Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell ; 39(3): 421-32, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20705243

RESUMEN

Cyclic phosphatidic acid (1-acyl-2,3-cyclic-glycerophosphate, CPA), one of nature's simplest phospholipids, is found in cells from slime mold to humans and has a largely unknown function. We find here that CPA is generated in mammalian cells in a stimulus-coupled manner by phospholipase D2 (PLD2) and binds to and inhibits the nuclear hormone receptor PPARgamma with nanomolar affinity and high specificity through stabilizing its interaction with the corepressor SMRT. CPA production inhibits the PPARgamma target-gene transcription that normally drives adipocytic differentiation of 3T3-L1 cells, lipid accumulation in RAW264.7 cells and primary mouse macrophages, and arterial wall remodeling in a rat model in vivo. Inhibition of PLD2 by shRNA, a dominant-negative mutant, or a small molecule inhibitor blocks CPA production and relieves PPARgamma inhibition. We conclude that CPA is a second messenger and a physiological inhibitor of PPARgamma, revealing that PPARgamma is regulated by endogenous agonists as well as by antagonists.


Asunto(s)
Adipocitos/metabolismo , Macrófagos/metabolismo , PPAR gamma/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfolipasa D/metabolismo , Células 3T3-L1 , Animales , Diferenciación Celular/fisiología , Ratones , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , PPAR gamma/genética , Ácidos Fosfatidicos/genética , Fosfolipasa D/genética , Ratas , Transcripción Genética/fisiología
2.
J Biol Chem ; 287(23): 19018-28, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22511778

RESUMEN

DiGeorge Critical Region 8 (DGCR8) is a double-stranded RNA-binding protein that interacts with Drosha and facilitates microRNA (miRNA) maturation. However, the role of DGCR8 in vascular smooth muscle cells (VSMCs) is not well understood. To investigate whether DGCR8 contributes to miRNA maturation in VSMCs, we generated DGCR8 conditional knockout (cKO) mice by crossing VSMC-specific Cre mice (SM22-Cre) with DGCR8(loxp/loxp) mice. We found that loss of DGCR8 in VSMCs resulted in extensive liver hemorrhage and embryonic mortality between embryonic days (E) 12.5 and E13.5. DGCR8 cKO embryos displayed dilated blood vessels and disarrayed vascular architecture. Blood vessels were absent in the yolk sac of DGCR8 KOs after E12.5. Disruption of DGCR8 in VSMCs reduced VSMC proliferation and promoted apoptosis in vitro and in vivo. In DGCR8 cKO embryos and knockout VSMCs, differentiation marker genes, including αSMA, SM22, and CNN1, were significantly down-regulated, and the survival pathways of ERK1/2 mitogen-activated protein kinase and the phosphatidylinositol 3-kinase/AKT were attenuated. Knockout of DGCR8 in VSMCs has led to down-regulation of the miR-17/92 and miR-143/145 clusters. We further demonstrated that the miR-17/92 cluster promotes VSMC proliferation and enhances VSMC marker gene expression, which may contribute to the defects of DGCR8 cKO mutants. Our results indicate that the DGCR8 gene is required for vascular development through the regulation of VSMC proliferation, apoptosis, and differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , MicroARNs/metabolismo , Músculo Liso Vascular/embriología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas/metabolismo , Animales , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Ratones , Ratones Noqueados , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Proteínas/genética , Proteínas de Unión al ARN , Saco Vitelino/irrigación sanguínea , Saco Vitelino/citología , Saco Vitelino/enzimología
3.
Front Immunol ; 14: 1174140, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37638013

RESUMEN

Introduction: The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods: Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results: Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion: These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.


Asunto(s)
Endotoxemia , Traumatismos por Radiación , alfa-Defensinas , Humanos , Adulto , Animales , Ratones , Células de Paneth , Disbiosis , Endotoxemia/etiología , ARN Ribosómico 16S , Traumatismos por Radiación/etiología , Citocinas , Inflamación
4.
Am J Respir Cell Mol Biol ; 46(4): 461-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22052879

RESUMEN

Both hyperoxia and mechanical ventilation can independently cause lung injury. In combination, these insults produce accelerated and severe lung injury. We recently reported that pre-exposure to hyperoxia for 12 hours, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone. We also reported that such injury and apoptosis are inhibited by antioxidant treatment. In this study, we hypothesized that apoptosis signal-regulating kinase-1 (ASK-1), a redox-sensitive, mitogen-activated protein kinase kinase kinase, plays a role in lung injury and apoptosis in this model. To determine the role of ASK-1 in lung injury, the release of inflammatory mediators and apoptosis, attributable to 12 hours of hyperoxia, were followed by large tidal volume mechanical ventilation with hyperoxia. Wild-type and ASK-1 knockout mice were subjected to hyperoxia (Fi(O(2)) = 0.9) for 12 hours before 4 hours of large tidal mechanical ventilation (tidal volume = 25 µl/g) with hyperoxia, and were compared with nonventilated control mice. Lung injury, apoptosis, and cytokine release were measured. The deletion of ASK-1 significantly inhibited lung injury and apoptosis, but did not affect the release of inflammatory mediators, compared with the wild-type mice. ASK-1 is an important regulator of lung injury and apoptosis in this model. Further study is needed to determine the mechanism of lung injury and apoptosis by ASK-1 and its downstream mediators in the lung.


Asunto(s)
MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control , Animales , Apoptosis/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Células Epiteliales/patología , Femenino , Hiperoxia/enzimología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , Alveolos Pulmonares/patología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
5.
Biochem Soc Trans ; 40(1): 31-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22260662

RESUMEN

LPA (lysophosphatidic acid, 1-acyl-2-hydroxy-sn-glycero-3-phosphate), is a growth factor-like lipid mediator that regulates many cellular functions, many of which are unique to malignantly transformed cells. The simple chemical structure of LPA and its profound effects in cancer cells has attracted the attention of the cancer therapeutics field and drives the development of therapeutics based on the LPA scaffold. In biological fluids, LPA is generated by ATX (autotaxin), a lysophospholipase D that cleaves the choline/serine headgroup from lysophosphatidylcholine and lysophosphatidylserine to generate LPA. In the present article, we review some of the key findings that make the ATX-LPA signalling axis an emerging target for cancer therapy.


Asunto(s)
Neoplasias/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Lisofosfolípidos/metabolismo , Terapia Molecular Dirigida , Invasividad Neoplásica , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Organofosfonatos/farmacología , Organofosfonatos/uso terapéutico , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Biochem Biophys Res Commun ; 408(3): 369-74, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21371421

RESUMEN

Dicer is a RNAase III enzyme that cleaves double stranded RNA and generates small interfering RNA (siRNA) and microRNA (miRNA). The goal of this study is to examine the role of Dicer and miRNAs in vascular smooth muscle cells (VSMCs). We deleted Dicer in VSMCs of mice, which caused a developmental delay that manifested as early as embryonic day E12.5, leading to embryonic death between E14.5 and E15.5 due to extensive hemorrhage in the liver, brain, and skin. Dicer KO embryos showed dilated blood vessels and a disarray of vascular architecture between E14.5 and E15.5. VSMC proliferation was significantly inhibited in Dicer KOs. The expression of VSMC marker genes were significantly downregulated in Dicer cKO embryos. The vascular structure of the yolk sac and embryo in Dicer KOs was lost to an extent that no blood vessels could be identified after E15.5. Expression of most miRNAs examined was compromised in VSMCs of Dicer KO. Our results indicate that Dicer is required for vascular development and regulates vascular remodeling by modulating VSMC proliferation and differentiation.


Asunto(s)
ARN Helicasas DEAD-box/genética , Discapacidades del Desarrollo/genética , Pérdida del Embrión/genética , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Neovascularización Fisiológica/genética , Ribonucleasa III/genética , Animales , Proliferación Celular , Eliminación de Gen , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología
7.
Cell Signal ; 78: 109850, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33253914

RESUMEN

The TP53 gene has been widely studied for its roles in cell cycle control, maintaining genome stability, activating repair mechanisms upon DNA damage, and initiating apoptosis should repair mechanisms fail. Thus, it is not surprising that mutations of p53 are the most common genetic alterations found in human cancer. Emerging evidence indicates that dysregulation of lipid metabolism by p53 can have a profound impact not only on cancer cells but also cells of the tumor microenvironment (TME). In particular, intermediates of the sphingolipid and lysophospholipid pathways regulate many cellular responses common to p53 such as cell survival, migration, DNA damage repair and apoptosis. The majority of these cellular events become dysregulated in cancer as well as cell senescence. In this review, we will provide an account on the seminal contributions of Prof. Lina Obeid, who deciphered the crosstalk between p53 and the sphingolipid pathway particularly in modulating DNA damage repair and apoptosis in non-transformed as well as transformed cells. We will also provide insights on the integrative role of p53 with the lysophosphatidic acid (LPA) signaling pathway in cancer progression and TME regulation.


Asunto(s)
Lisofosfolípidos/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Microambiente Tumoral , Proteína p53 Supresora de Tumor/metabolismo , Humanos , Lisofosfolípidos/genética , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética
8.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L711-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20833778

RESUMEN

Both high tidal volume mechanical ventilation (HV) and hyperoxia (HO) have been implicated in ventilator-induced lung injury. However, patients with acute lung injury are often exposed to HO before the application of mechanical ventilation. The potential priming of the lungs for subsequent injury by exposure to HO has not been extensively studied. We provide evidence that HO (90%) for 12 h followed by HV (25 µl/g) combined with HO for 2 or 4 h (HO-12h+HVHO-2h or -4h) induced severe lung injury in mice. Analysis of lung homogenates showed that lung injury was associated with cleavage of executioner caspases, caspases-3 and -7, and their downstream substrate poly(ADP-ribose) polymerase-1 (PARP-1). No significant lung injury or caspase cleavage was seen with either HO for 16 h or HV for up to 4 h. Ventilation for 4 h with HO (HVHO) did not cause significant lung injury without preexposure to HO. Twelve-hour HO followed by lower tidal volume (6 µl/g) mechanical ventilation failed to produce significant injury or caspase cleavage. We also evaluated the initiator caspases, caspases-8 and -9, to determine whether the death receptor or mitochondrial-mediated pathways were involved. Caspase-9 cleavage was observed in HO-12h+HVHO-2h and -4h as well as HO for 16 h. Caspase-8 activation was observed only in HO-12h+HVHO-4h, indicating the involvement of both pathways. Immunohistochemistry and in vitro stretch studies showed caspase cleavage in alveolar epithelial cells. In conclusion, preexposure to HO followed by HV produced severe lung injury associated with alveolar epithelial cell apoptosis.


Asunto(s)
Apoptosis/fisiología , Células Epiteliales/patología , Células Epiteliales/fisiología , Hiperoxia/complicaciones , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/etiología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología , Animales , Caspasas/metabolismo , Línea Celular , Activación Enzimática , Células Epiteliales/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Poli Adenosina Difosfato Ribosa/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Respiración Artificial/efectos adversos , Estrés Mecánico
9.
Am J Physiol Lung Cell Mol Physiol ; 298(6): L819-29, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20348281

RESUMEN

Pulmonary fibroblasts regulate extracellular matrix production and degradation and are critical in maintenance of lung structure, function, and repair, but they also play a central role in lung fibrosis. cAMP-elevating agents inhibit cytokine- and growth factor-stimulated myofibroblast differentiation and collagen synthesis in pulmonary fibroblasts. In the present study, we overexpressed adenylyl cyclase 6 (AC6) in pulmonary fibroblasts and measured cAMP production and collagen synthesis. AC6 overexpression enhanced cAMP production and the inhibition of collagen synthesis mediated by isoproterenol and beraprost, but not the responses to butaprost or PGE(2). To examine if increased AC6 expression would impact the development of fibrosis in an animal model, we generated transgenic mice that overexpress AC6 under a fibroblast-specific promoter, FTS1. Lung fibrosis was induced in FTS1-AC6(+/-) mice and littermate controls by intratracheal instillation of saline or bleomycin. Wild-type mice treated with bleomycin showed extensive peribronchial and interstitial fibrosis and collagen deposition. By contrast, FTS1-AC6(+/-) mice displayed decreased fibrotic development, lymphocyte infiltration (as determined by pathological scoring), and lung collagen content. Thus, AC6 overexpression inhibits fibrogenesis in the lung by reducing pulmonary fibroblast-mediated collagen synthesis and myofibroblast differentiation. Because AC6 overexpression does not lead to enhanced basal or PGE(2)-stimulated levels of cAMP, we conclude that endogenous catecholamines or prostacyclin is produced during bleomycin-induced lung fibrosis and that these signals have antifibrotic potential.


Asunto(s)
Adenilil Ciclasas/biosíntesis , Fibroblastos/fisiología , Fibrosis Pulmonar/fisiopatología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Bleomicina , Proteínas de Unión al Calcio/fisiología , Catecolaminas/fisiología , Colforsina/farmacología , Colágeno/biosíntesis , AMP Cíclico/biosíntesis , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Fibroblastos/efectos de los fármacos , Isoproterenol/farmacología , Pulmón/metabolismo , Pulmón/patología , Microdominios de Membrana/enzimología , Ratones , Ratones Transgénicos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Ratas , Proteína de Unión al Calcio S100A4 , Proteínas S100 , Transducción de Señal
10.
J Exp Med ; 199(6): 763-74, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15007093

RESUMEN

Neointimal lesions are characterized by accumulation of cells within the arterial wall and are a prelude to atherosclerotic disease. Here we report that a brief exposure to either alkyl ether analogs of the growth factor-like phospholipid lysophosphatidic acid (LPA), products generated during the oxidative modification of low density lipoprotein, or to unsaturated acyl forms of LPA induce progressive formation of neointima in vivo in a rat carotid artery model. This effect is completely inhibited by the peroxisome proliferator-activated receptor (PPAR)gamma antagonist GW9662 and mimicked by PPARgamma agonists Rosiglitazone and 1-O-hexadecyl-2-azeleoyl-phosphatidylcholine. In contrast, stearoyl-oxovaleryl phosphatidylcholine, a PPARalpha agonist and polypeptide epidermal growth factor, platelet-derived growth factor, and vascular endothelial growth factor failed to elicit neointima. The structure-activity relationship for neointima induction by LPA analogs in vivo is identical to that of PPARgamma activation in vitro and disparate from that of LPA G protein-coupled receptor activation. Neointima-inducing LPA analogs up-regulated the CD36 scavenger receptor in vitro and in vivo and elicited dedifferentiation of cultured vascular smooth muscle cells that was prevented by GW9662. These results suggest that selected LPA analogs are important novel endogenous PPARgamma ligands capable of mediating vascular remodeling and that activation of the nuclear transcription factor PPARgamma is both necessary and sufficient for neointima formation by components of oxidized low density lipoprotein.


Asunto(s)
Anilidas/farmacología , Arteriosclerosis/inducido químicamente , Enfermedades de las Arterias Carótidas/inducido químicamente , Lisofosfolípidos/toxicidad , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Análisis de Varianza , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Células Cultivadas , Cartilla de ADN , Modelos Animales de Enfermedad , Sustancias de Crecimiento/metabolismo , Ligandos , Lipoproteínas LDL/metabolismo , Masculino , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Relación Estructura-Actividad , Tiazolidinedionas/toxicidad , Factores de Tiempo , Factores de Transcripción/agonistas
11.
Cancers (Basel) ; 12(5)2020 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-32397679

RESUMEN

The tumor microenvironment (TME) may be best conceptualized as an ecosystem comprised of cancer cells interacting with a multitude of stromal components such as the extracellular matrix (ECM), blood and lymphatic networks, fibroblasts, adipocytes, and cells of the immune system. At the center of this crosstalk between cancer cells and their TME is the bioactive lipid lysophosphatidic acid (LPA). High levels of LPA and the enzyme generating it, termed autotaxin (ATX), are present in many cancers. It is also well documented that LPA drives tumor progression by promoting angiogenesis, proliferation, survival, invasion and metastasis. One of the hallmarks of cancer is the ability to modulate and escape immune detection and eradication. Despite the profound role of LPA in regulating immune functions and inflammation, its role in the context of tumor immunity has not received much attention until recently where emerging studies highlight that this signaling axis may be a means that cancer cells adopt to evade immune detection and eradication. The present review aims to look at the immunomodulatory actions of LPA in baseline immunity to provide a broad understanding of the subject with a special emphasis on LPA and cancer immunity, highlighting the latest progress in this area of research.

12.
Adv Biol Regul ; 71: 183-193, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30243984

RESUMEN

The lipid mediator lysophosphatidic acid (LPA) in biological fluids is primarily produced by cleavage of lysophospholipids by the lysophospholipase D enzyme Autotaxin (ATX). LPA has been identified and abundantly detected in the culture medium of various cancer cell types, tumor effusates, and ascites fluid of cancer patients. Our current understanding of the physiological role of LPA established its role in fundamental biological responses that include cell proliferation, metabolism, neuronal differentiation, angiogenesis, cell migration, hematopoiesis, inflammation, immunity, wound healing, regulation of cell excitability, and the promotion of cell survival by protecting against apoptotic death. These essential biological responses elicited by LPA are seemingly hijacked by cancer cells in many ways; transcriptional upregulation of ATX leading to increased LPA levels, enhanced expression of multiple LPA GPCR subtypes, and the downregulation of its metabolic breakdown. Recent studies have shown that overexpression of ATX and LPA GPCR can lead to malignant transformation, enhanced proliferation of cancer stem cells, increased invasion and metastasis, reprogramming of the tumor microenvironment and the metastatic niche, and development of resistance to chemo-, immuno-, and radiation-therapy of cancer. The fundamental role of LPA in cancer progression and the therapeutic inhibition of the ATX-LPA axis, although highly appealing, remains unexploited as drug development to these targets has not reached into the clinic yet. The purpose of this brief review is to highlight some unique signaling mechanisms engaged by the ATX-LPA axis and emphasize the therapeutic potential that lies in blocking the molecular targets of the LPA system.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Lisofosfolípidos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/patología , Humanos , Neoplasias/patología
13.
Sci Transl Med ; 11(482)2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842314

RESUMEN

The failure of anti-CD20 antibody (Rituximab) as therapy for lupus may be attributed to the transient and incomplete B cell depletion achieved in clinical trials. Here, using an alternative approach, we report that complete and sustained CD19+ B cell depletion is a highly effective therapy in lupus models. CD8+ T cells expressing CD19-targeted chimeric antigen receptors (CARs) persistently depleted CD19+ B cells, eliminated autoantibody production, reversed disease manifestations in target organs, and extended life spans well beyond normal in the (NZB × NZW) F1 and MRL fas/fas mouse models of lupus. CAR T cells were active for 1 year in vivo and were enriched in the CD44+CD62L+ T cell subset. Adoptively transferred splenic T cells from CAR T cell-treated mice depleted CD19+ B cells and reduced disease in naive autoimmune mice, indicating that disease control was cell-mediated. Sustained B cell depletion with CD19-targeted CAR T cell immunotherapy is a stable and effective strategy to treat murine lupus, and its effectiveness should be explored in clinical trials for lupus.


Asunto(s)
Antígenos CD19/metabolismo , Linfocitos B/inmunología , Inmunoterapia Adoptiva , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Depleción Linfocítica , Linfocitos T/metabolismo , Animales , Femenino , Lupus Eritematoso Sistémico/sangre , Ratones , Fenotipo , Proteoma/metabolismo , Análisis de Supervivencia
14.
Sci Rep ; 8(1): 16241, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389960

RESUMEN

Alcohol consumption has been shown to cause dysbiosis, but the mechanism involved in it is unknown. Recurrent colitis is known to induce expression of α-defensins in the colon, but the effect of alcohol consumption on it is not known. We investigated the effect of ethanol on α-defensin expression in the small intestine and colitis-induced expression in colon in mice. Furthermore, we evaluated the effect of human defensin-5 (HD5) on ethanol and colitis-induced gut barrier dysfunction and mucosal damage. Recurrent colitis was induced by feeding dextran sulfate sodium (DSS), 3 cycles of 5-days each with 15 days intervals, followed by 30-days remission. Ethanol was fed during the intervals and recovery in a liquid diet with or without HD5. Expression of α-defensins, tight junction (TJ) integrity and cytokine/chemokine expression were analyzed. Chronic ethanol feeding reduced α-defensin expression in the small intestine and colitis-induced defensin expression in the colon. HD5 attenuated the growth of enterotoxigenic Bacteriodes fragilis and E. coli, but had no effect on non-toxigenic Bacteriodes fragilis or probiotics, the Lactobacilli. Ethanol and colitis elevated Enterobacteriaceae, Firmicutes and Firmicutes to Bacteriodetes ratio in colonic mucosa. HD5 feeding attenuated ethanol and colitis-induced dysbiosis, disruption of intestinal epithelial TJ, mucosal inflammation, expression of pro-inflammatory cytokines and chemokines in the small intestine and colon, and endotoxemia. These results demonstrate that ethanol suppresses intestinal α-defensin expression, leading to dysbiosis, barrier dysfunction, inflammation and endotoxemia. HD5 feeding attenuates intestinal injury caused by ethanol and colitis, indicating that defensin expression is a potential target for treatment of alcoholic tissue injury and colitis.


Asunto(s)
Colitis Ulcerosa/tratamiento farmacológico , Disbiosis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , alfa-Defensinas/administración & dosificación , Administración Oral , Animales , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/microbiología , Colon/patología , ADN Bacteriano/aislamiento & purificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/microbiología , Disbiosis/patología , Etanol/toxicidad , Femenino , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Resultado del Tratamiento , alfa-Defensinas/síntesis química
15.
Cell Signal ; 16(3): 385-99, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14687668

RESUMEN

Lipid phosphate phosphatase 1 (LPP-1) is presumed to regulate the balance between lipid phosphates and their dephosphorylated counterparts. The currently prevailing hypothesis based on in vitro studies proposes that LPP-1 should regulate phospholipid lipid growth factors and second messengers, including lysophosphatidic acid (LPA), sphingosine 1-phosphate (S1P), diacylglycerol (DAG), and phosphatidic acid (PA). To evaluate the role of LPP-1 in vivo, three transgenic lines were established. RT-PCR, Western blotting, and enzymatic activity measurement confirmed a copy number-dependent ubiquitous overexpression of LPP-1. PMA-stimulated PA production in immortalized LPP-1 fibroblasts led to an elevation in the accumulation of DAG without major changes in the phospholipid classes isolated from the liver. The LPP-1 phenotype showed reduced body size, birth weight, and abnormalities in fur growth, whereas histological abnormalities included significantly decreased number of hair follicles, disrupted hair structure, and a severely impaired spermatogenesis. Implantation of LPP-1 or wild-type embryos into pseudopregnant LPP-1 mothers yielded a reduced litter size. The plasma level of alanine-leucine aminotransferase was significantly elevated. Unexpectedly, plasma concentrations of the five major acyl-species of LPA were indistinguishable between wild-type and LPP-1 animals. In contrast with previous studies using plasmid-mediated overexpression in vitro, transgenic overexpression of LPP-1 did not affect ERK1/2 activation elicited by LPA, S1P, thrombin, epidermal growth factor (EGF), and platelet-derived growth factor (PDGF), which was presumed to be a major signaling event regulated by LPP-1. Thus, transgenic overexpression of LPP-1 in mice elicited a number of unexpected phenotypic alterations without affecting several aspects of LPA signaling, which point to previously unappreciated mechanisms and roles of lipid phosphates in select organs.


Asunto(s)
Lisofosfolípidos/metabolismo , Fosfatidato Fosfatasa/metabolismo , Animales , Animales Modificados Genéticamente , Diacilglicerol Quinasa/metabolismo , Implantación del Embrión , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Cabello/ultraestructura , Hígado/enzimología , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Fosfatidato Fosfatasa/genética , Ácidos Fosfatidicos/metabolismo , Regiones Promotoras Genéticas , Espermatogénesis , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Testículo/enzimología , Transgenes
16.
Radiat Res ; 183(4): 465-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25807318

RESUMEN

We have previously demonstrated that the small molecule octadecenyl thiophosphate (OTP), a synthetic mimic of the growth factor-like mediator lysophosphatidic acid (LPA), showed radioprotective activity in a mouse model of total-body irradiation (TBI) when given orally or intraperitoneally 30 min before exposure to 9 Gy γ radiation. In the current study, we evaluated the effects of OTP, delivered subcutaneously, for radioprotection or radiomitigation from -24 h before to up to +72 h postirradiation using a mouse TBI model with therapeutic doses at around 1 mg/kg. OTP was injected at 10 mg/kg without observable toxic side effects in mice, providing a comfortable safety margin. Treatment of C57BL/6 mice with a single dose of OTP over the time period from -12 h before to +26 h after a lethal dose of TBI reduced mortality by 50%. When administered at +48 h to +72 h postirradiation (LD50/30 to LD100/30), OTP reduced mortality by ≥34%. OTP administered at +24 h postirradiation significantly elevated peripheral white blood cell and platelet counts, increased crypt survival in the jejunum, enhanced intestinal glucose absorption and reduced endotoxin seepage into the blood. In the 6.4-8.6 Gy TBI range using LD50/10 as the end point, OTP yielded a dose modification factor of 1.2. The current data indicate that OTP is a potent radioprotector and radiomitigator ameliorating the mortality and tissue injury of acute hematopoietic as well as acute gastrointestinal radiation syndrome.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de la radiación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Lisofosfolípidos/metabolismo , Compuestos Organofosforados/farmacología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos CD34/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/efectos de la radiación , Materiales Biomiméticos/efectos adversos , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Clonales/citología , Células Clonales/efectos de los fármacos , Células Clonales/efectos de la radiación , Relación Dosis-Respuesta a Droga , Femenino , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Glucosa/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Proteínas con Dominio LIM/metabolismo , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Compuestos Organofosforados/efectos adversos , Compuestos Organofosforados/farmacocinética , Fosfoproteínas/metabolismo , Recuento de Plaquetas , Complejo de la Endopetidasa Proteasomal , Protectores contra Radiación/efectos adversos , Protectores contra Radiación/farmacocinética , Protectores contra Radiación/farmacología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Factores de Transcripción/metabolismo , Irradiación Corporal Total/efectos adversos
17.
Cell Signal ; 27(9): 1751-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26027517

RESUMEN

In this study we characterized the effects of radiation injury on the expression and function of the autotaxin (ATX)-LPA2 GPCR axis. In IEC-6 crypt cells and jejunum enteroids quantitative RT-PCR showed a time- and dose-dependent upregulation of lpa2 in response to γ-irradiation that was abolished by mutation of the NF-κB site in the lpa2 promoter or by inhibition of ATM/ATR kinases with CGK-733, suggesting that lpa2 is a DNA damage response gene upregulated by ATM via NF-κB. The resolution kinetics of the DNA damage marker γ-H2AX in LPA-treated IEC-6 cells exposed to γ-irradiation was accelerated compared to vehicle, whereas pharmacological inhibition of LPA2 delayed the resolution of γ-H2AX. In LPA2-reconstituted MEF cells lacking LPA1&3 the levels of γ-H2AX decreased rapidly, whereas in Vector MEF were high and remained sustained. Inhibition of ERK1&2 or PI3K/AKT signaling axis by pertussis toxin or the C311A/C314A/L351A mutation in the C-terminus of LPA2 abrogated the effect of LPA on DNA repair. LPA2 transcripts in Lin(-)Sca-1(+)c-Kit(+) enriched for bone marrow stem cells were 27- and 5-fold higher than in common myeloid or lymphoid progenitors, respectively. Furthermore, after irradiation higher residual γ-H2AX levels were detected in the bone marrow or jejunum of irradiated LPA2-KO mice compared to WT mice. We found that γ-irradiation increases plasma ATX activity and LPA level that is in part due to the previously established radiation-induced upregulation of TNFα. These findings identify ATX and LPA2 as radiation-regulated genes that appear to play a physiological role in DNA repair.


Asunto(s)
Daño del ADN , Rayos gamma , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Hidrolasas Diéster Fosfóricas/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Línea Celular , Yeyuno/metabolismo , Yeyuno/patología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/patología , Ratas , Receptores del Ácido Lisofosfatídico/genética , Elementos de Respuesta
18.
Mol Cancer Res ; 13(1): 174-85, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25158955

RESUMEN

UNLABELLED: Autotaxin (ENPP2/ATX) and lysophosphatidic acid (LPA) receptors represent two key players in regulating cancer progression. The present study sought to understand the mechanistic role of LPA G protein-coupled receptors (GPCR), not only in the tumor cells but also in stromal cells of the tumor microenvironment. B16F10 melanoma cells predominantly express LPA5 and LPA2 receptors but lack LPA1. LPA dose dependently inhibited invasion of cells across a Matrigel layer. RNAi-mediated knockdown of LPA5 relieved the inhibitory effect of LPA on invasion without affecting basal invasion. This suggests that LPA5 exerts an anti-invasive action in melanoma cells in response to LPA. In addition, both siRNA-mediated knockdown and pharmacologic inhibition of LPA2 reduced the basal rate invasion. Unexpectedly, when probing the role of this GPCR in host tissues, it was found that the incidence of melanoma-derived lung metastasis was greatly reduced in LPA5 knockout (KO) mice compared with wild-type (WT) mice. LPA1-KO but not LPA2-KO mice also showed diminished melanoma-derived lung metastasis, suggesting that host LPA1 and LPA5 receptors play critical roles in the seeding of metastasis. The decrease in tumor cell residence in the lungs of LPA1-KO and LPA5-KO animals was apparent 24 hours after injection. However, KO of LPA1, LPA2, or LPA5 did not affect the subcutaneous growth of melanoma tumors. IMPLICATIONS: These findings suggest that tumor and stromal LPA receptors, in particular LPA1 and LPA5, play different roles in invasion and the seeding of metastasis.


Asunto(s)
Neoplasias Pulmonares/genética , Melanoma Experimental/genética , Receptores del Ácido Lisofosfatídico/genética , Animales , Carcinogénesis/genética , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Hidrolasas Diéster Fosfóricas/genética , Transducción de Señal/genética , Microambiente Tumoral
19.
Chem Biol ; 22(2): 206-16, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25619933

RESUMEN

Pharmacological mitigation of injuries caused by high-dose ionizing radiation is an unsolved medical problem. A specific nonlipid agonist of the type 2 G protein coupled receptor for lysophosphatidic acid (LPA2) 2-[4-(1,3-dioxo-1H,3H-benzoisoquinolin-2-yl)butylsulfamoyl]benzoic acid (DBIBB) when administered with a postirradiation delay of up to 72 hr reduced mortality of C57BL/6 mice but not LPA2 knockout mice. DBIBB mitigated the gastrointestinal radiation syndrome, increased intestinal crypt survival and enterocyte proliferation, and reduced apoptosis. DBIBB enhanced DNA repair by augmenting the resolution of γ-H2AX foci, increased clonogenic survival of irradiated IEC-6 cells, attenuated the radiation-induced death of human CD34(+) hematopoietic progenitors and enhanced the survival of the granulocyte/macrophage lineage. DBIBB also increased the survival of mice suffering from the hematopoietic acute radiation syndrome after total-body irradiation. DBIBB represents a drug candidate capable of mitigating acute radiation syndrome caused by high-dose γ-radiation to the hematopoietic and gastrointestinal system.


Asunto(s)
Apoptosis/efectos de los fármacos , Lisofosfolípidos/farmacología , Naftalimidas/farmacología , Receptores del Ácido Lisofosfatídico/agonistas , Sulfonamidas/farmacología , Síndrome de Radiación Aguda/metabolismo , Síndrome de Radiación Aguda/patología , Síndrome de Radiación Aguda/prevención & control , Animales , Apoptosis/efectos de la radiación , Sitios de Unión , Caspasa 8/metabolismo , Línea Celular , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/efectos de la radiación , Rayos gamma , Histonas/metabolismo , Humanos , Lisofosfolípidos/química , Lisofosfolípidos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación del Acoplamiento Molecular , Naftalimidas/química , Naftalimidas/uso terapéutico , Estructura Terciaria de Proteína , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Sulfonamidas/química , Sulfonamidas/uso terapéutico
20.
J Appl Physiol (1985) ; 111(5): 1467-76, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21799126

RESUMEN

Both prolonged exposure to hyperoxia and large tidal volume mechanical ventilation can each independently cause lung injury. However, the combined impact of these insults is poorly understood. We recently reported that preexposure to hyperoxia for 12 h, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone (Makena et al. Am J Physiol Lung Cell Mol Physiol 299: L711-L719, 2010). The upstream mechanisms of this lung injury and apoptosis have not been clearly elucidated. We hypothesized that lung injury in this model was dependent on oxidative signaling via the c-Jun NH(2)-terminal kinases (JNK). We, therefore, evaluated lung injury and apoptosis in the presence of N-acetyl-cysteine (NAC) in both mouse and cell culture models, and we provide evidence that NAC significantly inhibited lung injury and apoptosis by reducing the production of ROS, activation of JNK, and apoptosis. To confirm JNK involvement in apoptosis, cells treated with a specific JNK inhibitor, SP600125, and subjected to preexposure to hyperoxia, followed by mechanical stretch, exhibited significantly reduced evidence of apoptosis. In conclusion, lung injury and apoptosis caused by preexposure to hyperoxia, followed by high tidal volume mechanical ventilation, induces ROS-mediated activation of JNK and mitochondrial-mediated apoptosis. NAC protects lung injury and apoptosis by inhibiting ROS-mediated activation of JNK and downstream proapoptotic signaling.


Asunto(s)
Hiperoxia/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lesión Pulmonar/metabolismo , Oxidantes/metabolismo , Acetilcisteína/farmacología , Animales , Antracenos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Inhibidores de Caspasas , Línea Celular , Citocromos c/antagonistas & inhibidores , Citocromos c/metabolismo , Células Epiteliales/metabolismo , Hiperoxia/etiología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Volumen de Ventilación Pulmonar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA