neoplasias/terapia , Peritoneo/patología , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal
13.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34155144

RESUMEN

Aberrant inflammation, such as that associated with inflammatory bowel disease (IBD), is fueled by the inordinate activity of RelA/NF-κB factors. As such, the canonical NF-κB module mediates controlled nuclear activation of RelA dimers from the latent cytoplasmic complexes. What provokes pathological RelA activity in the colitogenic gut remains unclear. The noncanonical NF-κB pathway typically promotes immune organogenesis involving Nfkb2 gene products. Because NF-κB pathways are intertwined, we asked whether noncanonical signaling aggravated inflammatory RelA activity. Our investigation revealed frequent engagement of the noncanonical pathway in human IBD. In a mouse model of experimental colitis, we established that Nfkb2-mediated regulations escalated the RelA-driven proinflammatory gene response in intestinal epithelial cells, exacerbating the infiltration of inflammatory cells and colon pathologies. Our mechanistic studies clarified that cell-autonomous Nfkb2 signaling supplemented latent NF-κB dimers, leading to a hyperactive canonical RelA response in the inflamed colon. In sum, the regulation of latent NF-κB dimers appears to link noncanonical Nfkb2 signaling to RelA-driven inflammatory pathologies and may provide for therapeutic targets.


Asunto(s)
Inflamación/patología , Intestinos/patología , Subunidad p52 de NF-kappa B/metabolismo , FN-kappa B/metabolismo , Multimerización de Proteína , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Animales , Colitis/metabolismo , Colitis/patología , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Receptor beta de Linfotoxina/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Subunidad p52 de NF-kappa B/deficiencia , Células del Estroma/metabolismo
14.
Cells ; 10(4)2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805271

RESUMEN

The pleiotropic functions of lymphotoxin (LT)ß receptor (LTßR) signaling are linked to the control of secondary lymphoid organ development and structural maintenance, inflammatory or autoimmune disorders, and carcinogenesis. Recently, LTßR signaling in endothelial cells has been revealed to regulate immune cell migration. Signaling through LTßR is comprised of both the canonical and non-canonical-nuclear factor κB (NF-κB) pathways, which induce chemokines, cytokines, and cell adhesion molecules. Here, we focus on the novel functions of LTßR signaling in lymphatic endothelial cells for migration of regulatory T cells (Tregs), and specific targeting of LTßR signaling for potential therapeutics in transplantation and cancer patient survival.


Asunto(s)
Movimiento Celular , Leucocitos/citología , Leucocitos/metabolismo , Sistema Linfático/citología , Receptor beta de Linfotoxina/metabolismo , Transducción de Señal , Células Endoteliales/metabolismo , Humanos
15.
Infect Immun ; 89(6)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33753412

RESUMEN

The lymphotoxin ß receptor (LTßR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTßR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTßR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTßR-deficient (LTßR-/-) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTßR-/- mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTßR in cytokine regulation and adaptive immune responses to control T. gondii.


Asunto(s)
Inmunidad Adaptativa , Interacciones Huésped-Parásitos/inmunología , Inmunidad Innata , Receptor beta de Linfotoxina/metabolismo , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Toxoplasmosis/metabolismo , Animales , Modelos Animales de Enfermedad , Receptor beta de Linfotoxina/genética , Ratones , Ratones Noqueados , Toxoplasmosis/parasitología
16.
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
17.
Mucosal Immunol ; 14(3): 679-690, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33568785

RESUMEN

Inflammatory bowel disease is characterized by an exacerbated intestinal immune response, but the critical mechanisms regulating immune activation remain incompletely understood. We previously reported that the TNF-superfamily molecule TNFSF14 (LIGHT) is required for preventing severe disease in mouse models of colitis. In addition, deletion of lymphotoxin beta receptor (LTßR), which binds LIGHT, also led to aggravated colitis pathogenesis. Here, we aimed to determine the cell type(s) requiring LTßR and the mechanism critical for exacerbation of colitis. Specific deletion of LTßR in neutrophils (LTßRΔN), but not in several other cell types, was sufficient to induce aggravated colitis and colonic neutrophil accumulation. Mechanistically, RNA-Seq analysis revealed LIGHT-induced suppression of cellular metabolism, and mitochondrial function, that was dependent on LTßR. Functional studies confirmed increased mitochondrial mass and activity, associated with excessive mitochondrial ROS production and elevated glycolysis at steady-state and during colitis. Targeting these metabolic changes rescued exacerbated disease severity. Our results demonstrate that LIGHT signals to LTßR on neutrophils to suppress metabolic activation and thereby prevents exacerbated immune pathogenesis during colitis.


Asunto(s)
Colitis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Receptor beta de Linfotoxina/metabolismo , Mitocondrias/metabolismo , Neutrófilos/metabolismo , Activación Metabólica , Animales , Sulfato de Dextran , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Receptor beta de Linfotoxina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
18.
Nat Commun ; 12(1): 1065, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594067

RESUMEN

The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-ß receptor (LTßR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTßR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTßR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTßR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTßR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Receptor beta de Linfotoxina/metabolismo , Células Madre Neoplásicas/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Antígenos CD34/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Daño del ADN , Fluorouracilo/farmacología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
19.
PLoS One ; 16(2): e0247368, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606781

RESUMEN

Osteoporosis is a progressive systemic skeletal disease associated with decreased bone mineral density and deterioration of bone quality, and it affects millions of people worldwide. Currently, it is treated mainly using antiresorptive and osteoanabolic agents. However, these drugs have severe adverse effects. Cell replacement therapy using mesenchymal stem cells (MSCs) could serve as a treatment strategy for osteoporosis in the future. LIGHT (HVEM-L, TNFSF14, or CD258) is a member of the tumor necrosis factor superfamily. However, the effect of recombinant LIGHT (rhLIGHT) on osteogenesis in human bone marrow-derived MSCs (hBM-MSCs) is unknown. Therefore, we monitored the effects of LIGHT on osteogenesis of hBM-MSCs. Lymphotoxin-ß receptor (LTßR), which is a LIGHT receptor, was constitutively expressed on the surface of hBM-MSCs. After rhLIGHT treatment, calcium and phosphate deposition in hBM-MSCs, stained by Alizarin red and von Kossa, respectively, significantly increased. We performed quantitative real-time polymerase chain reaction to examine the expressions of osteoprogenitor markers (RUNX2/CBFA1 and collagen I alpha 1) and osteoblast markers (alkaline phosphatase, osterix/Sp7, and osteocalcin) and immunoblotting to assess the underlying biological mechanisms following rhLIGHT treatment. We found that rhLIGHT treatment enhanced von Kossa- and Alizarin red-positive hBM-MSCs and induced the expression of diverse differentiation markers of osteogenesis in a dose-dependent manner. WNT/ß-catenin pathway activation strongly mediated rhLIGHT-induced osteogenesis of hBM-MSCs, accelerating the differentiation of hBM-MSCs into osteocytes. In conclusion, the interaction between LIGHT and LTßR enhances osteogenesis of hBM-MSCs. Therefore, LIGHT might play an important role in stem cell therapy.


Asunto(s)
Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Marcadores Genéticos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Proteínas Recombinantes/farmacología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vía de Señalización Wnt/efectos de los fármacos
20.
J Cell Physiol ; 236(6): 4725-4737, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33269476

RESUMEN

The differentiation of mature medullary thymic epithelial cells (mTECs) is critical for the induction of central immune tolerance. Although the critical effect of mechanistic target of rapamycin complex 1 (mTORC1) in shaping mTEC differentiation has been studied, the regulatory role of mTORC2 in the differentiation and maturation of mTECs is poorly understood. We herein reported that TEC-specific ablation of a rapamycin-insensitive companion of mTOR (RICTOR), a key component of mTORC2, significantly decreased the thymus size and weight, the total cell number of TECs, and the cell number of mTECs with a smaller degree of reduced cortical thymic epithelial cells. Interestingly, RICTOR deficiency significantly accelerated the mTEC maturation process, as indicated by the increased ratios of mature mTECs (MHCIIhi , CD80+ , and Aire+ ) to immature mTECs (MHCIIlo , CD80- , and Aire- ) in Rictor-deficient mice. The RNA-sequencing assays showed that the upregulated nuclear factor-κB (NF-κB) signaling pathway in Rictor-deficient mTECs was one of the obviously altered pathways compared with wild-type mTECs. Our studies further showed that Rictor-deficient mTECs exhibited upregulated expression of receptor activator of NF-κB (RANK) and lymphotoxin ß receptor (LTßR), as well as increased activity of canonical and noncanonical NF-κB signaling pathways as determined by ImageStream and Simple Western. Finally, our results showed that inhibition of NF-κB signaling pathways could partially reverse the accelerated maturation of mTECs in Rictor conditional KO mice. Thus, mTORC2 negatively controls the kinetics of the mTEC maturation process by inhibiting the LTßR/RANK-NF-κB signal axis.


Asunto(s)
Diferenciación Celular , Células Epiteliales/enzimología , Receptor beta de Linfotoxina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , FN-kappa B/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Timo/enzimología , Animales , Células Epiteliales/patología , Regulación de la Expresión Génica , Cinética , Receptor beta de Linfotoxina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ratones Noqueados , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal , Timocitos/enzimología , Timocitos/patología , Timo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...