Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Biosci Bioeng ; 131(2): 207-212, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33051155

RESUMEN

Finding peaks in chromatograms and determining their start and end points (peak picking) is a core task in chromatography based biotechnology. Construction of peak-picking neural networks by deep learning was, however, hampered from the preparation of exact peak-picked or "labeled" chromatograms since the exact start and end points were often unclear in overlapping peaks in real chromatograms. We present a design of a fake chromatogram generator, along with a method for deep learning of peak-picking neural networks. Fake chromatograms were generated by generation of fake peaks, random sampling of peak positions from feature distributions, and merging with real blank sample chromatograms. Information on the exact start and end points, as labeled on the fake chromatograms, were effective for training and evaluating peak-picking neural networks. The peak-picking neural networks constructed herein outperformed conventional peak-picking software and showed comparable performance with that of experienced operators for processing the widely targeted metabolome data. Results of this study indicate that generation of fake chromatograms would be crucial for developing peak-picking neural networks and a key technology for further improvement of peak picking neural networks.


Asunto(s)
Aprendizaje Profundo , Metabolómica/métodos , Cromatografía , Programas Informáticos
2.
Int J Mol Sci ; 20(5)2019 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-30832329

RESUMEN

Pericytes are mesenchymal cells that surround the endothelial cells of small vessels in various organs. These cells express several markers, such as NG2, CD146, and PDGFRß, and play an important role in the stabilization and maturation of blood vessels. It was also recently revealed that like mesenchymal stem cells (MSCs), pericytes possess multilineage differentiation capacity, especially myogenic, adipogenic, and fibrogenic differentiation capacities. Although some previous studies have reported that pericytes also have osteogenic potential, the osteogenesis of pericytes can still be further elucidated. In the present study, we established novel methods for isolating and culturing primary murine pericytes. An immortalized pericyte line was also established. Multilineage induction of the pericyte line induced osteogenesis, adipogenesis, and chondrogenesis of the cells in vitro. In addition, pericytes that were injected into the fracture site of a bone fracture mouse model contributed to callus formation. Furthermore, in vivo pericyte-lineage-tracing studies demonstrated that endogenous pericytes also differentiate into osteoblasts and osteocytes and contribute to bone fracture healing as a cellular source of osteogenic cells. Pericytes can be a promising therapeutic candidate for treating bone fractures with a delayed union or nonunion as well as bone diseases causing bone defects.


Asunto(s)
Condrogénesis , Curación de Fractura , Osteogénesis , Pericitos/citología , Cultivo Primario de Células/métodos , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Condrocitos/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Ratones Endogámicos BALB C , Osteoblastos/citología , Pericitos/trasplante
3.
Bio Protoc ; 9(3): e3151, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33654960

RESUMEN

Bone is one of common metastasis sites for many types of cancer. In bone metastatic microenvironment, tumor-bone interactions play a significant role in the regulation of osteolytic or osteoblastic bone metastasis. In order to investigate the direct interaction between tumor cells and bone tissue, it is essential to generate appropriate animal models that mimic the behavior of tumor cells in bone metastatic lesions. Calvarial implantation model (bone invasion model) is a newly-established animal model that accurately recapitulates the behavior of tumor cells in the tumor-bone microenvironment. The surgical technique for tumor cell implantation is simpler than intracardiac, intra-arterial, or intraosseous injection techniques. This model can be useful for the identification of key factors driving tumor-induced osteolytic or osteoblastic changes.

4.
Nat Metab ; 1(9): 868-875, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-32694743

RESUMEN

Receptor activator of NF-κB ligand (RANKL) is a multifunctional cytokine known to affect immune and skeletal systems, as well as oncogenesis and metastasis1-4. RANKL is synthesized as a membrane-bound molecule, and cleaved into its soluble form by proteases5-7. As the soluble form of RANKL does not contribute greatly to bone remodelling or ovariectomy-induced bone loss8, whether soluble RANKL has a role in pathological settings remains unclear. Here we show that soluble RANKL promotes the formation of tumour metastases in bone. Mice that selectively lack soluble RANKL (Tnfsf11ΔS/ΔS)5-7,9 have normal bone homoeostasis and develop a normal immune system but display markedly reduced numbers of bone metastases after intracardiac injection of RANK-expressing melanoma and breast cancer cells. Deletion of soluble RANKL does not affect osteoclast numbers in metastatic lesions or tumour metastasis to non-skeletal tissues. Therefore, soluble RANKL is dispensable for physiological regulation of bone and immune systems, but has a distinct and pivotal role in the promotion of bone metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias/patología , Receptor Activador del Factor Nuclear kappa-B/fisiología , Animales , Remodelación Ósea/fisiología , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Osteoclastos/citología , Receptor Activador del Factor Nuclear kappa-B/genética
5.
Proc Natl Acad Sci U S A ; 115(9): 2204-2209, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29440427

RESUMEN

Bone metastatic lesions are classified as osteoblastic or osteolytic lesions. Prostate and breast cancer patients frequently exhibit osteoblastic-type and osteolytic-type bone metastasis, respectively. In metastatic lesions, tumor cells interact with many different cell types, including osteoblasts, osteoclasts, and mesenchymal stem cells, resulting in an osteoblastic or osteolytic phenotype. However, the mechanisms responsible for the modification of bone remodeling have not been fully elucidated. MicroRNAs (miRNAs) are transferred between cells via exosomes and serve as intercellular communication tools, and numerous studies have demonstrated that cancer-secreted miRNAs are capable of modifying the tumor microenvironment. Thus, cancer-secreted miRNAs can induce an osteoblastic or osteolytic phenotype in the bone metastatic microenvironment. In this study, we performed a comprehensive expression analysis of exosomal miRNAs secreted by several human cancer cell lines and identified eight types of human miRNAs that were highly expressed in exosomes from osteoblastic phenotype-inducing prostate cancer cell lines. One of these miRNAs, hsa-miR-940, significantly promoted the osteogenic differentiation of human mesenchymal stem cells in vitro by targeting ARHGAP1 and FAM134A Interestingly, although MDA-MB-231 breast cancer cells are commonly known as an osteolytic phenotype-inducing cancer cell line, the implantation of miR-940-overexpressing MDA-MB-231 cells induced extensive osteoblastic lesions in the resulting tumors by facilitating the osteogenic differentiation of host mesenchymal cells. Our results suggest that the phenotypes of bone metastases can be induced by miRNAs secreted by cancer cells in the bone microenvironment.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias de la Mama/patología , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/metabolismo , Animales , Neoplasias Óseas/secundario , Sustitutos de Huesos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Proteínas Activadoras de GTPasa/genética , Humanos , Masculino , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas , Ratones , MicroARNs/genética , Neoplasias Experimentales/metabolismo
6.
Autophagy ; 11(9): 1520-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26102061

RESUMEN

Sepsis/endotoxemia is elicited by the circulatory distribution of pathogens/endotoxins into whole bodies, and causes profound effects on human health by causing inflammation in multiple organs. Mitochondrial damage is one of the characteristics of the cellular degeneration observed during sepsis/endotoxemia. Elimination of damaged mitochondria through the autophagy-lysosome system has been reported in the liver, indicating that autophagy should play an important role in liver homeostasis during sepsis/endotoxemia. An increased appearance of mitochondrial DNA and proteins in the plasma is another feature of sepsis/endotoxemia, suggesting that damaged mitochondria are not only eliminated within the cells, but also extruded through currently unknown mechanisms. Here we provide evidence for the secretion of mitochondrial proteins and DNA from lipopolysaccharide (LPS)-stimulated rat hepatocytes as well as mouse embryonic fibroblasts (MEFs). The secretion of mitochondrial contents is accompanied by the secretion of proteins that reside in the lumenal space of autolysosomes (LC3-II and CTSD/cathepsin D), but not by a lysosomal membrane protein (LAMP1). The pharmacological inhibition of autophagy by 3MA blocks the secretion of mitochondrial constituents from LPS-stimulated hepatocytes. LPS also stimulates the secretion of mitochondrial as well as autolysosomal lumenal proteins from wild-type (Atg5(+/+)) MEFs, but not from atg5(-/-) MEFs. Furthermore, we show that direct exposure of purified mitochondria activates polymorphonuclear leukocytes (PMNs), as evident by the induction of IL1B/interlekin-1ß, a pro-inflammatory cytokine. Taken together, the data suggest the active extrusion of mitochondrial contents, which provoke an inflammatory response of immune cells, through the exocytosis of autolysosomes by cells stimulated with LPS.


Asunto(s)
Autofagia/efectos de los fármacos , Lipopolisacáridos/farmacología , Mitocondrias/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Autofagia/genética , Proteína 5 Relacionada con la Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , ADN Mitocondrial/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Lisosomas/efectos de los fármacos , Lisosomas/genética , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Activación Neutrófila/efectos de los fármacos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo
7.
Cell ; 158(1): 84-97, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995980

RESUMEN

DNA transcription, replication, and repair are regulated by histone acetylation, a process that requires the generation of acetyl-coenzyme A (CoA). Here, we show that all the subunits of the mitochondrial pyruvate dehydrogenase complex (PDC) are also present and functional in the nucleus of mammalian cells. We found that knockdown of nuclear PDC in isolated functional nuclei decreased the de novo synthesis of acetyl-CoA and acetylation of core histones. Nuclear PDC levels increased in a cell-cycle-dependent manner and in response to serum, epidermal growth factor, or mitochondrial stress; this was accompanied by a corresponding decrease in mitochondrial PDC levels, suggesting a translocation from the mitochondria to the nucleus. Inhibition of nuclear PDC decreased acetylation of specific lysine residues on histones important for G1-S phase progression and expression of S phase markers. Dynamic translocation of mitochondrial PDC to the nucleus provides a pathway for nuclear acetyl-CoA synthesis required for histone acetylation and epigenetic regulation.


Asunto(s)
Acetilcoenzima A/biosíntesis , Núcleo Celular/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/enzimología , Epigénesis Genética , Histonas/metabolismo , Humanos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Transporte de Proteínas
8.
Am J Respir Crit Care Med ; 180(11): 1131-42, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19713449

RESUMEN

RATIONALE: Bronchopulmonary dysplasia (BPD) and emphysema are characterized by arrested alveolar development or loss of alveoli; both are significant global health problems and currently lack effective therapy. Bone marrow-derived mesenchymal stem cells (BMSCs) prevent adult lung injury, but their therapeutic potential in neonatal lung disease is unknown. OBJECTIVES: We hypothesized that intratracheal delivery of BMSCs would prevent alveolar destruction in experimental BPD. METHODS: In vitro, BMSC differentiation and migration were assessed using co-culture assays and a modified Boyden chamber. In vivo, the therapeutic potential of BMSCs was assessed in a chronic hyperoxia-induced model of BPD in newborn rats. MEASUREMENTS AND MAIN RESULTS: In vitro, BMSCs developed immunophenotypic and ultrastructural characteristics of type II alveolar epithelial cells (AEC2) (surfactant protein C expression and lamellar bodies) when co-cultured with lung tissue, but not with culture medium alone or liver. Migration assays revealed preferential attraction of BMSCs toward oxygen-damaged lung versus normal lung. In vivo, chronic hyperoxia in newborn rats led to air space enlargement and loss of lung capillaries, and this was associated with a decrease in circulating and resident lung BMSCs. Intratracheal delivery of BMSCs on Postnatal Day 4 improved survival and exercise tolerance while attenuating alveolar and lung vascular injury and pulmonary hypertension. Engrafted BMSCs coexpressed the AEC2-specific marker surfactant protein C. However, engraftment was disproportionately low for cell replacement to account for the therapeutic benefit, suggesting a paracrine-mediated mechanism. In vitro, BMSC-derived conditioned medium prevented O(2)-induced AEC2 apoptosis, accelerated AEC2 wound healing, and enhanced endothelial cord formation. CONCLUSIONS: BMSCs prevent arrested alveolar and vascular growth in part through paracrine activity. Stem cell-based therapies may offer new therapeutic avenues for lung diseases that currently lack efficient treatments.


Asunto(s)
Lesión Pulmonar/prevención & control , Células Madre Mesenquimatosas , Alveolos Pulmonares/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Médula Ósea , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Tolerancia al Ejercicio , Hiperoxia , Hipertensión Pulmonar/prevención & control , Alveolos Pulmonares/ultraestructura , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia
9.
Antimicrob Agents Chemother ; 52(4): 1318-24, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18227186

RESUMEN

The in vitro and in vivo antifungal activities of T-2307, a novel arylamidine, were evaluated and compared with those of fluconazole, voriconazole, micafungin, and amphotericin B. T-2307 exhibited broad-spectrum activity against clinically significant pathogens, including Candida species (MIC range, 0.00025 to 0.0078 microg/ml), Cryptococcus neoformans (MIC range, 0.0039 to 0.0625 microg/ml), and Aspergillus species (MIC range, 0.0156 to 4 microg/ml). Furthermore, T-2307 exhibited potent activity against fluconazole-resistant and fluconazole-susceptible-dose-dependent Candida albicans strains as well as against azole-susceptible strains. T-2307 exhibited fungicidal activity against some Candida and Aspergillus species and against Cryptococcus neoformans. In mouse models of disseminated candidiasis, cryptococcosis, and aspergillosis, the 50% effective doses of T-2307 were 0.00755, 0.117, and 0.391 mg.kg(-1).dose(-1), respectively. This agent was considerably more active than micafungin and amphotericin B against candidiasis and than amphotericin B against cryptococcosis, and its activity was comparable to the activities of micafungin and amphotericin B against aspergillosis. The results of preclinical in vitro and in vivo evaluations performed thus far indicate that T-2307 could represent a potent injectable agent for the treatment of candidiasis, cryptococcosis, and aspergillosis.


Asunto(s)
Antifúngicos/farmacología , Aspergillus fumigatus/efectos de los fármacos , Benzamidinas/farmacología , Candida albicans/efectos de los fármacos , Cryptococcus neoformans/efectos de los fármacos , Micosis/tratamiento farmacológico , Animales , Antifúngicos/química , Antifúngicos/uso terapéutico , Aspergilosis/tratamiento farmacológico , Aspergilosis/microbiología , Benzamidinas/química , Benzamidinas/uso terapéutico , Candidiasis/tratamiento farmacológico , Candidiasis/microbiología , Criptococosis/tratamiento farmacológico , Criptococosis/microbiología , Masculino , Ratones , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Micosis/microbiología , Organismos Libres de Patógenos Específicos , Resultado del Tratamiento
10.
Pediatr Res ; 63(2): 176-81, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18091358

RESUMEN

Patent ductus arteriosus (PDA) complicates the hospital course of premature infants. Impaired oxygen (O2)-induced vasoconstriction in preterm ductus arteriosus (DA) contributes to PDA and results, in part, from decreased function/expression of O2-sensitive, voltage-gated potassium channels (Kv) in DA smooth muscle cells (DASMCs). This paradigm suggests that activation of the voltage-sensitive L-type calcium channels (CaL), which increases cytosolic calcium ([Ca2+]i), is a passive consequence of membrane depolarization. However, effective Kv gene transfer only partially matures O2 responsiveness in preterm DA. Thus, we hypothesized that CaL are directly O2 sensitive and that immaturity of CaL function in preterm DA contributes to impaired O2 constriction. We show that preterm rabbit DA rings have reduced O2- and 4-aminopyridine (Kv blocker)-induced constriction. Preterm rabbit DASMCs have reduced O2-induced whole-cell calcium current (ICa) and [Ca2+]i. BAY K8644, a CaL activator, increased O2 constriction, ICa, and [Ca]i in preterm DASMCs to levels seen at term but had no effect on human and rabbit term DA. Preterm rabbit DAs have decreased gamma and increased alpha subunit protein expression. We conclude that the CaL in term rabbit and human DASMCs is directly O2 sensitive. Functional immaturity of CaL O2 sensitivity contributes to impaired O2 constriction in premature DA and can be reversed by BAY K8644.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Conducto Arterial/metabolismo , Miocitos del Músculo Liso/citología , Oxígeno/metabolismo , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Conducto Arterioso Permeable/etiología , Electrofisiología , Humanos , Modelos Biológicos , Técnicas de Placa-Clamp , Conejos , Vasoconstricción
11.
Proc Natl Acad Sci U S A ; 104(27): 11418-23, 2007 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-17596340

RESUMEN

In pulmonary arterial hypertension (PAH), antiapoptotic, proliferative, and inflammatory diatheses converge to create an obstructive vasculopathy. A selective down-regulation of the Kv channel Kv1.5 has been described in human and animal PAH. The resultant increase in intracellular free Ca(2+) ([Ca(2+)](i)) and K(+) ([K(+)](i)) concentrations explains the pulmonary artery smooth muscle cell (PASMC) contraction, proliferation and resistance to apoptosis. The recently described PASMC hyperpolarized mitochondria and increased bcl-2 levels also contribute to apoptosis resistance in PAH. The cause of the Kv1.5, mitochondrial, and inflammatory abnormalities remains unknown. We hypothesized that these abnormalities can be explained in part by an activation of NFAT (nuclear factor of activated T cells), a Ca(2+)/calcineurin-sensitive transcription factor. We studied PASMC and lungs from six patients with and four without PAH and blood from 23 PAH patients and 10 healthy volunteers. Compared with normal, PAH PASMC had decreased Kv current and Kv1.5 expression and increased [Ca(2+)](i), [K(+)](i), mitochondrial potential (Delta Psi m), and bcl-2 levels. PAH but not normal PASMC and lungs showed activation of NFATc2. Inhibition of NFATc2 by VIVIT or cyclosporine restored Kv1.5 expression and current, decreased [Ca(2+)](i), [K(+)](i), bcl-2, and Delta Psi m, leading to decreased proliferation and increased apoptosis in vitro. In vivo, cyclosporine decreased established rat monocrotaline-PAH. NFATc2 levels were increased in circulating leukocytes in PAH versus healthy volunteers. CD3-positive lymphocytes with activated NFATc2 were seen in the arterial wall in PAH but not normal lungs. The generalized activation of NFAT in human and experimental PAH might regulate the ionic, mitochondrial, and inflammatory remodeling and be a therapeutic target and biomarker.


Asunto(s)
Marcación de Gen , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/terapia , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/genética , Arteria Pulmonar/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Canales de Calcio/biosíntesis , Línea Celular , Ciclosporina/administración & dosificación , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/patología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/biosíntesis , Canal de Potasio Kv1.5/genética , Pulmón/irrigación sanguínea , Pulmón/metabolismo , Pulmón/patología , Masculino , Tamaño Mitocondrial/efectos de los fármacos , Tamaño Mitocondrial/fisiología , Monocrotalina/administración & dosificación , Factores de Transcripción NFATC/fisiología , Oligopéptidos/administración & dosificación , Arteria Pulmonar/patología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
12.
Circulation ; 115(13): 1777-88, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17353442

RESUMEN

BACKGROUND: Constriction of the ductus arteriosus (DA) is initiated at birth by inhibition of O2-sensitive K+ channels in DA smooth muscle cells. Subsequent membrane depolarization and calcium influx through L-type calcium channels initiates functional closure. We hypothesize that Rho-kinase activation is an additional mechanism that sustains DA constriction. METHODS AND RESULTS: The effect of increased PO2 on the activity and expression of Rho-kinase was assessed in DAs from neonates with hypoplastic left-heart syndrome (n=15) and rabbits (339 term and 99 preterm rabbits). Rho-kinase inhibitors (Y-27632 and fasudil) prevent and reverse O2 constriction. Heterogeneity exists in the sensitivity of constrictors (PO2=endothelin=phenylephrine>KCl) and of fetal vessels (DA=pulmonary artery>aorta) to Rho-kinase inhibition. Inhibition of L-type calcium channels (nifedipine) or removal of extracellular calcium inhibits approximately two thirds of O2 constriction. Residual DA constriction reflects calcium sensitization, which persists after removal of extracellular calcium and blocking of sarcoplasmic reticulum Ca2+-ATPase. In term DA, an increase in PO2 activates Rho-kinase and thereby increases RhoB and ROCK-1 expression. Activation of Rho-kinase in DA smooth muscle cells is initiated by a PO2-dependent, rotenone-sensitive increase in mitochondrion-derived reactive O2 species. O2 effects on Rho-kinase are mimicked by exogenous H2O2. In preterm DAs, immaturity of mitochondrial reactive oxygen species generation is associated with reduced and delayed O2 constriction and lack of PO2-dependent upregulation of Rho-kinase expression. CONCLUSIONS: O2 activates Rho-kinase and increases Rho-kinase expression in term DA smooth muscle cells by a redox-regulated, positive-feedback mechanism that promotes sustained vasoconstriction. Conversely, Rho-kinase inhibitors may be useful in maintaining DA patency, as a bridge to congenital heart surgery.


Asunto(s)
Conducto Arterial/efectos de los fármacos , Oxígeno/farmacología , Proteínas Serina-Treonina Quinasas/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP rhoA/efectos de los fármacos , Proteína de Unión al GTP rhoB/biosíntesis , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Amidas/farmacología , Animales , Animales Recién Nacidos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Conducto Arterial/enzimología , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Retroalimentación Fisiológica , Femenino , Corazón Fetal/fisiología , Edad Gestacional , Humanos , Peróxido de Hidrógeno/metabolismo , Síndrome del Corazón Izquierdo Hipoplásico/patología , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Nifedipino/farmacología , Técnicas de Cultivo de Órganos , Oxidación-Reducción , Oxígeno/sangre , Presión Parcial , Fenilefrina/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Conejos , Superóxidos/metabolismo , Vasoconstricción/fisiología , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoB/genética
13.
Cancer Cell ; 11(1): 37-51, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17222789

RESUMEN

The unique metabolic profile of cancer (aerobic glycolysis) might confer apoptosis resistance and be therapeutically targeted. Compared to normal cells, several human cancers have high mitochondrial membrane potential (DeltaPsim) and low expression of the K+ channel Kv1.5, both contributing to apoptosis resistance. Dichloroacetate (DCA) inhibits mitochondrial pyruvate dehydrogenase kinase (PDK), shifts metabolism from glycolysis to glucose oxidation, decreases DeltaPsim, increases mitochondrial H2O2, and activates Kv channels in all cancer, but not normal, cells; DCA upregulates Kv1.5 by an NFAT1-dependent mechanism. DCA induces apoptosis, decreases proliferation, and inhibits tumor growth, without apparent toxicity. Molecular inhibition of PDK2 by siRNA mimics DCA. The mitochondria-NFAT-Kv axis and PDK are important therapeutic targets in cancer; the orally available DCA is a promising selective anticancer agent.


Asunto(s)
Apoptosis/fisiología , Mitocondrias/metabolismo , Neoplasias/metabolismo , Canales de Potasio/metabolismo , Animales , Línea Celular Tumoral , Ácido Dicloroacético/farmacología , Humanos , Immunoblotting , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Microscopía Confocal , Mitocondrias/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ratas , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Am J Physiol Lung Cell Mol Physiol ; 292(4): L872-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17142350

RESUMEN

Pulmonary arterial hypertension (PAH) is associated with mutations of bone morphogenetic protein receptor 2 (BMPR2), and BMPR2 expression decreases with the development of experimental PAH. Decreased BMPR2 expression and impaired intracellular BMP signaling in pulmonary artery (PA) smooth muscle cells (PASMC) suppresses apoptosis and promotes proliferation, thereby contributing to the pathogenesis of PAH. We hypothesized that overexpression of BMPR2 in resistance PAs would ameliorate established monocrotaline PAH. Human BMPR2 was inserted into a serotype 5 adenovirus with a green fluorescent protein (GFP) reporter. Dose-dependent transgene expression was confirmed in PASMC using fluorescence microscopy, quantitative RT-PCR, and immunoblots. PAH was induced by injecting Sprague-Dawley rats with monocrotaline (60 mg/kg ip) or saline. On day 14, post-monocrotaline (MCT) rats received 5 x 10(9) plaque-forming units of either Ad-human BMPR2 (Ad-hBMPR2) or Ad-GFP. Transgene expression was confirmed by fluorescence microscopy, quantitative RT-PCR of whole lung samples, and laser-capture microdissected resistance PAs. Invasive hemodynamic and echocardiographic end points of pulmonary hypertension were assessed on day 24. Endogenous BMPR2 mRNA levels were greatest in resistance PAs, and expression declined with MCT PAH. Despite robust hBMPR2 expression in all lung lobes and within resistance PAs of treated rats, hBMPR2 did not lower mean PA pressure, pulmonary vascular resistance index, right ventricular hypertrophy, or remodeling of resistance PAs. Nebulized intratracheal adenoviral gene therapy with hBMPR2 reliably distributed hBMPR2 to resistance PAs but did not ameliorate PAH. Depressed BMPR2 expression may be a marker of PAH but is not central to the pathogenesis of this model of PAH.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/biosíntesis , Hipertensión Pulmonar/fisiopatología , Animales , Presión Sanguínea , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Gasto Cardíaco , Terapia Genética , Hipertensión Pulmonar/terapia , Masculino , Monocrotalina , Arteria Pulmonar , Ratas , Ratas Sprague-Dawley , Resistencia Vascular/genética
15.
Circulation ; 112(16): 2477-86, 2005 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-16230500

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) and pulmonary emphysema, both significant global health problems, are characterized by a loss of alveoli. Vascular endothelial growth factor (VEGF) is a trophic factor required for endothelial cell survival and is abundantly expressed in the lung. METHODS AND RESULTS: We report that VEGF blockade decreases lung VEGF and VEGF receptor 2 (VEGFR-2) expression in newborn rats and impairs alveolar development, leading to alveolar simplification and loss of lung capillaries, mimicking BPD. In hyperoxia-induced BPD in newborn rats, air space enlargement and loss of lung capillaries are associated with decreased lung VEGF and VEGFR-2 expression. Postnatal intratracheal adenovirus-mediated VEGF gene therapy improves survival, promotes lung capillary formation, and preserves alveolar development in this model of irreversible lung injury. Combined VEGF and angiopoietin-1 gene transfer matures the new vasculature, reducing the vascular leakage seen in VEGF-induced capillaries. CONCLUSIONS: These findings underscore the importance of the vasculature in what is traditionally thought of as an airway disease and open new therapeutic avenues for lung diseases characterized by irreversible loss of alveoli through the modulation of angiogenic growth factors.


Asunto(s)
Displasia Broncopulmonar/terapia , Terapia Genética , Hiperoxia/terapia , Pulmón/irrigación sanguínea , Neovascularización Fisiológica , Alveolos Pulmonares/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Adenoviridae , Angiografía , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Endotelio Vascular/fisiología , Endotelio Vascular/ultraestructura , Vectores Genéticos , Humanos , Recién Nacido , Lesión Pulmonar , Arteria Pulmonar , Ratas
16.
Am J Respir Crit Care Med ; 172(6): 750-6, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15947285

RESUMEN

RATIONALE: Bronchopulmonary dysplasia (BPD), the chronic lung disease of preterm infants, and pulmonary emphysema, both significant global health problems, are characterized by an arrest in alveolar growth/loss of alveoli structures. Mechanisms that inhibit distal lung growth are poorly understood, but recent studies suggest that impaired vascular endothelial growth factor signaling and reduced nitric oxide (NO) production decreases alveolar and vessel growth in the developing lung, features observed in experimental oxygen-induced BPD. NO exerts its biological activity by stimulating guanosine 3',5'-cyclic monophosphate (cGMP) production. OBJECTIVES: Because cGMP is inactivated by phosphodiesterase (PDE) enzymes, we hypothesized that the cGMP-specific PDE5 inhibitor sildenafil would promote angiogenesis and attenuate oxygen-induced lung injury in newborn rats. METHODS, MEASUREMENTS, AND MAIN RESULTS: In vitro, sildenafil (10(-4) M) increased endothelial capillary network formation of human pulmonary endothelial cells exposed to hyperoxia. In vivo, rat pups were randomly exposed from birth to normoxia, hyperoxia (95% O(2), BPD model), and hyperoxia+sildenafil (100 mg/kg/day subcutaneously). Rat pups exposed to hyperoxia showed fewer and enlarged air spaces as well as decreased capillary density, mimicking pathologic features seen in human BPD. These structural anomalies were associated with echographic (decreased pulmonary acceleration time) and structural (right ventricular hypertrophy and increased medial wall thickness) signs of pulmonary hypertension. Sildenafil preserved alveolar growth and lung angiogenesis, and decreased pulmonary vascular resistance, right ventricular hypertrophy and medial wall thickness. CONCLUSIONS: Our findings suggest a role for the NO/cGMP pathway during alveolar development. Sildenafil may have therapeutic potential in diseases associated with impaired alveolar structures.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Enfermedades Pulmonares/fisiopatología , Pulmón/patología , Oxígeno , Inhibidores de Fosfodiesterasa/farmacología , Piperazinas/farmacología , Alveolos Pulmonares/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Células Cultivadas , GMP Cíclico/sangre , Humanos , Hipertensión Pulmonar/diagnóstico por imagen , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Neovascularización Fisiológica/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Purinas , Ratas , Ratas Sprague-Dawley , Citrato de Sildenafil , Sulfonas , Ultrasonografía Doppler
17.
Circ Res ; 95(8): 830-40, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15375007

RESUMEN

The pulmonary arteries (PA) in pulmonary arterial hypertension (PAH) are constricted and remodeled;. They have suppressed apoptosis, partly attributable to suppression of the bone morphogenetic protein axis and selective downregulation of PA smooth muscle cell (PASMC) voltage-gated K+ channels, including Kv1.5. The Kv downregulation-induced increase in [K+]i, tonically inhibits caspases, further suppressing apoptosis. Mitochondria control apoptosis and produce activated oxygen species like H2O2, which regulate vascular tone by activating K+ channels, but their role in PAH is unknown. We show that dichloroacetate (DCA), a metabolic modulator that increases mitochondrial oxidative phosphorylation, prevents and reverses established monocrotaline-induced PAH (MCT-PAH), significantly improving mortality. Compared with MCT-PAH, DCA-treated rats (80 mg/kg per day in drinking water on day 14 after MCT, studied on day 21) have decreased pulmonary, but not systemic, vascular resistance (63% decrease, P<0.002), PA medial thickness (28% decrease, P<0.0001), and right ventricular hypertrophy (34% decrease, P<0.001). DCA is similarly effective when given at day 1 or day 21 after MCT (studied day 28) but has no effect on normal rats. DCA depolarizes MCT-PAH PASMC mitochondria and causes release of H2O2 and cytochrome c, inducing a 10-fold increase in apoptosis within the PA media (TUNEL and caspase 3 activity) and decreasing proliferation (proliferating-cell nuclear antigen and BrdU assays). Immunoblots, immunohistochemistry, laser-captured microdissection-quantitative reverse-transcription polymerase chain reaction and patch-clamping show that DCA reverses the Kv1.5 downregulation in resistance PAs. In summary, DCA reverses PA remodeling by increasing the mitochondria-dependent apoptosis/proliferation ratio and upregulating Kv1.5 in the media. We identify mitochondria-dependent apoptosis as a potential target for therapy and DCA as an effective and selective treatment for PAH.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácido Dicloroacético/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Animales , División Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Ácido Dicloroacético/farmacología , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/prevención & control , Hemodinámica/efectos de los fármacos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/patología , Canal de Potasio Kv1.5 , Mitocondrias/efectos de los fármacos , Monocrotalina/toxicidad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Especificidad de Órganos , Fosforilación Oxidativa/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Arteria Pulmonar/citología , Ratas , Canales de Potasio Shab , Resistencia Vascular/efectos de los fármacos
18.
Circulation ; 110(11): 1372-9, 2004 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-15353504

RESUMEN

BACKGROUND: Oxygen (O2)-sensitive K+ channels mediate acute O2 sensing in many tissues. At birth, initial functional closure of the ductus arteriosus (DA) results from O2-induced vasoconstriction. This mechanism often fails in premature infants, resulting in persistent DA, a common form of congenital heart disease. We hypothesized that the basis for impaired O2 constriction in preterm DA is reduced expression and function of O2-sensitive, voltage-gated (Kv) channels. METHODS AND RESULTS: Preterm rabbit DA rings have reduced O2 constriction (even after inhibition of prostaglandin and nitric oxide synthases), and preterm DA smooth muscle cells (DASMCs) display reduced O2-sensitive K+ current. This is associated with decreased mRNA and protein expression of certain O2-sensitive Kv channels (Kv1.5 and Kv2.1) but equivalent expression of the L-type calcium channel. Transmural Kv1.5 or Kv2.1 gene transfer "rescues" the developmental deficiency, conferring O2 responsiveness to preterm rabbit DAs. Targeted SMC Kv1.5 gene transfer also enhances O2 constriction in human DAs. CONCLUSIONS: These data demonstrate a central role for developmentally regulated DASMC O2-sensitive Kv channels in the functional closure of the DA. Modulation of Kv channels may have therapeutic potential in diseases associated with impaired O2 responsiveness, including persistent DA.


Asunto(s)
Conducto Arterioso Permeable/etiología , Conducto Arterial/fisiopatología , Oxígeno/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Potasio/metabolismo , Nacimiento Prematuro , 4-Aminopiridina/farmacología , Adenoviridae/genética , Animales , Animales Recién Nacidos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/análisis , Humanos , Recién Nacido , Canal de Potasio Kv1.5 , Canales de Potasio con Entrada de Voltaje/deficiencia , Canales de Potasio con Entrada de Voltaje/genética , Embarazo , ARN Mensajero/biosíntesis , Conejos , Ratas , Canales de Potasio Shab , Transducción Genética , Vasoconstricción/efectos de los fármacos
19.
FASEB J ; 18(12): 1382-91, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15333581

RESUMEN

Female sexual function is under-studied, and mechanisms of clitoral engorgement-relaxation are incompletely understood. Penile erection results from nitric oxide (NO) -induced cyclic guanosine monophosphate (cGMP) accumulation. cGMP-dependent protein kinase (PKG) activates large-conductance, calcium-activated potassium channels (BK(Ca)), thereby hyperpolarizing and relaxing vascular and trabecular smooth muscle cells, allowing engorgement. We hypothesize rat clitorises relax by a similar mechanism. Rat clitorises express components of the proposed pathway: neuronal and endothelial NO synthases, soluble guanylyl cyclase (sGC), type 5 phosphodiesterase (PDE-5), and BK(Ca) channels. The NO donor diethylamine NONOate (DEANO), the PKG activator 8-pCPT-cGMP, and the PDE-5 inhibitor sildenafil, cause dose-dependent clitoral relaxation that is inhibited by antagonists of PKG (Rp-8-Br-cGMPS) or BK(Ca) channels (iberiotoxin). Electrical field stimulation induces tetrodotoxin-sensitive NO release and relaxation that is inhibited by the Na+ channel blocker tetrodotoxin or sGC inhibitor 1H-(1,2,4)oxadiozolo(4,3-a)quinoxalin-1-one. Human BK(Ca) channels, transferred to Chinese hamster ovary cells via an adenoviral vector, and endogenous rat clitoral smooth muscle K+ current are activated by this PKG-dependent mechanism. Laser confocal microscopy reveals protein expression of BK(Ca) channels on clitoral smooth muscle cells; these cells exhibit BK(Ca) channel activity that is activated by both DEANO and sildenafil. We conclude that neurovascular derived NO causes clitoral relaxation via a PKG-dependent activation of BK(Ca) channels. The BK(Ca) channel is an appealing target for drug therapy of female erectile dysfunction.


Asunto(s)
Clítoris/fisiología , GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Canales de Potasio Calcio-Activados/metabolismo , Animales , Células CHO , Calcio/metabolismo , Clítoris/anatomía & histología , Clítoris/irrigación sanguínea , Clítoris/inervación , Cricetinae , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Estimulación Eléctrica , Electrofisiología , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Canales de Potasio de Gran Conductancia Activados por el Calcio , Rayos Láser , Microdisección , Relajación Muscular/efectos de los fármacos , Músculo Liso/irrigación sanguínea , Músculo Liso/efectos de los fármacos , Músculo Liso/inervación , Músculo Liso/fisiología , Óxido Nítrico/biosíntesis , Piperazinas/farmacología , Canales de Potasio Calcio-Activados/genética , Purinas , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Citrato de Sildenafil , Sulfonas
20.
Org Lett ; 6(16): 2645-8, 2004 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15281734

RESUMEN

2'-Beta-methyl- and 2'-beta-hydroxymethyl-2'-deoxy-4'-thionucleosides have been synthesized through PhSeCl-mediated electrophilic glycosidation using 4-thiofuranoid glycals having carbon substituents at the C2-position as a glycosyl donor. Preparation of these glycals were carried out by means of the C2 lithiation of 1-chloro-4-thiofuranoid glycal with LTMP followed by the Birch reduction of the chlorine atom. [reaction: see text]


Asunto(s)
Siloxanos/química , Tionucleósidos/síntesis química , Tiofenos/química , Estructura Molecular , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA