Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 238(5): 976-991, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36852591

RESUMEN

Voltage-dependent potassium channel Kv1.3 plays a key role on T-cell activation; however, lack of reliable antibodies has prevented its accurate detection under endogenous circumstances. To overcome this limitation, we created a Jurkat T-cell line with endogenous Kv1.3 channel tagged, to determine the expression, location, and changes upon activation of the native Kv1.3 channels. CRISPR-Cas9 technique was used to insert a Flag-Myc peptide at the C terminus of the KCNA3 gene. Basal or activated channel expression was studied using western blot analysis and imaging techniques. We identified two isoforms of Kv1.3 other than the canonical channel (54 KDa) differing on their N terminus: a longer isoform (70 KDa) and a truncated isoform (43 KDa). All three isoforms were upregulated after T-cell activation. We focused on the functional characterization of the truncated isoform (short form, SF), because it has not been previously described and could be present in the available Kv1.3-/- mice models. Overexpression of SF in HEK cells elicited small amplitude Kv1.3-like currents, which, contrary to canonical Kv1.3, did not induce HEK proliferation. To explore the role of endogenous SF isoform in a native system, we generated both a knockout Jurkat clone and a clone expressing only the SF isoform. Although the canonical isoform (long form) localizes mainly at the plasma membrane, SF remains intracellular, accumulating perinuclearly. Accordingly, SF Jurkat cells did not show Kv1.3 currents and exhibited depolarized resting membrane potential (VM ), decreased Ca2+ influx, and a reduction in the [Ca2+ ]i increase upon stimulation. Functional characterization of these Kv1.3 channel isoforms showed their differential contribution to signaling pathways involved in formation of the immunological synapse. We conclude that alternative translation initiation generates at least three endogenous Kv1.3 channel isoforms in T cells that exhibit different functional roles. For some of these functions, Kv1.3 proteins do not need to form functional plasma membrane channels.


Asunto(s)
Canal de Potasio Kv1.3 , Animales , Humanos , Ratones , Línea Celular , Membrana Celular/metabolismo , Células Jurkat , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo
2.
J Cell Physiol ; 236(6): 4330-4347, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33230847

RESUMEN

The voltage-dependent potassium channel Kv1.3 has been implicated in proliferation in many cell types, based on the observation that Kv1.3 blockers inhibited proliferation. By modulating membrane potential, cell volume, and/or Ca2+ influx, K+  channels can influence cell cycle progression. Also, noncanonical channel functions could contribute to modulate cell proliferation independent of K+ efflux. The specificity of the requirement of Kv1.3 channels for proliferation suggests the involvement of molecule-specific interactions, but the underlying mechanisms are poorly identified. Heterologous expression of Kv1.3 channels in HEK cells has been shown to increase proliferation independently of K+ fluxes. Likewise, some of the molecular determinants of Kv1.3-induced proliferation have been located in the C-terminus region, where individual point mutations of putative phosphorylation sites (Y447A and S459A) abolished Kv1.3-induced proliferation. Here, we investigated the mechanisms linking Kv1.3 channels to proliferation exploring the correlation between Kv1.3 voltage-dependent molecular dynamics and cell cycle progression. Using transfected HEK cells, we analyzed both the effect of changes in resting membrane potential on Kv1.3-induced proliferation and the effect of mutated Kv1.3 channels with altered voltage dependence of gating. We conclude that voltage-dependent transitions of Kv1.3 channels enable the activation of proliferative pathways. We also found that Kv1.3 associated with IQGAP3, a scaffold protein involved in proliferation, and that membrane depolarization facilitates their interaction. The functional contribution of Kv1.3-IQGAP3 interplay to cell proliferation was demonstrated both in HEK cells and in vascular smooth muscle cells. Our data indicate that voltage-dependent conformational changes of Kv1.3 are an essential element in Kv1.3-induced proliferation.


Asunto(s)
Proliferación Celular , Activación del Canal Iónico , Canal de Potasio Kv1.3/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Células HEK293 , Humanos , Canales KATP/genética , Canales KATP/metabolismo , Canal de Potasio Kv1.3/química , Canal de Potasio Kv1.3/genética , Potenciales de la Membrana , Mutación , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
3.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768773

RESUMEN

The FASTK family of proteins have been recently reported to play a key role in the post-transcriptional regulation of mitochondrial gene expression, including mRNA stability and translation. Accumulated studies have provided evidence that the expression of some FASTK genes is altered in certain types of cancer, in agreement with the central role of mitochondria in cancer development. Here, we obtained a pan-cancer overview of the genomic and transcriptomic alterations of FASTK genes. FASTK, FASTKD1, FASTKD3 and FASTKD5 showed the highest rates of genetic alterations. FASTK and FASTKD3 alterations consisted mainly of amplifications that were seen in more than 8% of ovarian and lung cancers, respectively. FASTKD1 and FASTKD5 were the most frequently mutated FASTK genes, and the mutations were identified in 5-7% of uterine cancers, as well as in 4% of melanomas. Our results also showed that the mRNA levels of all FASTK members were strongly upregulated in esophageal, stomach, liver and lung cancers. Finally, the protein-protein interaction network for FASTK proteins uncovers the interaction of FASTK, FASTKD2, FASTKD4 and FASTKD5 with cancer signaling pathways. These results serve as a starting point for future research into the potential of the FASTK family members as diagnostic and therapeutic targets for certain types of cancer.


Asunto(s)
Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Mapas de Interacción de Proteínas/genética , ARN Mensajero/metabolismo , Transducción de Señal/genética , Transcriptoma/genética
4.
Arterioscler Thromb Vasc Biol ; 39(12): e273-e286, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597447

RESUMEN

OBJECTIVE: We have previously described that changes in the expression of Kv channels associate to phenotypic modulation (PM), so that Kv1.3/Kv1.5 ratio is a landmark of vascular smooth muscle cells phenotype. Moreover, we demonstrated that the Kv1.3 functional expression is relevant for PM in several types of vascular lesions. Here, we explore the efficacy of Kv1.3 inhibition for the prevention of remodeling in human vessels, and the mechanisms linking the switch in Kv1.3 /Kv1.5 ratio to PM. Approach and Results: Vascular remodeling was explored using organ culture and primary cultures of vascular smooth muscle cells obtained from human vessels. We studied the effects of Kv1.3 inhibition on serum-induced remodeling, as well as the impact of viral vector-mediated overexpression of Kv channels or myocardin knock-down. Kv1.3 blockade prevented remodeling by inhibiting proliferation, migration, and extracellular matrix secretion. PM activated Kv1.3 via downregulation of Kv1.5. Hence, both Kv1.3 blockers and Kv1.5 overexpression inhibited remodeling in a nonadditive fashion. Finally, myocardin knock-down induced vessel remodeling and Kv1.5 downregulation and myocardin overexpression increased Kv1.5, while Kv1.5 overexpression inhibited PM without changing myocardin expression. CONCLUSIONS: We demonstrate that Kv1.5 channel gene is a myocardin-regulated, vascular smooth muscle cells contractile marker. Kv1.5 downregulation upon PM leaves Kv1.3 as the dominant Kv1 channel expressed in dedifferentiated cells. We demonstrated that the inhibition of Kv1.3 channel function with selective blockers or by preventing Kv1.5 downregulation can represent an effective, novel strategy for the prevention of intimal hyperplasia and restenosis of the human vessels used for coronary angioplasty procedures.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Vasos Coronarios/patología , Regulación de la Expresión Génica , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.5/genética , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/genética , Transactivadores/genética , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Humanos , Inmunohistoquímica , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/biosíntesis , Canal de Potasio Kv1.5/biosíntesis , Músculo Liso Vascular/patología , Proteínas Nucleares/biosíntesis , Técnicas de Cultivo de Órganos , Fenotipo , ARN/genética , Transactivadores/biosíntesis , Remodelación Vascular
5.
Am J Physiol Endocrinol Metab ; 317(5): E805-E819, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479304

RESUMEN

Inhibition of insulin-degrading enzyme (IDE) has been proposed as a possible therapeutic target for type 2 diabetes treatment. However, many aspects of IDE's role in glucose homeostasis need to be clarified. In light of this, new preclinical models are required to elucidate the specific role of this protease in the main tissues related to insulin handling. To address this, here we generated a novel line of mice with selective deletion of the Ide gene within pancreatic beta-cells, B-IDE-KO mice, which have been characterized in terms of multiple metabolic end points, including blood glucose, plasma C-peptide, and intraperitoneal glucose tolerance tests. In addition, glucose-stimulated insulin secretion was quantified in isolated pancreatic islets and beta-cell differentiation markers and insulin secretion machinery were characterized by RT-PCR. Additionally, IDE was genetically and pharmacologically inhibited in INS-1E cells and rodent and human islets, and insulin secretion was assessed. Our results show that, in vivo, life-long deletion of IDE from beta-cells results in increased plasma C-peptide levels. Corroborating these findings, isolated islets from B-IDE-KO mice showed constitutive insulin secretion, a hallmark of beta-cell functional immaturity. Unexpectedly, we found 60% increase in Glut1 (a high-affinity/low-Km glucose transporter), suggesting increased glucose transport into the beta-cell at low glucose levels, which may be related to constitutive insulin secretion. In parallel, IDE inhibition in INS-1E and islet cells resulted in impaired insulin secretion after glucose challenge. We conclude that IDE is required for glucose-stimulated insulin secretion. When IDE is inhibited, insulin secretion machinery is perturbed, causing either inhibition of insulin release at high glucose concentrations or constitutive secretion.


Asunto(s)
Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Insulisina/metabolismo , Animales , Glucemia/metabolismo , Péptido C/sangre , Femenino , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 1/metabolismo , Homeostasis , Humanos , Insulisina/genética , Masculino , Ratones , Ratones Noqueados , ARN Interferente Pequeño/farmacología , Ratas
6.
Nucleic Acids Res ; 45(19): 10941-10947, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29036396

RESUMEN

The FASTK family proteins have recently emerged as key post-transcriptional regulators of mitochondrial gene expression. FASTK, the founding member and its homologs FASTKD1-5 are architecturally related RNA-binding proteins, each having a different function in the regulation of mitochondrial RNA biology, from mRNA processing and maturation to ribosome assembly and translation. In this review, we outline the structure, evolution and function of these FASTK proteins and discuss the individual role that each has in mitochondrial RNA biology. In addition, we highlight the aspects of FASTK research that still require more attention.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Mitocondriales/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/genética , ARN/genética , Humanos , Proteínas Mitocondriales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mitocondrial , Proteínas de Unión al ARN/metabolismo
7.
J Allergy Clin Immunol ; 142(6): 1894-1908.e7, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29470999

RESUMEN

BACKGROUND: Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown. OBJECTIVE: The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation. METHODS: Isolated dendritic cells and bone marrow-derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC-/-, MHC class II-/-, Wiskott-Aldrich syndrome protein (WASP)-/-, 5C.C7 recombination-activating gene 2 (Rag2)-/-, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection. RESULTS: Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes. CONCLUSIONS: Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Mastocitos/efectos de los fármacos , NAD/farmacología , Adulto , Animales , Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Humanos , Listeria monocytogenes , Listeriosis/tratamiento farmacológico , Listeriosis/inmunología , Mastocitos/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , NAD/uso terapéutico
8.
Adv Exp Med Biol ; 1071: 51-59, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30357733

RESUMEN

The molecular mechanisms underlying O2-sensing by carotid body (CB) chemoreceptors remain undetermined. Mitochondria have been implicated, due to the sensitivity of CB response to electron transport chain (ETC) blockers. ETC is one of the major sources of reactive oxygen species, proposed as mediators in oxygen sensing. Fas-activated serine/threonine phosphoprotein is a sensor of mitochondrial stress that modulates protein translation to promote survival of cells exposed to adverse conditions. A translational variant of Fas-activated serine/threonine kinase (FASTK) is required for the biogenesis of ND6 mRNA, the mitochondrial encoded subunit 6 of the NADH dehydrogenase complex (Complex I). Ablating FASTK expression reduced Complex I activity in vivo by about 50%. We have tested the hypothesis of Complex I participation in O2-sensing structures by studying the effect of hypoxia in FASTK-/- knockout mice. Ventilatory response to acute hypoxia and hypercapnia tests showed similar sensitivity and CB catecholaminergic activity in knockout and wild type mice; hypoxic pulmonary vasoconstriction response also was similar. Pulmonary artery contractility in vitro, using small vessel myography, showed a significantly decreased relaxation to rotenone in knockout mice pre-constricted vessels with PGF2α. In conclusion, FASTK-/- knockout mice maintain respiratory chemoreflex under hypoxia and hypercapnia stress suggesting that completely functional Complex I ND6 protein is not required for these responses.


Asunto(s)
Cuerpo Carotídeo/fisiología , Complejo I de Transporte de Electrón/metabolismo , Hipoxia/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Hipercapnia/fisiopatología , Ratones , Ratones Noqueados , Mitocondrias , Proteínas Serina-Treonina Quinasas/genética
9.
J Biol Chem ; 291(50): 25877-25887, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27789713

RESUMEN

The Fas-activated serine/threonine kinase (FASTK) family of proteins has recently emerged as a central regulator of mitochondrial gene expression through the function of an unusual RNA-binding domain named RAP (for RNA-binding domain abundant in Apicomplexans), shared by all six members of the family. Here we describe the role of one of the less characterized members, FASTKD3, in mitochondrial RNA metabolism. First, we show that, in contrast to FASTK, FASTKD2, and FASTKD5, FASTKD3 does not localize in mitochondrial RNA granules, which are sites of processing and maturation of mtRNAs and ribosome biogenesis. Second, we generated FASTKD3 homozygous knock-out cell lines by homologous recombination and observed that the absence of FASTKD3 resulted in increased steady-state levels and half-lives of a subset of mature mitochondrial mRNAs: ND2, ND3, CYTB, COX2, and ATP8/6. No aberrant processing of RNA precursors was observed. Rescue experiments demonstrated that RAP domain is required for FASTKD3 function in mRNA stability. Besides, we describe that FASTKD3 is required for efficient COX1 mRNA translation without altering mRNA levels, which results in a decrease in the steady-state levels of COX1 protein. This finding is associated with reduced mitochondrial complex IV assembly and activity. Our observations suggest that the function of this family of proteins goes beyond RNA processing and ribosome assembly and includes RNA stability and translation regulation within mitochondria.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/metabolismo , ARN/metabolismo , Línea Celular Tumoral , Ciclooxigenasa 1/biosíntesis , Ciclooxigenasa 1/genética , Complejo IV de Transporte de Electrones/biosíntesis , Complejo IV de Transporte de Electrones/genética , Humanos , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Serina-Treonina Quinasas/genética , ARN/genética , Estabilidad del ARN , ARN Mensajero/genética , ARN Mitocondrial
10.
Blood ; 119(12): 2819-28, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22302739

RESUMEN

Wiskott Aldrich syndrome (WAS) is caused by mutations in the WAS gene that encodes for a protein (WASp) involved in cytoskeleton organization in hematopoietic cells. Several distinctive abnormalities of T, B, and natural killer lymphocytes; dendritic cells; and phagocytes have been found in WASp-deficient patients and mice; however, the in vivo consequence of WASp deficiency within individual blood cell lineages has not been definitively evaluated. By conditional gene deletion we have generated mice with selective deficiency of WASp in the B-cell lineage (B/WcKO mice). We show that this is sufficient to cause a severe reduction of marginal zone B cells and inability to respond to type II T-independent Ags, thereby recapitulating phenotypic features of complete WASp deficiency. In addition, B/WcKO mice showed prominent signs of B-cell dysregulation, as indicated by an increase in serum IgM levels, expansion of germinal center B cells and plasma cells, and elevated autoantibody production. These findings are accompanied by hyperproliferation of WASp-deficient follicular and germinal center B cells in heterozygous B/WcKO mice in vivo and excessive differentiation of WASp-deficient B cells into class-switched plasmablasts in vitro, suggesting that WASp-dependent B cell-intrinsic mechanisms critically contribute to WAS-associated autoimmunity.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Recuento de Células , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética
11.
PLoS One ; 18(9): e0291442, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37725617

RESUMEN

Increasing evidence demonstrate that the electron transfer chain plays a critical role in controlling the effector functions of macrophages. In this work, we have generated a Ndufs4-/- murine macrophage cell lines. The Ndufs4 gene, which encodes a supernumerary subunit of complex I, is a mutational hotspot in Leigh syndrome patients. Ndufs4-/- macrophages showed decreased complex I activity, altered complex I assembly, and lower levels of maximal respiration and ATP production. These mitochondrial respiration alterations were associated with a shift towards a pro-inflammatory cytokine profile after lipopolysaccharide challenge and improved ability to phagocytose Gram-negative bacteria.


Asunto(s)
Complejo I de Transporte de Electrón , Enfermedad de Leigh , Humanos , Animales , Ratones , Complejo I de Transporte de Electrón/genética , Macrófagos , Fagocitosis , Línea Celular
12.
J Immunol ; 184(9): 5325-32, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20363972

RESUMEN

We generated Fas-activated serine threonine phosphoprotein (FAST)-deficient mice (FAST(-/-)) to study the in vivo role of FAST in immune system function. In a model of house dust mite-induced allergic pulmonary inflammation, wild type mice develop a mixed cellular infiltrate composed of eosinophils, lymphocytes, and neutrophils. FAST(-/-) mice develop airway inflammation that is distinguished by the near absence of neutrophils. Similarly, LPS-induced alveolar neutrophil recruitment is markedly reduced in FAST(-/-) mice compared with wild type controls. This is accompanied by reduced concentrations of cytokines (TNF-alpha and IL-6 and -23) and chemoattractants (MIP-2 and keratinocyte chemoattractant) in bronchoalveolar lavage fluids. Because FAST(-/-) neutrophils exhibit normal chemotaxis and survival, impaired neutrophil recruitment is likely to be due to reduced production of chemoattractants within the pulmonary parenchyma. Studies using bone marrow chimeras implicate lung resident hematopoietic cells (e.g., pulmonary dendritic cells and/or alveolar macrophages) in this process. In conclusion, our results introduce FAST as a proinflammatory factor that modulates the function of lung resident hematopoietic cells to promote neutrophil recruitment and pulmonary inflammation.


Asunto(s)
Proteínas Mitocondriales/fisiología , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/patología , Receptor fas/fisiología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Alérgenos/administración & dosificación , Alérgenos/inmunología , Animales , Células Cultivadas , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Dermatophagoides pteronyssinus/inmunología , Polvo/inmunología , Femenino , Hematopoyesis/genética , Hematopoyesis/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/fisiología , Hipersensibilidad Respiratoria/genética , Antígeno Intracelular 1 de las Células T
13.
J Allergy Clin Immunol ; 128(1): 139-146, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21570718

RESUMEN

BACKGROUND: Cartilage-hair hypoplasia (CHH) is characterized by metaphyseal dysplasia, bone marrow failure, increased risk of malignancies, and a variable degree of immunodeficiency. CHH is caused by mutations in the RNA component of the mitochondrial RNA processing (RMRP) endoribonuclease gene, which is involved in ribosomal assembly, telomere function, and cell cycle control. OBJECTIVES: We aimed to define thymic output and characterize immune function in a cohort of patients with molecularly defined CHH with and without associated clinical immunodeficiency. METHODS: We studied the distribution of B and T lymphocytes (including recent thymic emigrants), in vitro lymphocyte proliferation, cell cycle, and apoptosis in 18 patients with CHH compared with controls. RESULTS: Patients with CHH have a markedly reduced number of recent thymic emigrants, and their peripheral T cells show defects in cell cycle control and display increased apoptosis, resulting in poor proliferation on activation. CONCLUSION: These data confirm that RMRP mutations result in significant defects of cell-mediated immunity and provide a link between the cellular phenotype and the immunodeficiency in CHH.


Asunto(s)
Apoptosis/inmunología , Ciclo Celular/inmunología , Enfermedad de Hirschsprung/inmunología , Síndromes de Inmunodeficiencia/inmunología , Osteocondrodisplasias/congénito , Linfocitos T/inmunología , Timo/inmunología , Adolescente , Separación Celular , Niño , Preescolar , Femenino , Citometría de Flujo , Genotipo , Cabello/anomalías , Cabello/inmunología , Cabello/patología , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/patología , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Lactante , Masculino , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/inmunología , Osteocondrodisplasias/patología , Fenotipo , Reacción en Cadena de la Polimerasa , Enfermedades de Inmunodeficiencia Primaria , ARN Largo no Codificante , ARN no Traducido/genética , Adulto Joven
15.
PLoS One ; 16(4): e0237413, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33930025

RESUMEN

Homologous recombination (HR) faithfully restores DNA double-strand breaks. Defects in this HR repair pathway are associated with cancer predisposition. In genetic engineering, HR has been used extensively to study gene function and it represents an ideal method of gene therapy for single gene disorders. Here, we present a novel assay to measure HR in living cells. The HR substrate consisted of a non-fluorescent 3' truncated form of the eGFP gene and was integrated into the AAVS1 locus, known as a safe harbor. The donor DNA template comprised a 5' truncated eGFP copy and was delivered via AAV particles. HR mediated repair restored full-length eGFP coding sequence, resulting in eGFP+ cells. The utility of our assay in quantifying HR events was validated by exploring the impact of the overexpression of HR promoters and the siRNA-mediated silencing of genes known to play a role in DNA repair on the frequency of HR. We conclude that this novel assay represents a useful tool to further investigate the mechanisms that control HR and test continually emerging tools for HR-mediated genome editing.


Asunto(s)
Proteínas Fluorescentes Verdes/genética , Recombinación Homóloga/genética , Línea Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Fluorescencia , Ingeniería Genética/métodos , Células HCT116 , Células HEK293 , Humanos , Neoplasias/genética , Regiones Promotoras Genéticas/genética , Reparación del ADN por Recombinación/genética
16.
Blood ; 112(10): 4139-47, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18772454

RESUMEN

Development of hematopoietic cells depends on a dynamic actin cytoskeleton. Here we demonstrate that expression of the cytoskeletal regulator WASP, mutated in the Wiskott-Aldrich syndrome, provides selective advantage for the development of naturally occurring regulatory T cells, natural killer T cells, CD4(+) and CD8(+) T lymphocytes, marginal zone (MZ) B cells, MZ macrophages, and platelets. To define the relative contribution of MZ B cells and MZ macrophages for MZ development, we generated wild-type and WASP-deficient bone marrow chimeric mice, with full restoration of the MZ. However, even in the presence of MZ macrophages, only 10% of MZ B cells were of WASP-deficient origin. We show that WASP-deficient MZ B cells hyperproliferate in vivo and fail to respond to sphingosine-1-phosphate, a crucial chemoattractant for MZ B-cell positioning. Abnormalities of the MZ compartment in WASP(-/-) mice lead to aberrant uptake of Staphylococcus aureus and to a reduced immune response to TNP-Ficoll. Moreover, WASP-deficient mice have increased levels of "natural" IgM antibodies. Our findings reveal that WASP regulates both development and function of hematopoietic cells. We demonstrate that WASP deficiency leads to an aberrant MZ that may affect responses to blood-borne pathogens and peripheral B-cell tolerance.


Asunto(s)
Linfocitos B/inmunología , Células Madre Hematopoyéticas/inmunología , Homeostasis/inmunología , Proteína del Síndrome de Wiskott-Aldrich/inmunología , Animales , Proliferación Celular/efectos de los fármacos , Factores Quimiotácticos/inmunología , Factores Quimiotácticos/farmacología , Homeostasis/efectos de los fármacos , Inmunoglobulina M/inmunología , Lisofosfolípidos/inmunología , Lisofosfolípidos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Esfingosina/análogos & derivados , Esfingosina/inmunología , Esfingosina/farmacología , Staphylococcus aureus/inmunología , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/genética
17.
Mol Cell Biol ; 26(14): 5214-25, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809760

RESUMEN

The adapter protein 3BP2 is expressed in lymphocytes; binds to Syk/ZAP-70, Vav, and phospholipase C-gamma (PLC-gamma); and is thought to be important for interleukin-2 gene transcription in T cells. To define the role of 3BP2 in lymphocyte development and function, we generated 3BP2-deficient mice. T-cell development, proliferation, cytokine secretion, and signaling in response to T-cell receptor (TCR) ligation were all normal in 3BP2(-/-) mice. 3BP2(-/-) mice had increased accumulation of pre-B cells in the bone marrow and a block in the progression of transitional B cells in the spleen from the T1 to the T2 stage, but normal numbers of mature B cells. B-cell proliferation, cell cycle progression, PLC-gamma2 phosphorylation, calcium mobilization, NF-ATp dephosphorylation, and Erk and Jnk activation in response to B-cell receptor (BCR) ligation were all impaired. These results suggest that 3BP2 is important for BCR, but not for TCR signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Secuencia de Bases , Diferenciación Celular , Supervivencia Celular , ADN Complementario/genética , Inmunoglobulinas/sangre , Linfopoyesis , Ratones , Ratones Noqueados , Transducción de Señal , Bazo/citología , Bazo/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo
19.
Sci Rep ; 6: 22325, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26928119

RESUMEN

CD4(+) CD25(+) Foxp3(+) Tregs have been shown to play a central role in immune homeostasis while preventing from fatal inflammatory responses, while Th17 cells have traditionally been recognized as pro-inflammatory mediators implicated in a myriad of diseases. Studies have shown the potential of Tregs to convert into Th17 cells, and Th17 cells into Tregs. Increasing evidence have pointed out CD25 as a key molecule during this transdifferentiation process, however molecules that allow such development remain unknown. Here, we investigated the impact of NAD(+) on the fate of CD4(+) CD25(+) Foxp3(+) Tregs in-depth, dissected their transcriptional signature profile and explored mechanisms underlying their conversion into IL-17A producing cells. Our results demonstrate that NAD(+) promotes Treg conversion into Th17 cells in vitro and in vivo via CD25 cell surface marker. Despite the reduced number of Tregs, known to promote homeostasis, and an increased number of pro-inflammatory Th17 cells, NAD(+) was able to promote an impressive allograft survival through a robust systemic IL-10 production that was CD4(+) CD25(+) Foxp3(+) independent. Collectively, our study unravels a novel immunoregulatory mechanism of NAD(+) that regulates Tregs fate while promoting allograft survival that may have clinical applications in alloimmunity and in a wide spectrum of inflammatory conditions.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Supervivencia de Injerto/efectos de los fármacos , Interleucina-10/metabolismo , NAD/uso terapéutico , Trasplante de Piel , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Aloinjertos/trasplante , Animales , Antígenos CD4/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Factores de Transcripción Forkhead/genética , Homeostasis/efectos de los fármacos , Humanos , Interleucina-10/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
20.
Nat Commun ; 5: 5101, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25290058

RESUMEN

CD4(+) T cells are involved in the development of autoimmunity, including multiple sclerosis (MS). Here we show that nicotinamide adenine dinucleotide (NAD(+)) blocks experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, by inducing immune homeostasis through CD4(+)IFNγ(+)IL-10(+) T cells and reverses disease progression by restoring tissue integrity via remyelination and neuroregeneration. We show that NAD(+) regulates CD4(+) T-cell differentiation through tryptophan hydroxylase-1 (Tph1), independently of well-established transcription factors. In the presence of NAD(+), the frequency of T-bet(-/-) CD4(+)IFNγ(+) T cells was twofold higher than wild-type CD4(+) T cells cultured in conventional T helper 1 polarizing conditions. Our findings unravel a new pathway orchestrating CD4(+) T-cell differentiation and demonstrate that NAD(+) may serve as a powerful therapeutic agent for the treatment of autoimmune and other diseases.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Vaina de Mielina/efectos de los fármacos , NAD/farmacología , Regeneración/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Homeostasis/efectos de los fármacos , Ratones , Triptófano Hidroxilasa/efectos de los fármacos , Triptófano Hidroxilasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA