Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.271
Filtrar
Más filtros

Intervalo de año de publicación
1.
PLoS Biol ; 18(11): e3000946, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33253145

RESUMEN

Inflammation of the central nervous system (CNS) induces endothelial blood-brain barrier (BBB) opening as well as the formation of a tight junction barrier between reactive astrocytes at the Glia Limitans. We hypothesized that the CNS parenchyma may acquire protection from the reactive astrocytic Glia Limitans not only during neuroinflammation but also when BBB integrity is compromised in the resting state. Previous studies found that astrocyte-derived Sonic hedgehog (SHH) stabilizes the BBB during CNS inflammatory disease, while endothelial-derived desert hedgehog (DHH) is expressed at the BBB under resting conditions. Here, we investigated the effects of endothelial Dhh on the integrity of the BBB and Glia Limitans. We first characterized DHH expression within endothelial cells at the BBB, then demonstrated that DHH is down-regulated during experimental autoimmune encephalomyelitis (EAE). Using a mouse model in which endothelial Dhh is inducibly deleted, we found that endothelial Dhh both opens the BBB via the modulation of forkhead box O1 (FoxO1) transcriptional activity and induces a tight junctional barrier at the Glia Limitans. We confirmed the relevance of this glial barrier system in human multiple sclerosis active lesions. These results provide evidence for the novel concept of "chronic neuroinflammatory tolerance" in which BBB opening in the resting state is sufficient to stimulate a protective barrier at the Glia Limitans that limits the severity of subsequent neuroinflammatory disease. In summary, genetic disruption of the BBB generates endothelial signals that drive the formation under resting conditions of a secondary barrier at the Glia Limitans with protective effects against subsequent CNS inflammation. The concept of a reciprocally regulated CNS double barrier system has implications for treatment strategies in both the acute and chronic phases of multiple sclerosis pathophysiology.


Asunto(s)
Barrera Hematoencefálica/fisiología , Barrera Hematoencefálica/fisiopatología , Uniones Adherentes/patología , Uniones Adherentes/fisiología , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Astrocitos/patología , Astrocitos/fisiología , Cadherinas/genética , Cadherinas/fisiología , Permeabilidad Capilar/genética , Permeabilidad Capilar/fisiología , Claudina-5/genética , Claudina-5/fisiología , Regulación hacia Abajo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Células Endoteliales/patología , Células Endoteliales/fisiología , Femenino , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Neuroglía/patología , Neuroglía/fisiología , Uniones Estrechas/patología , Uniones Estrechas/fisiología
2.
J Biol Chem ; 296: 100426, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33609526

RESUMEN

Birt-Hogg-Dubé (BHD) syndrome is a multiorgan disorder caused by inactivation of the folliculin (FLCN) protein. Previously, we identified FLCN as a binding protein of Rab11A, a key regulator of the endocytic recycling pathway. This finding implies that the abnormal localization of specific proteins whose transport requires the FLCN-Rab11A complex may contribute to BHD. Here, we used human kidney-derived HEK293 cells as a model, and we report that FLCN promotes the binding of Rab11A with transferrin receptor 1 (TfR1), which is required for iron uptake through continuous trafficking between the cell surface and the cytoplasm. Loss of FLCN attenuated the Rab11A-TfR1 interaction, resulting in delayed recycling transport of TfR1. This delay caused an iron deficiency condition that induced hypoxia-inducible factor (HIF) activity, which was reversed by iron supplementation. In a Drosophila model of BHD syndrome, we further demonstrated that the phenotype of BHD mutant larvae was substantially rescued by an iron-rich diet. These findings reveal a conserved function of FLCN in iron metabolism and may help to elucidate the mechanisms driving BHD syndrome.


Asunto(s)
Antígenos CD/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Transferrina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Síndrome de Birt-Hogg-Dubé/metabolismo , Síndrome de Birt-Hogg-Dubé/fisiopatología , Citoplasma/metabolismo , Proteínas de Drosophila , Drosophila melanogaster , Células HEK293 , Homeostasis , Humanos , Hierro/metabolismo , Modelos Animales , Proteínas Proto-Oncogénicas/fisiología , Receptores de Transferrina/genética , Receptores de Transferrina/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Unión al GTP rab/metabolismo
3.
J Biol Chem ; 296: 100433, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33610554

RESUMEN

Defects in protein O-mannosylation lead to severe congenital muscular dystrophies collectively known as α-dystroglycanopathy. A hallmark of these diseases is the loss of the O-mannose-bound matriglycan on α-dystroglycan, which reduces cell adhesion to the extracellular matrix. Mutations in protein O-mannose ß1,2-N-acetylglucosaminyltransferase 1 (POMGNT1), which is crucial for the elongation of O-mannosyl glycans, have mainly been associated with muscle-eye-brain (MEB) disease. In addition to defects in cell-extracellular matrix adhesion, aberrant cell-cell adhesion has occasionally been observed in response to defects in POMGNT1. However, specific molecular consequences of POMGNT1 deficiency on cell-cell adhesion are largely unknown. We used POMGNT1 knockout HEK293T cells and fibroblasts from an MEB patient to gain deeper insight into the molecular changes in POMGNT1 deficiency. Biochemical and molecular biological techniques combined with proteomics, glycoproteomics, and glycomics revealed that a lack of POMGNT1 activity strengthens cell-cell adhesion. We demonstrate that the altered intrinsic adhesion properties are due to an increased abundance of N-cadherin (N-Cdh). In addition, site-specific changes in the N-glycan structures in the extracellular domain of N-Cdh were detected, which positively impact on homotypic interactions. Moreover, in POMGNT1-deficient cells, ERK1/2 and p38 signaling pathways are activated and transcriptional changes that are comparable with the epithelial-mesenchymal transition (EMT) are triggered, defining a possible molecular mechanism underlying the observed phenotype. Our study indicates that changes in cadherin-mediated cell-cell adhesion and other EMT-related processes may contribute to the complex clinical symptoms of MEB or α-dystroglycanopathy in general and suggests that the impact of changes in O-mannosylation on N-glycosylation has been underestimated.


Asunto(s)
Adhesión Celular/fisiología , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo , Antígenos CD/metabolismo , Antígenos CD/fisiología , Cadherinas/metabolismo , Cadherinas/fisiología , Adhesión Celular/genética , Distroglicanos/metabolismo , Glicómica , Glicosilación , Glicosiltransferasas/deficiencia , Glicosiltransferasas/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Manosa/química , Distrofias Musculares/genética , N-Acetilglucosaminiltransferasas/fisiología , Polisacáridos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Int J Cancer ; 150(4): 688-704, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34716584

RESUMEN

The surface inhibitory receptor NKG2A forms heterodimers with the invariant CD94 chain and is expressed on a subset of activated CD8 T cells. As antibodies to block NKG2A are currently tested in several efficacy trials for different tumor indications, it is important to characterize the NKG2A+ CD8 T cell population in the context of other inhibitory receptors. Here we used a well-controlled culture system to study the kinetics of inhibitory receptor expression. Naïve mouse CD8 T cells were synchronously and repeatedly activated by artificial antigen presenting cells in the presence of the homeostatic cytokine IL-7. The results revealed NKG2A as a late inhibitory receptor, expressed after repeated cognate antigen stimulations. In contrast, the expression of PD-1, TIGIT and LAG-3 was rapidly induced, hours after first contact and subsequently down regulated during each resting phase. This late, but stable expression kinetics of NKG2A was most similar to that of TIM-3 and CD39. Importantly, single-cell transcriptomics of human tumor-infiltrating lymphocytes (TILs) showed indeed that these receptors were often coexpressed by the same CD8 T cell cluster. Furthermore, NKG2A expression was associated with cell division and was promoted by TGF-ß in vitro, although TGF-ß signaling was not necessary in a mouse tumor model in vivo. In summary, our data show that PD-1 reflects recent TCR triggering, but that NKG2A is induced after repeated antigen stimulations and represents a late inhibitory receptor. Together with TIM-3 and CD39, NKG2A might thus mark actively dividing tumor-specific TILs.


Asunto(s)
Proteínas de Punto de Control Inmunitario/fisiología , Subfamília C de Receptores Similares a Lectina de Células NK/fisiología , Animales , Antígenos CD/fisiología , Linfocitos T CD8-positivos/inmunología , División Celular , Receptor 2 Celular del Virus de la Hepatitis A/fisiología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología , Receptores Inmunológicos/fisiología , Factor de Crecimiento Transformador beta/farmacología , Microambiente Tumoral , Proteína del Gen 3 de Activación de Linfocitos
5.
EMBO J ; 37(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29724755

RESUMEN

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Asunto(s)
Antígenos Bacterianos/fisiología , Antígenos CD/fisiología , Proteínas Bacterianas/fisiología , Moléculas de Adhesión Celular/fisiología , Helicobacter pylori/fisiología , Proteínas Oncogénicas/fisiología , Antígenos CD/química , Proteínas Bacterianas/química , Moléculas de Adhesión Celular/química , Células HEK293 , Humanos , Mutagénesis , Multimerización de Proteína , Transporte de Proteínas
6.
EMBO J ; 37(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29858229

RESUMEN

The human gastric pathogen Helicobacter pylori is a major causative agent of gastritis, peptic ulcer disease, and gastric cancer. As part of its adhesive lifestyle, the bacterium targets members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family by the conserved outer membrane adhesin HopQ. The HopQ-CEACAM1 interaction is associated with inflammatory responses and enables the intracellular delivery and phosphorylation of the CagA oncoprotein via a yet unknown mechanism. Here, we generated crystal structures of HopQ isotypes I and II bound to the N-terminal domain of human CEACAM1 (C1ND) and elucidated the structural basis of H. pylori specificity toward human CEACAM receptors. Both HopQ alleles target the ß-strands G, F, and C of C1ND, which form the trans dimerization interface in homo- and heterophilic CEACAM interactions. Using SAXS, we show that the HopQ ectodomain is sufficient to induce C1ND monomerization and thus providing H. pylori a route to influence CEACAM-mediated cell adherence and signaling events.


Asunto(s)
Antígenos CD/fisiología , Proteínas Bacterianas/fisiología , Moléculas de Adhesión Celular/fisiología , Helicobacter pylori/fisiología , Animales , Antígenos CD/química , Proteínas Bacterianas/química , Células CHO , Moléculas de Adhesión Celular/química , Línea Celular Tumoral , Cricetulus , Humanos , Multimerización de Proteína
7.
Proc Natl Acad Sci U S A ; 116(47): 23698-23704, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31690656

RESUMEN

Pyrophosphate deficiency may explain the excessive vascular calcification found in children with Hutchinson-Gilford progeria syndrome (HGPS) and in a mouse model of this disease. The present study found that hydrolysis products of ATP resulted in a <9% yield of pyrophosphate in wild-type blood and aortas, showing that eNTPD activity (ATP → phosphate) was greater than eNPP activity (ATP → pyrophosphate). Moreover, pyrophosphate synthesis from ATP was reduced and pyrophosphate hydrolysis (via TNAP; pyrophosphate → phosphate) was increased in both aortas and blood obtained from mice with HGPS. The reduced production of pyrophosphate, together with the reduction in plasma ATP, resulted in marked reduction of plasma pyrophosphate. The combination of TNAP inhibitor levamisole and eNTPD inhibitor ARL67156 increased the synthesis and reduced the degradation of pyrophosphate in aortas and blood ex vivo, suggesting that these combined inhibitors could represent a therapeutic approach for this devastating progeroid syndrome. Treatment with ATP prevented vascular calcification in HGPS mice but did not extend longevity. By contrast, combined treatment with ATP, levamisole, and ARL67156 prevented vascular calcification and extended longevity by 12% in HGPS mice. These findings suggest a therapeutic approach for children with HGPS.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Fosfatasa Alcalina/fisiología , Enfermedades de la Aorta/prevención & control , Apirasa/antagonistas & inhibidores , Calcinosis/prevención & control , Difosfatos/metabolismo , Levamisol/uso terapéutico , Progeria/tratamiento farmacológico , Pirofosfatasas/antagonistas & inhibidores , Adenosina Trifosfato/uso terapéutico , Fosfatasa Alcalina/antagonistas & inhibidores , Animales , Antígenos CD/fisiología , Enfermedades de la Aorta/enzimología , Apirasa/deficiencia , Apirasa/fisiología , Calcinosis/enzimología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Lamina Tipo A/genética , Longevidad/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/metabolismo , Hidrolasas Diéster Fosfóricas/deficiencia , Hidrolasas Diéster Fosfóricas/fisiología , Progeria/genética , Progeria/metabolismo , Progeria/patología , Pirofosfatasas/deficiencia , Pirofosfatasas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Br J Haematol ; 195(1): 113-118, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34426978

RESUMEN

Richter syndrome (RS), an aggressive lymphoma occurring in the context of chronic lymphocytic leukaemia/small lymphocytic lymphoma, is associated with poor prognosis when treated with conventional immunochemotherapy, therefore, improved treatments are required. Immune checkpoint blockade has shown efficacy in some B-cell malignancies and modest responses in early clinical trials for RS. We investigated the immune checkpoint profile of RS as a basis to inform rational therapeutic investigations in RS. Formalin-fixed, paraffin-embedded biopsies of RS (n = 19), de novo diffuse large B-cell lymphoma (DLBCL; n = 58), transformed indolent lymphomas (follicular [tFL], n = 16; marginal zone [tMZL], n = 24) and non-transformed small lymphocytic lymphoma (SLL; n = 15) underwent gene expression profiling using the NanoString Human Immunology panel. Copy number assessment was performed using next-generation sequencing. Immunohistochemistry (IHC) for LAG3 and PD-1 was performed. LAG3 gene expression was higher in RS compared to DLBCL (P = 0·0002, log2FC 1·96), tFL (P < 0·0001, log2FC 2·61), tMZL (P = 0·0004, log2FC 1·79) and SLL (P = 0·0057, log2FC 1·45). LAG3 gene expression correlated with the gene expression of human leukocyte antigen Class I and II, and related immune genes and immune checkpoints. IHC revealed LAG3 protein expression on both malignant RS cells and tumour-infiltrating lymphocytes. Our findings support the investigation of LAG3 inhibition to enhance anti-tumour responses in RS.


Asunto(s)
Antígenos CD/fisiología , Inhibidores de Puntos de Control Inmunológico , Leucemia Linfocítica Crónica de Células B/inmunología , Linfoma de Células B de la Zona Marginal/tratamiento farmacológico , Linfoma Folicular/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/inmunología , Terapia Molecular Dirigida , Proteínas de Neoplasias/fisiología , Antígenos CD/biosíntesis , Antígenos CD/genética , Linfocitos B/metabolismo , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Linfocitos Infiltrantes de Tumor/metabolismo , Linfoma de Células B de la Zona Marginal/genética , Linfoma Folicular/genética , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Síndrome , Proteína del Gen 3 de Activación de Linfocitos
9.
Eur J Immunol ; 50(8): 1126-1141, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32222966

RESUMEN

TIM-3 has been considered as a target in cancer immunotherapy. In T cells, inhibitory as well as activating functions have been ascribed to this molecule. Its role may therefore depend on the state of T cells and on the presence of interaction partners capable to perform functional pairing. Carcinoembryonic antigen-related cell adhesion molecule (CEACAM1) has been proposed to bind TIM-3 and to regulate its function. Using a T cell reporter platform we confirmed CEACAM1-mediated inhibition, but CEACAM1 did not functionally engage TIM-3. TIM-3 and CEACAM1 coexpression was limited to a small subset of activated T cells. Moreover, results obtained in extensive binding studies were not in support of an interaction between TIM-3 and CEACAM1. Cytoplasmic sequences derived from TIM-3 induced inhibitory signaling in our human T cell reporter system. Our results indicate that TIM-3 functions are independent of CEACAM1 and that this receptor has the capability to promote inhibitory signaling pathways in human T cells.


Asunto(s)
Antígenos CD/fisiología , Moléculas de Adhesión Celular/fisiología , Receptor 2 Celular del Virus de la Hepatitis A/fisiología , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Receptor 2 Celular del Virus de la Hepatitis A/análisis , Humanos , Células Jurkat , Activación de Linfocitos , Transducción de Señal/fisiología , Linfocitos T/inmunología
10.
PLoS Pathog ; 15(1): e1007429, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30653605

RESUMEN

HIV causes several forms of immune dysfunction that need to be addressed in a functional cure for HIV. Immune exhaustion describes a dysfunctional phenotype caused by chronic cellular activation. Lymphocyte activation gene-3 (LAG3) is one of several negative coreceptors known as immune checkpoints that contribute to this exhaustion phenotype. Antibodies targeting immune checkpoints are now used clinically to restore immunity against cancer and hold promise in restoring immunity during HIV infection. Here, we summarize current knowledge surrounding LAG3 and discuss its relevance during HIV infection and the potential for LAG3-targeting antibodies in a functional HIV cure.


Asunto(s)
Antígenos CD/fisiología , Infecciones por VIH/inmunología , Anticuerpos , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/inmunología , Humanos , Activación de Linfocitos , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Proteína del Gen 3 de Activación de Linfocitos
11.
PLoS Pathog ; 15(3): e1007633, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30875408

RESUMEN

Memory CD8+ T cells in the circulation rapidly infiltrate non-lymphoid tissues following infection and provide protective immunity in an antigen-specific manner. However, the subsequent fate of memory CD8+ T cells after entering non-lymphoid tissues such as the skin during a secondary infection is largely unknown. Furthermore, because expression of CD62L is often used to identify the central memory (TCM) CD8+ T cell subset, uncoupling the physical requirement for CD62L-mediated lymph node homing versus other functional attributes of TCM CD8+ T cells remains unresolved. Here, we show that in contrast to naïve CD8+ T cells, memory CD8+ T cells traffic into the skin independent of CD62L-mediated lymph node re-activation and provide robust protective immunity against Vaccinia virus (VacV) infection. TCM, but not effector memory (TEM), CD8+ T cells differentiated into functional CD69+/CD103- tissue residents following viral clearance, which was also dependent on local recognition of antigen in the skin microenvironment. Finally, we found that memory CD8+ T cells expressed granzyme B after trafficking into the skin and utilized cytolysis to provide protective immunity against VacV infection. Collectively, these findings demonstrate that TCM CD8+ T cells become cytolytic following rapid infiltration of the skin to protect against viral infection and subsequently differentiate into functional CD69+ tissue-residents.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica/fisiología , Piel/inmunología , Animales , Antígenos CD/metabolismo , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígenos de Diferenciación de Linfocitos T/fisiología , Linfocitos T CD8-positivos/virología , Femenino , Selectina L/metabolismo , Lectinas Tipo C/metabolismo , Lectinas Tipo C/fisiología , Ganglios Linfáticos , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/virología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/fisiología , Virus Vaccinia/inmunología , Virus Vaccinia/patogenicidad
12.
Nat Immunol ; 10(9): 973-80, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19648922

RESUMEN

The adaptors SAP, EAT-2 and ERT are specific to cells of the immune system and belong to the SAP family. All three are expressed in natural killer (NK) cells. Here we examined the global function of the SAP family using mice lacking SAP, EAT-2 and ERT. These adaptors acted together in a mechanism that was essential for the elimination of hematopoietic but not nonhematopoietic cells by NK cells. This function was mediated by many receptors of the SLAM family on NK cells that were engaged by ligands found solely on hematopoietic cells. In the absence of SAP-related adaptors, SLAM receptors lost their activating function and became inhibitory receptors that repressed other activating receptors, such as NKG2D. Hence, the SAP family is essential for the elimination of unwanted hematopoietic cells by NK cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Sistema Hematopoyético/citología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células Asesinas Naturales/fisiología , Animales , Antígenos CD/fisiología , Antígenos Ly/fisiología , Antígeno CD48 , Células CHO , Cricetinae , Cricetulus , Antígenos de Histocompatibilidad Clase I/fisiología , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/fisiología , Ratas , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Transcripción/fisiología
13.
FASEB J ; 34(11): 14960-14976, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32924185

RESUMEN

Atherosclerosis is an inflammatory disease characterized by the accumulation of macrophages in the vessel wall. Macrophages depend on their polarization to exert either pro-inflammatory or anti-inflammatory effects. Macrophages of the anti-inflammatory phenotype express high levels of CD163, a scavenger receptor for the hemoglobin-haptoglobin complex. CD163 can also bind to the pro-inflammatory cytokine TWEAK. Using ApoE-deficient or ApoE/CD163 double-deficient mice we aim to investigate the involvement of CD163 in atherosclerosis development and its capacity to neutralize the TWEAK actions. ApoE/CD163 double-deficient mice displayed a more unstable plaque phenotype characterized by an increased lipid and macrophage content, plaque size, and pro-inflammatory cytokine expression. In vitro experiments demonstrated that the absence of CD163 in M2-type macrophages-induced foam cell formation through upregulation of CD36 expression. Moreover, exogenous TWEAK administration increased atherosclerotic lesion size, lipids, and macrophages content in ApoE-/- /CD163-/- compared with ApoE-/- /CD163+/+ mice. Treatment with recombinant CD163 was able to neutralize the proatherogenic effects of TWEAK in ApoE/CD163 double-deficient mice. Recombinant CD163 abolished the pro-inflammatory actions of TWEAK on vascular smooth muscle cells, decreasing NF-kB activation, cytokines and metalloproteinases expression, and macrophages migration. In conclusion, CD163-expressing macrophages serve as a protective mechanism to prevent the deleterious effects of TWEAK on atherosclerotic plaque development and progression.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Diferenciación Mielomonocítica/fisiología , Aterosclerosis/patología , Citocina TWEAK/metabolismo , Células Espumosas/patología , Macrófagos/patología , Placa Aterosclerótica/patología , Receptores de Superficie Celular/fisiología , Animales , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Citocinas/metabolismo , Femenino , Células Espumosas/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Placa Aterosclerótica/etiología , Placa Aterosclerótica/metabolismo
14.
PLoS Biol ; 16(12): e2006649, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30550561

RESUMEN

Cell-surface transferrin receptor (CD71+) erythroid cells are abundant in newborns with immunomodulatory properties. Here, we show that neonatal CD71+ erythroid cells express significant levels of V-domain Immunoglobulin (Ig) Suppressor of T Cell Activation (VISTA) and, via constitutive production of transforming growth factor (TGF)- ß, play a pivotal role in promotion of naïve CD4+ T cells into regulatory T cells (Tregs). Interestingly, we discovered that CD71+VISTA+ erythroid cells produce significantly higher levels of TGF-ß compared to CD71+VISTA- erythroid cells and CD71+ erythroid cells from the VISTA knock-out (KO) mice. As a result, CD71+VISTA+ erythroid cells-compared to CD71+VISTA- and CD71+ erythroid cells from the VISTA KO mice-significantly exceed promotion of naïve CD4+ T cells into induced Tregs (iTreg) via TGF-ß in vitro. However, depletion of CD71+ erythroid cells had no significant effects on the frequency of Tregs in vivo. Surprisingly, we observed that the remaining and/or newly generated CD71+ erythroid cells following anti-CD71 antibody administration exhibit a different gene expression profile, evidenced by the up-regulation of VISTA, TGF-ß1, TGF-ß2, and program death ligand-1 (PDL-1), which may account as a compensatory mechanism for the maintenance of Treg population. We also observed that iTreg development by CD71+ erythroid cells is mediated through the inhibition of key signaling molecules phosphorylated protein kinase B (phospho-Akt) and phosphorylated mechanistic target of rapamycin (phospho-mTOR). Finally, we found that elimination of Tregs using forkhead box P3 (FOXP3)-diptheria toxin receptor (DTR) mice resulted in a significant expansion in the frequency of CD71+ erythroid cells in vivo. Collectively, these studies provide a novel, to our knowledge, insight into the cross-talk between CD71+ erythroid cells and Tregs in newborns. Our results highlight the biological role of CD71+ erythroid cells in the neonatal period and possibly beyond.


Asunto(s)
Células Eritroides/inmunología , Proteínas de la Membrana/fisiología , Receptores de Transferrina/fisiología , Animales , Antígenos CD/fisiología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Células Eritroides/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Activación de Linfocitos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Receptores de Superficie Celular , Receptores de Transferrina/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología
15.
Circ Res ; 124(6): 891-903, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30707047

RESUMEN

RATIONALE: Endothelial barrier function depends on the proper localization and function of the adherens junction protein VE (vascular endothelial)-cadherin. Previous studies have suggested a functional relationship between integrin-mediated adhesion complexes and VE-cadherin yet the underlying molecular links are unclear. Binding of the cytoskeletal adaptor protein talin to the ß-integrin cytoplasmic domain is a key final step in regulating the affinity of integrins for extracellular ligands (activation) but the role of integrin activation in VE-cadherin mediated endothelial barrier function is unknown. OBJECTIVE: To test the requirement of talin-dependent activation of ß1 integrin in VE-cadherin organization and endothelial cell (EC) barrier function. METHODS AND RESULTS: EC-specific deletion of talin in adult mice resulted in impaired stability of intestinal microvascular blood vessels, hemorrhage, and death. Talin-deficient endothelium showed altered VE-cadherin organization at EC junctions in vivo. shRNA (short hairpin RNA)-mediated knockdown of talin1 expression in cultured ECs led to increased radial actin stress fibers, increased adherens junction width and increased endothelial monolayer permeability measured by electrical cell-substrate impedance sensing. Restoring ß1-integrin activation in talin-deficient cells with a ß1-integrin activating antibody normalized both VE-cadherin organization and EC barrier function. In addition, VE-cadherin organization was normalized by reexpression of talin or integrin activating talin head domain but not a talin head domain mutant that is selectively deficient in activating integrins. CONCLUSIONS: Talin-dependent activation of EC ß1-integrin stabilizes VE-cadherin at endothelial junctions and promotes endothelial barrier function.


Asunto(s)
Antígenos CD/fisiología , Cadherinas/fisiología , Células Endoteliales/fisiología , Integrina beta1/fisiología , Talina/fisiología , Animales , Antígenos CD/análisis , Cadherinas/análisis , Femenino , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Uniones Intercelulares/metabolismo , Masculino , Ratones
16.
Heart Surg Forum ; 24(4): E764-E768, 2021 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-34473045

RESUMEN

BACKGROUND: Hypoxia induced injury of pulmonary microvascular endothelial barrier is closely related to the pathogenesis of acute lung injury after lung transplantation. VE-cadherin is an important structural molecule for pulmonary microvascular endothelial barrier. In this study, we aim to investigate the roles of VE-cadherin in hypoxia induced injury of pulmonary microvascular endothelial barrier. METHODS: Rat model of hypoxia and cultured pulmonary microvascular endothelial cells (PMVECs) were utilized. Determination of PMVECs apoptosis, skeleton combination was conducted to verify the effects of hypoxia on injury of pulmonary microvascular endothelial barrier. In addition, VE-cadherin expression was modulated by administration of siRNA in order to investigate the roles of VE-cadherin in hypoxia induced PMVECs apoptosis and skeleton recombination. RESULTS: Our data indicated that expression of VE-cadherin was down-regulated in hypoxia-exposed PMVECs. Whereas, in the cells treated using siRNA, down-regulation of VE-cadherin did not trigger PMVECs apoptosis, but it increased the sensitivity of PMVECs to the hypoxia induced apoptosis. In cases of hypoxia, the expression of VE-cadherin was significantly down-regulated, together with endothelial skeleton recombination and increase of permeability, which then triggered endothelial barrier dysfunction. CONCLUSIONS: These data verify that VE-cadherin expression played an important role in hypoxia induced PMVECs apoptosis and cellular skeletal recombination.


Asunto(s)
Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Antígenos CD/fisiología , Cadherinas/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Microcirculación , Circulación Pulmonar , Animales , Apoptosis , Permeabilidad de la Membrana Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/patología , Hipoxia , Masculino , Ratas Sprague-Dawley
17.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067904

RESUMEN

Lymphocyte activation gene 3 (LAG-3) is a cell surface inhibitory receptor with multiple biological activities over T cell activation and effector functions. LAG-3 plays a regulatory role in immunity and emerged some time ago as an inhibitory immune checkpoint molecule comparable to PD-1 and CTLA-4 and a potential target for enhancing anti-cancer immune responses. LAG-3 is the third inhibitory receptor to be exploited in human anti-cancer immunotherapies, and it is considered a potential next-generation cancer immunotherapy target in human therapy, right next to PD-1 and CTLA-4. Unlike PD-1 and CTLA-4, the exact mechanisms of action of LAG-3 and its relationship with other immune checkpoint molecules remain poorly understood. This is partly caused by the presence of non-conventional signaling motifs in its intracellular domain that are different from other conventional immunoregulatory signaling motifs but with similar inhibitory activities. Here we summarize the current understanding of LAG-3 signaling and its role in LAG-3 functions, from its mechanisms of action to clinical applications.


Asunto(s)
Antígenos CD/metabolismo , Antígenos CD/fisiología , Transducción de Señal/fisiología , Humanos , Inmunoterapia , Activación de Linfocitos , Neoplasias/metabolismo , Receptores Inmunológicos/metabolismo , Linfocitos T/inmunología , Proteína del Gen 3 de Activación de Linfocitos
18.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360833

RESUMEN

CD39 is an enzyme which is responsible, together with CD73, for a cascade converting adenosine triphosphate into adenosine diphosphate and cyclic adenosine monophosphate, ultimately leading to the release of an immunosuppressive form of adenosine in the tumor microenvironment. Here, we first review the environmental and genetic factors shaping CD39 expression. Second, we report CD39 functions in the T cell compartment, highlighting its role in regulatory T cells, conventional CD4+ T cells and CD8+ T cells. Finally, we compile a list of studies, from preclinical models to clinical trials, which have made essential contributions to the discovery of novel combinatorial approaches in the treatment of cancer.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Linfocitos T/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD/fisiología , Apirasa/genética , Apirasa/inmunología , Apirasa/fisiología , Regulación de la Expresión Génica , Humanos , Linfocitos T/inmunología
19.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34445616

RESUMEN

Neovascularization restores blood flow recovery after ischemia in peripheral arterial disease. The main two components of neovascularization are angiogenesis and arteriogenesis. Both of these processes contribute to functional improvements of blood flow after occlusion. However, discriminating between the specific contribution of each process is difficult. A frequently used model for investigating neovascularization is the murine hind limb ischemia model (HLI). With this model, it is difficult to determine the role of angiogenesis, because usually the timing for the sacrifice of the mice is chosen to be optimal for the analysis of arteriogenesis. More importantly, the occurring angiogenesis in the distal calf muscles is probably affected by the proximally occurring arteriogenesis. Therefore, to understand and subsequently intervene in the process of angiogenesis, a model is needed which investigates angiogenesis without the influence of arteriogenesis. In this study we evaluated the in vivo Matrigel plug assay in genetic deficient mice to investigate angiogenesis. Mice deficient for interferon regulatory factor (IRF)3, IRF7, RadioProtective 105 (RP105), Chemokine CC receptor CCR7, and p300/CBP-associated factor (PCAF) underwent the in vivo Matrigel model. Histological analysis of the Matrigel plugs showed an increased angiogenesis in mice deficient of IRF3, IRF7, and RP105, and a decreased angiogenesis in PCAF deficient mice. Our results also suggest an involvement of CCR7 in angiogenesis. Comparing our results with results of the HLI model found in the literature suggests that the in vivo Matrigel plug assay is superior in evaluating the angiogenic response after ischemia.


Asunto(s)
Antígenos CD/fisiología , Miembro Posterior/irrigación sanguínea , Factor 3 Regulador del Interferón/fisiología , Factor 7 Regulador del Interferón/fisiología , Isquemia/patología , Neovascularización Patológica/patología , Factores de Transcripción p300-CBP/fisiología , Animales , Colágeno , Combinación de Medicamentos , Miembro Posterior/patología , Isquemia/metabolismo , Laminina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Proteoglicanos , Recuperación de la Función
20.
J Virol ; 93(11)2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30867310

RESUMEN

The cellular protein bone marrow stromal antigen-2 (BST-2)/tetherin acts against a variety of enveloped viruses by restricting their release from the plasma membrane. The HIV-1 accessory protein Vpu counteracts BST-2 by downregulating it from the cell surface and displacing it from virion assembly sites. Previous comparisons of Vpus from transmitted/founder viruses and between viruses isolated during acute and chronic infection led to the identification of a tryptophan at position 76 in Vpu (W76) as a key determinant for the displacement of BST-2 from virion assembly sites. Although present in Vpus from clades B, D, and G, W76 is absent from Vpus from clades A, C, and H. Mutagenesis of the C-terminal region of Vpu from two clade C viruses led to the identification of a conserved LL sequence that is functionally analogous to W76 of clade B. Alanine substitution of these leucines partially impaired virion release. This impairment was even greater when the mutations were combined with mutations of the Vpu ß-TrCP binding site, resulting in Vpu proteins that induced high surface levels of BST-2 and reduced the efficiency of virion release to less than that of virus lacking vpu Microscopy confirmed that these C-terminal leucines in clade C Vpu, like W76 in clade B, contribute to virion release by supporting the displacement of BST-2 from virion assembly sites. These results suggest that although encoded differently, the ability of Vpu to displace BST-2 from sites of virion assembly on the plasma membrane is evolutionarily conserved among clade B and C HIV-1 isolates.IMPORTANCE Although targeted by a variety of restriction mechanisms, HIV-1 establishes chronic infection in most cases, in part due to the counteraction of these host defenses by viral accessory proteins. Using conserved motifs, the accessory proteins exploit the cellular machinery to degrade or mistraffic host restriction factors, thereby counteracting them. The Vpu protein counteracts the virion-tethering factor BST-2 in part by displacing it from virion assembly sites along the plasma membrane, but a previously identified determinant of that activity is clade specific at the level of protein sequence and not found in the clade C viruses that dominate the pandemic. Here, we show that clade C Vpu provides this activity via a leucine-containing sequence rather than the tryptophan-containing sequence found in clade B Vpu. This difference seems likely to reflect the different evolutionary paths taken by clade B and clade C HIV-1 in human populations.


Asunto(s)
Antígenos CD/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Liberación del Virus/fisiología , Antígenos CD/fisiología , Línea Celular , Membrana Celular/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Proteínas Ligadas a GPI/fisiología , Infecciones por VIH/virología , Seropositividad para VIH , VIH-1/metabolismo , VIH-1/fisiología , Células HeLa , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Humanos , Proteínas Reguladoras y Accesorias Virales/genética , Virión/genética , Virión/metabolismo , Ensamble de Virus/fisiología , Proteínas con Repetición de beta-Transducina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA