Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Pediatr Pulmonol ; 57(7): 1600-1607, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35355453

RESUMEN

INTRODUCTION: Ventilator management is a critical part of managing congenital diaphragmatic hernia (CDH). We aimed to use a murine model and patient data to study CDH-associated differences in oxygenation, airway resistance, and pulmonary mechanics by disease severity. METHODS: We used the nitrofen model of CDH. For control and CDH rodents, data were collected within the first hour of life. Oxygen saturations (SpO2 ) were collected using MouseOx, and large airway resistance and inspiratory capacities were collected using flexiVent. A single-center, retrospective review of term CDH infants from 2014 to 2020 was performed. Tidal volumes were collected every 6 h for the first 48 h of life or until the patient was taken off conventional ventilation. Newborns that were mechanically ventilated but had no pulmonary pathology were used as controls. CDH severity was defined using the CDH Study Group (CDHSG) classification system. RESULTS: Control rodents had a median SpO2 of 94% (IQR: 88%-98%); CDH pups had a median SpO2 of 27.9% (IQR: 22%-30%) (p < 0.01). CDH rodents had lower inspiratory capacity than controls (median: 110 µl, IQR: 70-170 vs. median: 267 µl, IQR: 216-352; p < 0.01). CDH infants had a lower initial SpO2 than control infants. Overall, CDH infants had lower tidal volumes than control infants (median: 4.2 ml/kg, IQR: 3.3-5.0 vs. 5.4 ml/kg, IQR: 4.7-6.2; p = 0.03). Tidal volumes varied by CDHSG stage. CONCLUSION: Newborns with CDH have lower SpO2 and lower, CDHSG stage specific, tidal volumes than control infants. The nitrofen model of CDH reflects these differences. Rodent models may be useful in studying therapeutic ventilatory strategies for CDH infants.


Asunto(s)
Hernias Diafragmáticas Congénitas/terapia , Respiración Artificial , Animales , Animales Recién Nacidos , Humanos , Recién Nacido , Pulmón , Gravedad del Paciente , Estudios Retrospectivos , Roedores , Volumen de Ventilación Pulmonar
3.
Pediatr Pulmonol ; 55(9): 2402-2411, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32568428

RESUMEN

OBJECTIVE: Abnormal pulmonary vasculature directly affects the development and progression of congenital diaphragmatic hernia (CDH)-associated pulmonary hypertension (PH). Though overarching structural and cellular changes in CDH-affected pulmonary arteries have been documented, the precise role of the extracellular matrix (ECM) in the pulmonary artery (PA) pathophysiology remains undefined. Here, we quantify the structural, compositional, and mechanical CDH-induced changes in the main and distal PA ECM and investigate the efficacy of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) as a therapy to ameliorate pathological vascular ECM changes. METHODS: Pregnant Sprague-Dawley rodents were administered nitrofen to induce CDH-affected pulmonary vasculature in the offspring. A portion of CDH-affected pups was treated with intravenous infusion of MSC-EVs (1 × 1010 /mL) upon birth. A suite of histological, mechanical, and transmission electron microscopic analyses were utilized to characterize the PA ECM. RESULTS: The CDH model main PA presented significantly altered characteristics-including greater vessel thickness, greater lysyl oxidase (LOX) expression, and a relatively lower ultimate tensile strength of 13.6 MPa compared to control tissue (25.1 MPa), suggesting that CDH incurs ECM structural disorganization. MSC-EV treatment demonstrated the potential to reverse CDH-related changes, particularly through rapid inhibition of ECM remodeling enzymes (LOX and MMP-9). Additionally, MSC-EV treatment bolstered structural aspects of the PA ECM and mitigated pathological disorganization as exhibited by increased medial wall thickness and stiffness that, while not significantly altered, trends away from CDH-affected tissue. CONCLUSIONS: These data demonstrate notable ECM remodeling in the CDH pulmonary vasculature, along with the capacity of MSC-EVs to attenuate pathological ECM remodeling, identifying MSC-EVs as a potentially efficacious therapeutic for CDH-associated pulmonary hypertension.


Asunto(s)
Matriz Extracelular/patología , Vesículas Extracelulares , Hernias Diafragmáticas Congénitas/patología , Arteria Pulmonar/patología , Animales , Femenino , Hernias Diafragmáticas Congénitas/inducido químicamente , Hernias Diafragmáticas Congénitas/complicaciones , Hernias Diafragmáticas Congénitas/fisiopatología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Pulmón/patología , Intercambio Materno-Fetal , Células Madre Mesenquimatosas , Éteres Fenílicos , Embarazo , Arteria Pulmonar/fisiopatología , Ratas Sprague-Dawley
4.
Stem Cells Dev ; 29(15): 967-980, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32475301

RESUMEN

Congenital diaphragmatic hernia (CDH) leads to pathophysiologic pulmonary vasoreactivity. Previous studies show that mesenchymal stromal cell-derived extracellular vesicles (MSCEv) inhibit lung inflammation and vascular remodeling. We characterize MSCEv and human pulmonary artery endothelial cell (HPAEC) interaction, as well as the pulmonary artery (PA) response to MSCEv treatment. HPAECs were cultured with and without exposure to nitrofen (2,4-dichloro-phenyl-p-nitrophenylether) and treated with MSCEv. HPAEC viability, architecture, production of reactive oxygen species (ROS), endothelial dysfunction-associated protein levels (PPARγ, LOX-1, LOX-2, nuclear factor-κB [NF-κB], endothelial NO synthase [eNOS], ET-1 [endothelin 1]), and the nature of MSCEv-cellular interaction were assessed. Newborn rodents with and without CDH (nitrofen model and Sprague-Dawley) were treated with intravascular MSCEv or vehicle control, and their PAs were isolated. Contractility was assessed by wire myography. The contractile (KCL and ET-1) and relaxation (fasudil) responses were evaluated. HPAEC viability correlated inversely with nitrofen dose, while architectural compromise was directly proportional. There was a 2.1 × increase in ROS levels in nitrofen HPAECs (P < 0.001), and MSCEv treatment attenuated ROS levels by 1.5 × versus nitrofen HPAECs (P < 0.01). Nitrofen-induced alterations in endothelial dysfunction-associated proteins are shown, and exposure to MSCEv restored more physiologic expression. Nitrofen HPAEC displayed greater MSCEv uptake (80% increase, P < 0.05). Adenosine, a clathrin-mediated endocytosis inhibitor, decreased uptake by 46% (P < 0.05). CDH PA contraction was impaired with KCL (108.6% ± 1.4% vs. 112.0% ± 1.4%, P = 0.092) and ET-1 (121.7% ± 3.0% vs. 131.2% ± 1.8%, P < 0.01). CDH PA relaxation was impaired with fasudil (32.2% ± 1.9% vs. 42.1% ± 2.2%, P < 0.001). After MSCEv treatment, CDH PA contraction improved (125.9% ± 3.4% vs. 116.4 ± 3.5, P = 0.06), and relaxation was unchanged (32.5% ± 3.2% vs. 29.4% ± 3.1%, P = 0.496). HPAEC exposure to nitrofen led to changes consistent with vasculopathy in CDH, and MSCEv treatment led to a more physiologic cellular response. MSCEv were preferentially taken up by nitrofen-treated cells by clathrin-dependent endocytosis. In vivo, MSCEv exposure improved PA contractile response. These data reveal mechanisms of cellular and signaling alterations that characterize MSCEv-mediated attenuation of pulmonary vascular dysfunction in CDH-associated pulmonary hypertension.


Asunto(s)
Endotelio/fisiopatología , Vesículas Extracelulares/metabolismo , Hernias Diafragmáticas Congénitas/fisiopatología , Arteria Pulmonar/fisiopatología , Adulto , Animales , Muerte Celular , Clatrina/metabolismo , Endocitosis , Células Endoteliales/metabolismo , Endotelina-1/metabolismo , Endotelio/patología , Femenino , Colorantes Fluorescentes/metabolismo , Hernias Diafragmáticas Congénitas/patología , Humanos , FN-kappa B/metabolismo , Éteres Fenílicos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores Depuradores de Clase E/metabolismo , Vasoconstricción
5.
J Vasc Res ; 55(1): 26-34, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29216632

RESUMEN

Although it is well known that nitrofen induces congenital diaphragmatic hernia (CDH), including CDH-associated lung hypoplasia and pulmonary hypertension (PH) in rodents, the mechanism of pathogenesis remains largely unclear. It has been reported that pulmonary artery (PA) endothelial cell (EC) dysfunction contributes to the development of PH in CDH. Thus, we hypothesized that there is significant alteration of endothelial dysfunction-associated proteins in nitrofen-induced CDH PAs. Pregnant SD rats received either nitrofen or olive oil on gestational day 9.5. The newborn rats were sacrificed and divided into a CDH (n = 81) and a control (n = 23) group. After PA isolation, the expression of PA endothelial dysfunction-associated proteins was assessed on Western blot and immunostaining. We demonstrate that the expression of C-reactive protein and endothelin-1 and its receptors, ETA and ETB, were significantly increased in the CDH PAs. Levels of phosphorylated myosin light chain were significantly elevated, but those of phosphorylated endothelial nitric oxide synthase, caveolin-1, and mechanistic target of rapamycin were significantly decreased in the CDH PAs. In this work, we elucidate alterations in the expression of endothelial dysfunction-associated proteins specific to nitrofen-induced CDH rodent PAs, thereby advancing our understanding of the critical role of endothelial dysfunction-associated pathways in the pathogenesis of nitrofen-induced CDH.


Asunto(s)
Endotelio Vascular/fisiopatología , Hernias Diafragmáticas Congénitas/fisiopatología , Éteres Fenílicos , Arteria Pulmonar/fisiopatología , Animales , Animales Recién Nacidos , Proteínas Portadoras/metabolismo , Caveolina 1/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Edad Gestacional , Hernias Diafragmáticas Congénitas/inducido químicamente , Hernias Diafragmáticas Congénitas/metabolismo , Hernias Diafragmáticas Congénitas/patología , Exposición Materna , Cadenas Ligeras de Miosina/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Embarazo , Efectos Tardíos de la Exposición Prenatal , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Ratas Sprague-Dawley , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Remodelación Vascular
6.
Stem Cells ; 36(1): 79-90, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29076623

RESUMEN

Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been proposed to be a key mechanistic link in the therapeutic efficacy of cells in response to cellular injuries through paracrine effects. We hypothesize that inflammatory stimulation of MSCs results in the release of EVs that have greater anti-inflammatory effects. The present study evaluates the immunomodulatory abilities of EVs derived from inflammation-stimulated and naive MSCs (MSCEv+ and MSCEv, respectively) isolated using a current Good Manufacturing Practice-compliant tangential flow filtration system. Detailed characterization of both EVs revealed differences in protein composition, cytokine profiles, and RNA content, despite similarities in size and expression of common surface markers. MSCEv+ further attenuated release of pro-inflammatory cytokines in vitro when compared to MSCEv, with a distinctly different pattern of EV-uptake by activated primary leukocyte subpopulations. The efficacy of EVs was partially attributed to COX2/PGE2 expression. The present study demonstrates that inflammatory stimulation of MSCs renders release of EVs that have enhanced anti-inflammatory properties partially due to COX2/PGE2 pathway alteration. Stem Cells 2018;36:79-90.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión/métodos , Humanos
7.
Biomed Opt Express ; 7(9): 3184-3197, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27699091

RESUMEN

Multimodal imaging probes a variety of tissue properties in a single image acquisition by merging complimentary imaging technologies. Exploiting synergies amongst the data, algorithms can be developed that lead to better tissue characterization than could be accomplished by the constituent imaging modalities taken alone. The combination of optical coherence tomography (OCT) with fluorescence lifetime imaging microscopy (FLIM) provides access to detailed tissue morphology and local biochemistry. The optical system described here merges 1310 nm swept-source OCT with time-domain FLIM having excitation at 355 and 532 nm. The pulses from 355 and 532 nm lasers have been interleaved to enable simultaneous acquisition of endogenous and exogenous fluorescence signals, respectively. The multimodal imaging system was validated using tissue phantoms. Nonspecific tagging with Alexa Flour 532 in a Watanbe rabbit aorta and active tagging of the LOX-1 receptor in human coronary artery, demonstrate the capacity of the system for simultaneous acquisition of OCT, endogenous FLIM, and exogenous FLIM in tissues.

8.
Amino Acids ; 47(10): 2089-99, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26354329

RESUMEN

Autophagy (i.e., "self-eating") and apoptosis (i.e., type I programmed cell death) are essential and intimately involved in molecular, cellular, and whole-body homeostasis in humans and animals. Autophagy has been categorized as a mechanism of intracellular degradation, recycling, defense, and survival. To date, three types of autophagy have been identified: macroautophagy, microautophagy, and chaperone-mediated autophagy. Recent discoveries strongly suggest that macroautophagy also modulates type II programmed cell death under specific circumstances. Autophagy and apoptosis are fundamentally distinct processes, but are interconnected by common stress initiators and intermediate regulators. During the past two decades, the role of amino acid metabolism and signaling in the regulation of apoptosis and autophagy has been intensively studied. In this review, we summarize recent advances in our understanding of the molecular mechanisms that regulate both autophagy and apoptosis in the context of amino acid signaling.


Asunto(s)
Aminoácidos/metabolismo , Apoptosis , Autofagia , Comunicación Celular , Transducción de Señal , Animales , Humanos
9.
J Liposome Res ; 24(3): 182-90, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24443972

RESUMEN

Liposomes have been used to diagnose and treat cancer and, to a lesser extent, cardiovascular disease. We previously showed the uptake of anionic liposomes into the atheromas of Watanabe heritable hyperlipidemic rabbits within lipid pools. However, the cellular distribution of anionic liposomes in atherosclerotic plaque remains undescribed. In addition, how anionic liposomes are absorbed into atherosclerotic plaque is unclear. We investigated the uptake and distribution of anionic liposomes in atherosclerotic plaque in aortic tissues from apolipoprotein E-deficient (ApoE(-/-)) mice. To facilitate the tracking of liposomes, we used liposomes containing fluorescently labeled non-silencing small interfering RNA. Confocal microscopy analysis showed the uptake of anionic liposomes into atherosclerotic plaque and colocalization with macrophages. Transmission electron microscopy analysis revealed anionic liposomal accumulation in macrophages. To investigate how anionic liposomes cross the local endothelial barrier, we examined the role of clathrin-mediated endocytosis in human coronary artery endothelial cells (HCAECs) treated with or without the inflammatory cytokine tumor necrosis factor (TNF)-α. Pretreatment with amantadine, an inhibitor of clathrin-mediated endocytosis, significantly decreased liposomal uptake in HCAECs treated with or without TNF-α by 77% and 46%, respectively. Immunoblot analysis showed that endogenous clathrin expression was significantly increased in HCAECs stimulated with TNF-α but was inhibited by amantadine. These studies indicated that clathrin-mediated endocytosis is partly responsible for the uptake of liposomes by endothelial cells. Our results suggest that anionic liposomes target macrophage-rich areas of vulnerable plaque in ApoE(-)(/)(-) mice; this finding may lead to the development of novel diagnostic and therapeutic strategies for treating vulnerable plaque in humans.


Asunto(s)
Liposomas/metabolismo , Placa Aterosclerótica/fisiopatología , Amantadina/farmacología , Animales , Aniones/administración & dosificación , Aorta/patología , Apolipoproteínas E/deficiencia , Clatrina/biosíntesis , Clatrina/fisiología , Endocitosis/efectos de los fármacos , Endotelio Vascular , Humanos , Liposomas/administración & dosificación , Macrófagos/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
10.
Prostate ; 72(13): 1412-22, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22294520

RESUMEN

BACKGROUND: Androgen deprivation therapy, one of the standard treatments for prostate cancer (PCa) induces apoptosis, as well as autophagy in androgen-responsive PCa cells. As autophagy can promote either cell survival or death, it is important to understand its role in PCa treatment. The objective of this study was to elucidate the function of autophagy in lipid droplet (LD) homeostasis and survival in androgen-sensitive PCa cells. METHODS: To produce androgen deprivation, charcoal filtered serum or the androgen inhibitor casodex were used in LNCaP and LAPC4 cells. Autophagy was monitored by immunofluorescence/confocal microscopy and immunoblot analysis. Levels of intracellular LDs and triacyglycerols after the inhibition of autophagy by 3-methyladenine, bafilomycin A(1) , or si-ATG5 were quantified by three independent methods, Oil Red O staining, triacyglycerols lipase assay, and nuclear magnetic resonance. RESULTS: Androgen deprivation induced autophagy and the depletion of LDs in both of the androgen-sensitive PCa cell lines examined, whereas the blockage of autophagy by pharmacological or genetic means inhibited LD degradation and therefore lipolysis and cell growth. In addition, under androgen deprivation, increased colocalization of LDs and autophagic vesicles was observed in LNCaP cells, which can be further enhanced by blocking the autophagic flux. CONCLUSION: Autophagy mediates LD degradation and lipolysis in androgen-sensitive PCa cells during androgen deprivation which aids the survival of PCa cells during hormone therapy.


Asunto(s)
Autofagia/fisiología , Supervivencia Celular/fisiología , Metabolismo de los Lípidos/fisiología , Lipólisis/fisiología , Neoplasias de la Próstata/metabolismo , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Anilidas/farmacología , Anilidas/uso terapéutico , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Masculino , Neoplasias Hormono-Dependientes , Nitrilos/farmacología , Nitrilos/uso terapéutico , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Compuestos de Tosilo/farmacología , Compuestos de Tosilo/uso terapéutico , Células Tumorales Cultivadas
11.
J Biol Chem ; 286(31): 27389-98, 2011 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-21646352

RESUMEN

Inflammatory cytokine-regulated apoptosis and autophagy play pivotal roles in plaque rupture and thrombosis of atherosclerotic lesions. However, the molecular interplay between apoptosis and autophagy in vascular cells has not been investigated. Our prior study showed that human apolipoprotein L6 (ApoL6), a pro-apoptotic BH3-only member of the Bcl-2 family, was one of the downstream targets of interferon-γ (INFγ), which sensitizes atherosclerotic lesion-derived cells (LDCs) to Fas-induced apoptosis. To investigate whether ApoL6 plays a causal role in atherosclerotic apoptosis and autophagy, in this study, we demonstrate that IFNγ treatment itself strongly induces ApoL6, and ApoL6 is highly expressed and partially co-localized with activated caspase 3 in activated smooth muscle cells in atherosclerotic lesions. In addition, overexpression of ApoL6 promotes reactive oxygen species (ROS) generation, caspase activation, and subsequent apoptosis, which can be blocked by pan caspase inhibitor and ROS scavenger. Knockdown of ApoL6 expression by siApoL6 suppresses INFγ- and Fas-mediated apoptosis. Further, ApoL6 binds Bcl-X(L), one of the most abundant anti-death proteins in LDCs. Interestingly, forced ApoL6 expression in LDCs induces degradation of Beclin 1, accumulation of p62, and subsequent attenuation of LC3-II formation and translocation and thus autophagy, whereas siApoL6 treatment reverts the phenotype. Taken together, our results suggest that ApoL6 regulates both apoptosis and autophagy in SMCs. IFNγ-initiated, ApoL6-induced apoptosis in vascular cells may be an important factor causing plaque instability and a potential therapeutic target for treating atherosclerosis and cardiovascular disease.


Asunto(s)
Apolipoproteínas/fisiología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Aterosclerosis/metabolismo , Autofagia/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Antioxidantes/farmacología , Apolipoproteínas/biosíntesis , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Apolipoproteínas L , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Aterosclerosis/inmunología , Beclina-1 , Células Cultivadas , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/fisiología , Unión Proteica , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína bcl-X/metabolismo
12.
Autophagy ; 4(8): 1079-82, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18927493

RESUMEN

We recently reported the identification and characterization of a novel BH3-only pro-death protein, apolipoprotein L1 (ApoL1), that, when overexpressed, induces autophagic cell death (ACD) in a variety of cells, including those originated from normal and cancerous tissues. ApoL1 failed to induce ACD in autophagy-deficient Atg5(-/-) and Atg7(-/-) MEF cells, suggesting that ApoL1-induced cell death is indeed autophagy-dependent. In addition, a BH3 domain deletion allele of ApoL1 was unable to induce ACD, demonstrating that ApoL1 is a bona fide BH3-only pro-death protein. To further investigate regulation of ApoL1 expression, we showed that ApoL1 is inducible by interferon-gamma and tumor necrosis factor-alpha in human umbilical vein endothelial cells, suggesting that ApoL1 may play a role in cytokine-induced inflammatory response. Moreover, we observed that ApoL1 is a lipid-binding protein with high affinity for phosphatidic acid and cardiolipin and less affinity for various phosphoinositides. Functional genomics analysis identified 5 nonsynonymous single nucleotide polymorphisms (NSNPs) in the coding exons of the human ApoL1 structural gene-all the 5 NSNPs may cause deleterious alteration of ApoL1 activity. Finally, we discuss the link between ApoL1 and various human diseases.


Asunto(s)
Apolipoproteínas/metabolismo , Autofagia , Lipoproteínas HDL/metabolismo , Secuencia de Aminoácidos , Apolipoproteína L1 , Apolipoproteínas/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Metabolismo de los Lípidos , Lipoproteínas HDL/genética , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/metabolismo , Tripanosomiasis Africana/patología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/metabolismo
13.
Amino Acids ; 35(4): 655-64, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18506409

RESUMEN

Proline metabolism in mammals involves two other amino acids, glutamate and ornithine, and five enzymatic activities, Delta(1)-pyrroline-5-carboxylate (P5C) reductase (P5CR), proline oxidase, P5C dehydrogenase, P5C synthase and ornithine-delta-aminotransferase (OAT). With the exception of OAT, which catalyzes a reversible reaction, the other four enzymes are unidirectional, suggesting that proline metabolism is purpose-driven, tightly regulated, and compartmentalized. In addition, this tri-amino-acid system also links with three other pivotal metabolic systems, namely the TCA cycle, urea cycle, and pentose phosphate pathway. Abnormalities in proline metabolism are relevant in several diseases: six monogenic inborn errors involving metabolism and/or transport of proline and its immediate metabolites have been described. Recent advances in the Human Genome Project, in silico database mining techniques, and research in dissecting the molecular basis of proline metabolism prompted us to utilize functional genomic approaches to analyze human genes which encode proline metabolic enzymes in the context of gene structure, regulation of gene expression, mRNA variants, protein isoforms, and single nucleotide polymorphisms.


Asunto(s)
Apoptosis , Genómica , Polimorfismo de Nucleótido Simple , Prolina/metabolismo , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica , Genoma Humano , Humanos , Modelos Biológicos , Ornitina-Oxo-Ácido Transaminasa/metabolismo , Prolina Oxidasa/genética , Prolina Oxidasa/metabolismo , Elementos de Respuesta , Factores de Tiempo , Distribución Tisular , Urea/metabolismo
14.
J Biol Chem ; 283(31): 21540-9, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18505729

RESUMEN

The Bcl-2 family proteins are important regulators of type I programmed cell death apoptosis; however, their role in autophagic cell death (AuCD) or type II programmed cell death is still largely unknown. Here we report the cloning and characterization of a novel Bcl-2 homology domain 3 (BH3)-only protein, apolipoprotein L1 (apoL1), that, when overexpressed and accumulated intracellularly, induces AuCD in cells as characterized by the increasing formation of autophagic vacuoles and activating the translocation of LC3-II from the cytosol to the autophagic vacuoles. Wortmannin and 3-methyladenine, inhibitors of class III phosphatidylinostol 3-kinase and, subsequently, autophagy, blocked apoL1-induced AuCD. In addition, apoL1 failed to induce AuCD in autophagy-deficient ATG5(-/-) and ATG7(-/-) mouse embryonic fibroblast cells, suggesting that apoL1-induced cell death is indeed autophagy-dependent. Furthermore, a BH3 domain deletion construct of apoL1 failed to induce AuCD, demonstrating that apoL1 is a bona fide BH3-only pro-death protein. Moreover, we showed that apoL1 is inducible by p53 in p53-induced cell death and is a lipid-binding protein with high affinity for phosphatidic acid (PA) and cardiolipin (CL). Previously, it has been shown that PA directly interacted with mammalian target of rapamycin and positively regulated the ability of mammalian target of rapamycin to activate downstream effectors. In addition, CL has been shown to activate mitochondria-mediated apoptosis. Sequestering of PA and CL with apoL1 may alter the homeostasis between survival and death leading to AuCD. To our knowledge, this is the first BH3-only protein with lipid binding activity that, when overproduced intracellularly, induces AuCD.


Asunto(s)
Apolipoproteínas/fisiología , Lipoproteínas HDL/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Secuencia de Aminoácidos , Apolipoproteína L1 , Apolipoproteínas/metabolismo , Apoptosis , Autofagia , Línea Celular Tumoral , Humanos , Lípidos/química , Lipoproteínas HDL/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Conformación Molecular , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido , Serina-Treonina Quinasas TOR
15.
Oncol Rep ; 10(3): 537-43, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12684620

RESUMEN

This study was conducted to assess protective effect of an antioxidant protein, sericin, on tumor promotion in the 7,12-dimethylbenz (alpha) anthracene (DMBA)-initiated and 12-O-tetradecanoylphorbol 13-acetate (TPA)-promoted mouse skin tumorigenesis model. In experiment 1, sericin was applied topically to DMBA-initiated female ICR mouse skin at the doses of 2.5 and 5 mg twice per week for 16 weeks, 30 min prior to each promotion treatment with TPA. The protective effect of sericin was evident in terms of significant reduction in tumor incidence and tumor multiplicity at the doses of 2.5 and 5 mg per application, compared to the control group without receiving sericin. The expression of tumor necrosis factor (TNF)-alpha protein and the level of 4-hydroxynonenal (4-HNE) in normal epidermis were significantly reduced in both sericin treatment groups. In experiment 2, sericin at the dose of 5 mg was applied topically to the dorsal mouse skin 30 min before application of a TPA, and the same doses of TPA and sericin were applied twice at an interval of 24 h. Sericin treatment inhibited double TPA treatment-induced morphological changes reflecting inflammatory response, including leukocyte infiltration, hyperplasia and cell proliferation. Furthermore, sericin treatment significantly suppressed the elevation in 4-HNE level and elevated expressions of c-fos, c-myc and cyclooxygenase-2 (COX-2) in normal epidermis induced by double application of TPA. The results suggest that sericin possesses protective effect against tumor promotion in mouse skin by suppressing oxidative stress, inflammatory responses and TNF-alpha.


Asunto(s)
Anticarcinógenos/farmacología , Estrés Oxidativo/efectos de los fármacos , Péptidos Cíclicos/farmacología , Neoplasias Cutáneas/prevención & control , Factor de Necrosis Tumoral alfa/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Aldehídos/metabolismo , Animales , Carcinógenos/toxicidad , Ciclooxigenasa 2 , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Epidermis/patología , Femenino , Genes fos/fisiología , Genes myc/fisiología , Hiperplasia , Inflamación/patología , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos ICR , Monofenol Monooxigenasa/antagonistas & inhibidores , Prostaglandina-Endoperóxido Sintasas/metabolismo , Sericinas , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/metabolismo , Acetato de Tetradecanoilforbol/toxicidad
16.
J Photochem Photobiol B ; 71(1-3): 11-7, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14705634

RESUMEN

This study was conducted to assess protective effect of an antioxidant protein, sericin, on UVB-induced acute damage and tumor promotion in mouse skin. In experiment 1, HR-1 hairless mice were treated with 180 mJ/cm2 of ultraviolet B light (UVB) once daily for 1 and 7 days. The treatment for 7 days caused red sunburn lesions of the skin. The intensity of red color and area of these lesions were inhibited by the topical application of sericin at the dose of 5 mg after UVB treatment. Immunohistochemical analyses showed that the application of sericin significantly suppressed UVB-induced elevations in 4-hydroxynonenal (4-HNE), expression of cyclooxygenase-2 (COX-2) protein, and proliferating cell nuclear antigen (PCNA)-labeling index in the UVB-exposed epidermis. In experiment 2, HR-1 hairless mice were treated with 200 nmol of 7,12-dimethylbenz [alpha] anthracene (DMBA) followed 1 week later by irradiation with 180 mJ/ cm2 of UVB twice weekly for 22 weeks. The protective effect of sericin was evident in terms of significant reduction in tumor incidence and tumor multiplicity at the dose of 5 mg. The results suggest that sericin possesses photoprotective effect against UVB-induced acute damage and tumor promotion by reducing oxidative stress, COX-2 and cell proliferation in mouse skin.


Asunto(s)
Estrés Oxidativo , Péptidos Cíclicos/fisiología , Neoplasias Cutáneas/prevención & control , Rayos Ultravioleta , Animales , Ratones , Ratones Pelados , Sericinas , Neoplasias Cutáneas/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA