Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Cardiovasc Pathol ; 36: 53-63, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30056298

RESUMEN

BACKGROUND: The calcific aortic valve disease (CAVD) is a common heart pathology that involves inflammation, fibrosis, and calcification of aortic valve leaflets. All these processes could be affected by changes in the extracellular purinergic signaling that depend on the activity of ectonucleotidases, mainly ectonucleoside triphosphate diphosphohydrolase 1 (CD39, eNTPD1) and ecto-5'nucleotidase (CD73, e5NT). OBJECTIVE AND METHODS: We investigated the localization of CD39 and CD73 proteins in human noncalcified and calcified aortic valves using immunohistochemistry together with analysis of NTPDases and e5NT activities in aortic valve homogenates by analysis of substrate into product conversion by high-performance liquid chromatography. We also measured the rates of extracellular nucleotide catabolism on the surface of isolated cultured aortic valve endothelial (hAVECs) and interstitial cells (hAVICs) as well as characterized cellular CD39 and CD73 distribution. RESULTS: In noncalcified valves, CD39 and CD73 were expressed in both endothelial and interstitial cells, while in calcified valves, the expressions of CD39 and CD73 were significantly down-regulated with the exception of calcified regions where the expression of CD73 was maintained. This correlated with activities in valve homogenates. NTPDase was reduced by 35% and e5NT activity by 50% in calcified vs. noncalcified valve. CD39 and CD73 were present mainly in the cell membrane of hAVECs, but in hAVICs, these proteins were also present intracellularly. The rates of extracellular adenosine triphosphate and adenosine monophosphate hydrolysis in isolated hAVECs and hAVICs were comparable. CONCLUSION: The presence of ectonucleotidases in valves and especially in aortic valve interstitial cells highlights important local role of purinergic signaling and metabolism. Changes in the local expression and hence the activity of CD39 and CD73 in calcified valves suggest their potential role in CAVD.


Asunto(s)
5'-Nucleotidasa/metabolismo , Válvula Aórtica/enzimología , Apirasa/metabolismo , Calcinosis/enzimología , Enfermedades de las Válvulas Cardíacas/enzimología , Inmunohistoquímica , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Válvula Aórtica/patología , Calcinosis/patología , Células Cultivadas , Células Endoteliales/enzimología , Células Endoteliales/patología , Femenino , Proteínas Ligadas a GPI/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Hidrólisis , Masculino , Persona de Mediana Edad , Adulto Joven
2.
Cardiovasc Drugs Ther ; 32(3): 245-253, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29766336

RESUMEN

PURPOSE: Anthracyclines cause chronic irreversible cardiac failure, but the mechanism remains poorly understood. Emerging data indicate that cardiac damage begins early, suggesting protective modalities delivered in the acute stage may confer prolonged benefit. Ischaemic preconditioning (IPC) activates the pro-survival reperfusion injury salvage kinase (RISK) pathway which involves PI3-kinase and MAPK/ERK1/2. METHODS: We investigated whether simulated IPC (sIPC), in the form of a sublethal exposure to a hypoxic buffer simulating ischaemic conditions followed by reoxygenation, protects primary adult rat cardiomyocytes against anthracycline-induced injury. PI3-kinase and MAPK/ERK1/2 were inhibited using LY294002, and PD98059. The role of reactive oxygen species (ROS), mitochondrial membrane potential (Δψm) and mitochondrial permeability transition pore (mPTP) were also investigated in doxorubicin-treated cells. We further examined whether sIPC protected HeLa cancer cells from doxorubicin-induced death. RESULTS: sIPC protected cardiomyocytes against doxorubicin-induced death (35.4 ± 1.7% doxorubicin vs 14.7 ± 1.5% doxorubicin + sIPC; p < 0.01). This protection was abrogated by the PI3-kinase inhibitor, LY294002, but not the MAPK/ERK1/2 inhibitor, PD98059. A ROS scavenger failed to rescue cardiomyocytes from doxorubicin toxicity, and no significant influence on Δψm or mPTP opening was identified after subjecting cells to a doxorubicin insult. Importantly, sIPC did not protect HeLa cancer cells from doxorubicin-induced death. CONCLUSION: sIPC is able to protect cardiomyocytes against anthracycline injury via a pathway involving PI3-kinase. This mechanism appears to be independent of ROS, changes to Δψm, and mPTP. Further investigation of the mechanism of sIPC-induced protection against anthracycline-injury is warranted.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Cardiopatías/prevención & control , Precondicionamiento Isquémico Miocárdico , Miocitos Cardíacos/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Animales , Cardiotoxicidad , Hipoxia de la Célula , Femenino , Células HeLa , Cardiopatías/inducido químicamente , Cardiopatías/enzimología , Cardiopatías/patología , Humanos , Masculino , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Neoplasias del Cuello Uterino/enzimología , Neoplasias del Cuello Uterino/patología
3.
PLoS One ; 12(6): e0180571, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28662206

RESUMEN

Doxorubicin is a potent chemotherapeutic agent that is widely-used to treat a variety of cancers but causes acute and chronic cardiac injury, severely limiting its use. Clinically, the acute side effects of doxorubicin are mostly manageable, whereas the delayed consequences can lead to life-threatening heart failure, even decades after cancer treatment. The cardiotoxicity of doxorubicin is subject to a critical cumulative dose and so dosage limitation is considered to be the best way to reduce these effects. Hence, a number of studies have defined a "safe dose" of the drug, both in animal models and clinical settings, with the aim of avoiding long-term cardiac effects. Here we show that a dose generally considered as safe in a mouse model can induce harmful changes in the myocardium, as early as 2 weeks after infusion. The adverse changes include the development of fibrotic lesions, disarray of cardiomyocytes and a major transcription dysregulation. Importantly, low-dose doxorubicin caused specific changes in the transcriptional profile of several histone deacetylases (HDACs) which are epigenetic regulators of cardiac remodelling. This suggests that cardioprotective therapies, aimed at modulating HDACs during doxorubicin treatment, deserve further exploration.


Asunto(s)
Cardiomiopatías/inducido químicamente , Modelos Animales de Enfermedad , Doxorrubicina/efectos adversos , Histona Desacetilasas/metabolismo , Transcripción Genética , Animales , Cardiomiopatías/enzimología , Cardiomiopatías/genética , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Microscopía Confocal
4.
Cardiovasc Drugs Ther ; 31(1): 63-75, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28185035

RESUMEN

Anthracycline chemotherapy maintains a prominent role in treating many forms of cancer. Cardiotoxic side effects limit their dosing and improved cancer outcomes expose the cancer survivor to increased cardiovascular morbidity and mortality. The basic mechanisms of cardiotoxicity may involve direct pathways for reactive oxygen species generation and topoisomerase 2 as well as other indirect pathways. Cardioprotective treatments are few and those that have been examined include renin angiotensin system blockade, beta blockers, or the iron chelator dexrazoxane. New treatments exploiting the ErbB or other novel pro-survival pathways, such as conditioning, are on the cardioprotection horizon. Even in the forthcoming era of targeted cancer therapies, the substantial proportion of today's anthracycline-treated cancer patients may become tomorrow's cardiac patient.


Asunto(s)
Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/uso terapéutico , Cardiopatías/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Animales , Cardiotoxicidad , Fármacos Cardiovasculares/uso terapéutico , Citoprotección , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Cardiopatías/prevención & control , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Troponina/metabolismo
5.
PLoS Genet ; 11(3): e1005021, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25748626

RESUMEN

Skeletal muscle remodelling and contractile dysfunction occur through both acute and chronic disease processes. These include the accumulation of insoluble aggregates of misfolded amyloid proteins that is a pathological feature of Huntington's disease (HD). While HD has been described primarily as a neurological disease, HD patients' exhibit pronounced skeletal muscle atrophy. Given that huntingtin is a ubiquitously expressed protein, skeletal muscle fibres may be at risk of a cell autonomous HD-related dysfunction. However the mechanism leading to skeletal muscle abnormalities in the clinical and pre-clinical HD settings remains unknown. To unravel this mechanism, we employed the R6/2 transgenic and HdhQ150 knock-in mouse models of HD. We found that symptomatic animals developed a progressive impairment of the contractile characteristics of the hind limb muscles tibialis anterior (TA) and extensor digitorum longus (EDL), accompanied by a significant loss of motor units in the EDL. In symptomatic animals, these pronounced functional changes were accompanied by an aberrant deregulation of contractile protein transcripts and their up-stream transcriptional regulators. In addition, HD mouse models develop a significant reduction in muscle force, possibly as a result of a deterioration in energy metabolism and decreased oxidation that is accompanied by the re-expression of the HDAC4-DACH2-myogenin axis. These results show that muscle dysfunction is a key pathological feature of HD.


Asunto(s)
Enfermedad de Huntington/patología , Músculo Esquelético/patología , Animales , Atrofia , Técnicas de Sustitución del Gen , Histona Desacetilasas/metabolismo , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Miogenina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
6.
Front Physiol ; 5: 380, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25339908

RESUMEN

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by the expansion of a polyglutamine stretch within the huntingtin protein (HTT). The neurological symptoms, that involve motor, cognitive and psychiatric disturbances, are caused by neurodegeneration that is particularly widespread in the basal ganglia and cereberal cortex. HTT is ubiquitously expressed and in recent years it has become apparent that HD patients experience a wide array of peripheral organ dysfunction including severe metabolic phenotype, weight loss, HD-related cardiomyopathy and skeletal muscle wasting. Although skeletal muscles pathology became a hallmark of HD, the mechanisms underlying muscular atrophy in this disorder are unknown. Skeletal muscles account for approximately 40% of body mass and are highly adaptive to physiological and pathological conditions that may result in muscle hypertrophy (due to increased mechanical load) or atrophy (inactivity, chronic disease states). The atrophy is caused by degeneration of myofibers and their replacement by fibrotic tissue is the major pathological feature in many genetic muscle disorders. Under normal physiological conditions the muscle function is orchestrated by a network of intrinsic hypertrophic and atrophic signals linked to the functional properties of the motor units that are likely to be imbalanced in HD. In this article, we highlight the emerging field of research with particular focus on the recent studies of the skeletal muscle pathology and the identification of new disease-modifying treatments.

7.
J Mol Cell Cardiol ; 74: 340-52, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24984146

RESUMEN

Fetal cardiomyocyte adaptation to low levels of oxygen in utero is incompletely understood, and is of interest as hypoxia tolerance is lost after birth, leading to vulnerability of adult cardiomyocytes. It is known that cardiac mitochondrial morphology, number and function change significantly following birth, although the underlying molecular mechanisms and physiological stimuli are undefined. Here we show that the decrease in cardiomyocyte HIF-signaling in cardiomyocytes immediately after birth acts as a physiological switch driving mitochondrial fusion and increased postnatal mitochondrial biogenesis. We also investigated mechanisms of ATP generation in embryonic cardiac mitochondria. We found that embryonic cardiac cardiomyocytes rely on both glycolysis and the tricarboxylic acid cycle to generate ATP, and that the balance between these two metabolic pathways in the heart is controlled around birth by the reduction in HIF signaling. We therefore propose that the increase in ambient oxygen encountered by the neonate at birth acts as a key physiological stimulus to cardiac mitochondrial adaptation.


Asunto(s)
Ventrículos Cardíacos/metabolismo , Hipoxia/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Adaptación Fisiológica , Adenosina Trifosfato/biosíntesis , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Ciclo del Ácido Cítrico/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Glucólisis/efectos de los fármacos , Glucólisis/genética , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Oxígeno/farmacología , Transducción de Señal , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
8.
PLoS Biol ; 11(9): e1001666, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24086110

RESUMEN

Cardiomyocytes are vulnerable to hypoxia in the adult, but adapted to hypoxia in utero. Current understanding of endogenous cardiac oxygen sensing pathways is limited. Myocardial oxygen consumption is determined by regulation of energy metabolism, which shifts from glycolysis to lipid oxidation soon after birth, and is reversed in failing adult hearts, accompanying re-expression of several "fetal" genes whose role in disease phenotypes remains unknown. Here we show that hypoxia-controlled expression of the transcription factor Hand1 determines oxygen consumption by inhibition of lipid metabolism in the fetal and adult cardiomyocyte, leading to downregulation of mitochondrial energy generation. Hand1 is under direct transcriptional control by HIF1α. Transgenic mice prolonging cardiac Hand1 expression die immediately following birth, failing to activate the neonatal lipid metabolising gene expression programme. Deletion of Hand1 in embryonic cardiomyocytes results in premature expression of these genes. Using metabolic flux analysis, we show that Hand1 expression controls cardiomyocyte oxygen consumption by direct transcriptional repression of lipid metabolising genes. This leads, in turn, to increased production of lactate from glucose, decreased lipid oxidation, reduced inner mitochondrial membrane potential, and mitochondrial ATP generation. We found that this pathway is active in adult cardiomyocytes. Up-regulation of Hand1 is protective in a mouse model of myocardial ischaemia. We propose that Hand1 is part of a novel regulatory pathway linking cardiac oxygen levels with oxygen consumption. Understanding hypoxia adaptation in the fetal heart may allow development of strategies to protect cardiomyocytes vulnerable to ischaemia, for example during cardiac ischaemia or surgery.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , Consumo de Oxígeno/genética , Adenosina Trifosfato/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Potencial de la Membrana Mitocondrial/genética , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Oxígeno/metabolismo , Activación Transcripcional
9.
Plant Physiol Biochem ; 73: 56-62, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24056127

RESUMEN

Annexins are calcium- and membrane-binding proteins that have been shown to have diverse properties such as actin, integrin and GTP binding, both in animals and plants. Recently, Medicago truncatula annexin 1 (AnnMt1) has been suggested to participate in nodulation (Nod factor signaling) and mycorrhization in legume plants. In this report we demonstrate for the first time that recombinant AnnMt1 (rec-AnnMt1) mediates membrane permeabilization to cations with conductance ranging from 16 pS to 329 pS. In agreement with other structurally determined annexins, homology modeling of AnnMt1 suggests that most of the functional determinants are found on the convex surface of the modeled structure. In conclusion, we propose a potential constitutive role of AnnMt1 in Nod factor signaling as a non-specific ion channel.


Asunto(s)
Anexinas/metabolismo , Canales Iónicos/metabolismo , Medicago truncatula/metabolismo , Proteínas de Plantas/metabolismo , Nodulación de la Raíz de la Planta , Anexinas/química , Cationes/metabolismo , Permeabilidad de la Membrana Celular , Canales Iónicos/química , Medicago truncatula/química , Medicago truncatula/microbiología , Modelos Biológicos , Estructura Molecular , Micorrizas , Proteínas de Plantas/química , Conformación Proteica , Proteínas Recombinantes/metabolismo , Transducción de Señal
10.
J Biol Chem ; 288(12): 8332-8341, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23362253

RESUMEN

CLU (clusterin) is a tumor suppressor gene that we have previously shown to be negatively modulated by the MYCN proto-oncogene, but the mechanism of repression was unclear. Here, we show that MYCN inhibits the expression of CLU by direct interaction with the non-canonical E box sequence CACGCG in the 5'-flanking region. Binding of MYCN to the CLU gene induces bivalent epigenetic marks and recruitment of repressive proteins such as histone deacetylases and Polycomb members. MYCN physically binds in vitro and in vivo to EZH2, a component of the Polycomb repressive complex 2, required to repress CLU. Notably, EZH2 interacts with the Myc box domain 3, a segment of MYC known to be essential for its transforming effects. The expression of CLU can be restored in MYCN-amplified cells by epigenetic drugs with therapeutic results. Importantly, the anticancer effects of the drugs are ablated if CLU expression is blunted by RNA interference. Our study implies that MYC tumorigenesis can be effectively antagonized by epigenetic drugs that interfere with the recruitment of chromatin modifiers at repressive E boxes of tumor suppressor genes such as CLU.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Región de Flanqueo 5' , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Cromatina/metabolismo , Clusterina/genética , Clusterina/metabolismo , Elementos E-Box , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Datos de Secuencia Molecular , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/fisiología , Proteínas Oncogénicas/fisiología , Regiones Promotoras Genéticas , Unión Proteica , Proto-Oncogenes Mas
11.
Clin Cancer Res ; 19(5): 1116-25, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23322899

RESUMEN

PURPOSE: Neuroblastoma is a rare childhood cancer whose high risk, metastatic form has a dismal outcome in spite of aggressive therapeutic interventions. The toxicity of drug treatments is a major problem in this pediatric setting. In this study, we investigated whether Polyphenon E, a clinical grade mixture of green tea catechins under evaluation in multiple clinical cancer trials run by the National Cancer Institute (Bethesda, MD), has anticancer activity in mouse models of neuroblastoma. EXPERIMENTAL DESIGN: We used three neuroblastoma models: (i) transgenic TH-MYCN mouse developing spontaneous neuroblastomas; (ii) nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice xenotransplanted with human SHSY5Y cells; and (iii) A/J mice transplanted with syngeneic Neuro 2A cells. Mice were randomized in control and Polyphenon E-drinking groups. Blood from patients with neuroblastoma and normal controls was used to assess the phenotype and function of myeloid cells. RESULTS: Polyphenon E reduced the number of tumor-infiltrating myeloid cells, and inhibited the development of spontaneous neuroblastomas in TH-MYCN transgenic mice. In therapeutic models of neuroblastoma in A/J, but not in immunodeficient NOD/SCID mice, Polyphenon E inhibited tumor growth by acting on myeloid-derived suppressor cells (MDSC) and CD8 T cells. In vitro, Polyphenon E impaired the development and motility of MDSCs and promoted differentiation to more neutrophilic forms through the 67 kDa laminin receptor signaling and induction of granulocyte colony-stimulating factor. The proliferation of T cells infiltrating a patient metastasis was reactivated by Polyphenon E. CONCLUSIONS: These findings suggest that the neuroblastoma-promoting activity of MDSCs can be manipulated pharmacologically in vivo and that green tea catechins operate, at least in part, through this mechanism.


Asunto(s)
Catequina/análogos & derivados , Células Mieloides/inmunología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/inmunología , Linfocitos T/inmunología , Té/química , Animales , Catequina/farmacología , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Mieloides/efectos de los fármacos , Neuroblastoma/mortalidad , Receptores de Laminina/metabolismo , Tasa de Supervivencia , Linfocitos T/efectos de los fármacos
12.
Mitochondrion ; 12(1): 162-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21925619

RESUMEN

The 25 kDa branched polyethylenimine (PEI) is a highly efficient synthetic polycation used in transfection protocols, but also triggers mitochondrial-mediated apoptotic cell death processes where the mechanistic issues are poorly understood. We now demonstrate that PEI in a concentration- and time-dependent manner can affect functions (membrane potential, swelling and respiration) and ultrastructural integrity of freshly isolated rat liver mitochondria. The threshold concentration for detection of PEI-mediated impairment of rat liver mitochondrial functions is 3 µg/mL, however, lower PEI levels still exert some effects on mitochondrial morphology and respiration, and these may be related to the inherent membrane perturbing properties of this polycation. The PEI-mediated mitochondrial swelling phase is biphasic, with a fast decaying initial period (most prominent from 4 µg/mL PEI) followed by a slower, linear swelling response. The slow phase is presumably the result of a time-dependent transition permeability opening in mitochondria initially resistant to swelling/depolarization, but may further be related to PEI-induced nanoscale structural defects and/or formation of pores in the outer membrane. Respiration assessments further suggested that PEI in the presence of exogenous ADP behaves in a similar fashion to a slow-acting inhibitory compound. PEI further shows an uncoupling property that is detectable at low respiration rates. The relevance of these findings to PEI-mediated initiation of intrinsic apoptotic pathway is discussed.


Asunto(s)
Respiración de la Célula/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Polietileneimina/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Terapia Genética , Hepatocitos/efectos de los fármacos , Hepatocitos/ultraestructura , Hígado/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Membranas Mitocondriales/fisiología , Ácidos Nucleicos/genética , Ácidos Nucleicos/metabolismo , Ratas , Ratas Wistar
13.
Toxicon ; 58(4): 369-79, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21798278

RESUMEN

Entomopathogenic fungi are important natural regulatory factors of insect populations and have potential as biological control agents of insect pests. The cosmopolitan soil fungus Conidiobolus coronatus (Entomopthorales) easily attacks Galleria mellonella (Lepidoptera) larvae. Prompt death of invaded insects is attributed to the action of toxic metabolites released by the invader. Effect of fungal metabolites on hemocytes, insect blood cells involved in innate defense response, remains underexplored to date. C. coronatus isolate 3491 inducing 100% mortality of G. mellonella last instar larvae exposed to sporulating colonies, was cultivated at 20 °C in minimal medium. Post-incubation filtrates were used as a source of fungal metabolites. A two-step HPLC (1 step: Shodex KW-803 column eluted with 50 mM KH(2)PO(4) supplemented with 0.1 M KCl, pH 6.5; 2 step: ProteinPak™ CM 8HR column equilibrated with 5 mM KH(2)PO(4), pH 6.5, proteins eluted with a linear gradient of 0.5 M KCl) allowed the isolation of coronatin-1, an insecticidal 36 kDa protein showing both elastolytic and chitinolytic activities. Addition of coronatin-1 into primary in vitro cultures of G. mellonella hemocytes resulted in rapid disintegration of spherulocytes freely floating in culture medium and shrinkage of plasmatocytes adhering to the bottom of culture well. Coronatin-1 stimulated pseudopodia atrophy and, in consequence, disintegration of nets formed by cultured hemocytes. After incorporation of coronatin-1 into planar lipid membrane (PLM) ion channels selective for K(+) ions in 50/450 mM KCl solutions were observed. Potassium current flows were recorded in nearly 70% of experiments with conductance from 300 pS up to 1 nS. All observed channels were active at both positive and negative membrane potentials. Under experimental conditions incorporated coronatin-1 exhibited a zero current potential (E(rev)) of 47.7 mV, which indicates K(+)-selectivity of this protein. The success of the purification of coronatin-1 will allow further characterization of the mode of action of this molecule, including ability of coronatin-1 to form potassium channels in immunocompetent hemocytes.


Asunto(s)
Conidiobolus/química , Hemocitos/efectos de los fármacos , Insecticidas/farmacología , Mariposas Nocturnas/efectos de los fármacos , Micotoxinas/farmacología , Canales de Potasio/química , Animales , Capacidad Eléctrica , Insecticidas/química , Insecticidas/aislamiento & purificación , Larva/efectos de los fármacos , Membrana Dobles de Lípidos/química , Potenciales de la Membrana , Mariposas Nocturnas/citología , Mariposas Nocturnas/crecimiento & desarrollo , Micotoxinas/química , Micotoxinas/aislamiento & purificación
14.
Pflugers Arch ; 462(1): 135-42, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21484537

RESUMEN

Muscle LIM protein (MLP, also known as cysteine rich protein 3 (CSRP3, CRP3)) is a muscle-specific-expressed LIM-only protein. It consists of 194 amino-acids and has been described initially as a factor involved in myogenesis (Arber et al. Cell 79:221-231, 1994). MLP soon became an important model for experimental cardiology when it was first demonstrated that MLP deficiency leads to myocardial hypertrophy followed by a dilated cardiomyopathy and heart failure phenotype (Arber et al. Cell 88:393-403, 1997). At this time, this was the first genetically altered animal model to develop this devastating disease. Interestingly, MLP was also found to be down-regulated in humans with heart failure (Zolk et al. Circulation 101:2674-2677, 2000) and MLP mutations are able to cause hypertrophic and dilated forms of cardiomyopathy in humans (Bos et al. Mol Genet Metab 88:78-85, 2006; Geier et al. Circulation 107:1390-1395, 2003; Hershberger et al. Clin Transl Sci 1:21-26, 2008; Knöll et al. Cell 111:943-955, 2002; Knöll et al. Circ Res 106:695-704, 2010; Mohapatra et al. Mol Genet Metab 80:207-215, 2003). Although considerable efforts have been undertaken to unravel the underlying molecular mechanisms-how MLP mutations, either in model organisms or in the human setting cause these diseases are still unclear. In contrast, only precise knowledge of the underlying molecular mechanisms will allow the development of novel and innovative therapeutic strategies to combat this otherwise lethal condition. The focus of this review will be on the function of MLP in cardiac mechanosensation and we shall point to possible future directions in MLP research.


Asunto(s)
Corazón/fisiología , Mecanotransducción Celular/fisiología , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica/fisiopatología , Corazón/anatomía & histología , Corazón/fisiopatología , Humanos , Proteínas con Dominio LIM , Proteínas Musculares/genética , Miocardio/citología , Miocardio/patología , Estrés Mecánico
15.
J Cardiovasc Transl Res ; 4(3): 238-44, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21360311

RESUMEN

Mechanosensation (the ultimate conversion of a mechanical stimulus into a biochemical signal) as well as mechanotransduction (transmission of mechanically induced signals) belong to the most fundamental processes in biology. These effects, because of their dynamic nature, are particularly important for the cardiovascular system. Therefore, it is not surprising that defects in cardiac mechanosensation, are associated with various types of cardiomyopathy and heart failure. However, our current knowledge regarding the genetic basis of impaired mechanosensation in the cardiovascular system is beginning to shed light on this subject and is at the centre of this brief review.


Asunto(s)
Cardiomiopatías/genética , Insuficiencia Cardíaca/genética , Mecanotransducción Celular/genética , Miocardio/metabolismo , Sensación/genética , Angiotensina II/genética , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Conectina , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Filamentos Intermedios/metabolismo , Proteínas Musculares/genética , Mutación , Polimorfismo Genético , Proteínas Quinasas/genética , Sistema Renina-Angiotensina/genética , Sarcómeros/metabolismo , Estrés Mecánico
16.
Int J Cardiol ; 151(1): 18-33, 2011 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-20546954

RESUMEN

OBJECTIVES: We studied fibrosis, collagen metabolism, MMPs/TIMPs and cytokine expression in various forms of human heart failure (HF) by quantitative immunofluorescent microscopy, Western blot, zymography, RT-PCR and in situ hybridization. In explanted human hearts with HF due to either dilated (DCM, n=6) or ischemic (ICM-BZ-borderzone, ICM-RZ-remote zone, n=7) or inflammatory (myocarditis, MYO, n=6) cardiomyopathy and 8 controls MMP2, 8, 9, 19, and TIMP1, 2, 3, 4 as well as procollagens I and III (PINP, PIIINP), mature collagen III (IIINTP) and the cross-linked collagen I degradation product (ICTP) were measured. RESULTS: In comparison with controls, MMPs and TIMPs were significantly upregulated ranging (from highest to lowest) from ICM-BZ, DCM, ICM-RZ, MYO for all MMPs with the exception of MMP9 (highest in DCM), and for TIMPs from ICM-BZ, ICM-RZ, DCM and MYO. MMP2 and 9 were activated in all groups. The TIMP/MMP ratio was 1.3 for control, 1.9 in ICM-BZ (TIMP>MMP) and lowered to 1.0 in the other groups. Collagen I/collagen III ratio correlated significantly with the decrease in LVEDP. PINP was higher than ICTP in all groups. PIIINP elevation was present in DCM and ICM-RZ and IIINTP was up to 4-fold augmented in all groups. Fibrosin mRNA was upregulated in ICM-BZ, activin A in MYO but FGF1 and FGF2 remained unchanged. ANP mRNA was increased in all groups. CONCLUSIONS: Although different degrees of severity of collagen metabolism, MMP/TIMP imbalance and cytokine expression in diverse forms of HF are present, the end product is collagen deposition. These findings suggest multiple mechanisms acting alone or in concert in fibrosis development in HF.


Asunto(s)
Colágeno Tipo I/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Adulto , Colágeno Tipo III/metabolismo , Femenino , Fibrosis , Humanos , Masculino , Metaloproteinasa 8 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Microscopía Fluorescente , Persona de Mediana Edad , Miocarditis/metabolismo , Miocarditis/patología , Miocardio/metabolismo , Miocardio/patología , Índice de Severidad de la Enfermedad , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Regulación hacia Arriba/fisiología , Inhibidor Tisular de Metaloproteinasa-4
17.
Biochim Biophys Acta ; 1807(3): 275-85, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21167126

RESUMEN

Single-ion channel activities were measured after reconstitution of potato tuber mitochondrial inner membranes into planar lipid bilayers. In addition to the recently described large-conductance Ca(2+)-activated potassium channel activity (Koszela-Piotrowska et al., 2009), the following mitochondrial ion conductance pathways were recorded: (i) an ATP-regulated potassium channel (mitoK(ATP) channel) activity with a conductance of 164+/-8pS, (ii) a large-conductance Ca(2+)-insensitive iberiotoxin-sensitive potassium channel activity with a conductance of 312 pS+/-23, and (iii) a chloride 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS)-inhibited channel activity with a conductance of 117 pS+/-4. In isolated non-phosphorylating potato tuber mitochondria, individual and combined potassium channel activities caused significant (up to 14mV) but not collapsing K(+)-influx-induced membrane potential depolarisation. Under phosphorylating conditions, the coupling parameters were unchanged in the presence of high K(+) level, indicating that plant K(+) channels function as energy-dissipating systems that are not able to divert energy from oxidative phosphorylation. A potato tuber K(+) channel that is ATP-, 5-hydroxydecanonic acid-, glybenclamide-inhibited and diazoxide-stimulated caused low cation flux, modestly decreasing membrane potential (up to a few mV) and increasing respiration in non-phosphorylating mitochondria. Immunological analysis with antibodies raised against the mammalian plasma membrane ATP-regulated K(+) channel identified a pore-forming subunit of the Kir-like family in potato tuber mitochondrial inner membrane. These results suggest that a mitoK(ATP) channel similar to that of mammalian mitochondria is present in potato tuber mitochondria.


Asunto(s)
Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Canales de Potasio/metabolismo , Solanum tuberosum/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Electrofisiología , Immunoblotting , Activación del Canal Iónico/efectos de los fármacos , Membrana Dobles de Lípidos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Solanum tuberosum/efectos de los fármacos
18.
FEBS Lett ; 584(10): 2057-62, 2010 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-20083113

RESUMEN

The functional characterisation of potassium channels found in the mitochondria of plants and unicellular eukaryotes is critically discussed herein, with a focus on the ATP-sensitive potassium channel and the large-conductance Ca(2+)-activated potassium channel (mitoBK(Ca) channel). The physiological functions of these channels are not completely understood. We discuss the functional connections and roles of potassium channels, uncoupling protein and alternative oxidase, three energy-dissipating systems that exist in the mitochondrial respiratory chain of plants and some unicellular eukaryotes, which include preventing the production of reactive oxygen species.


Asunto(s)
Células Eucariotas/citología , Células Eucariotas/metabolismo , Mitocondrias/metabolismo , Células Vegetales , Plantas/metabolismo , Canales de Potasio/metabolismo , Animales , Metabolismo Energético , Humanos
19.
J Bioenerg Biomembr ; 41(4): 323-34, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19821034

RESUMEN

Mitochondrial potassium channels in the brain have been suggested to have an important role in neuroprotection. The single channel activity of mitochondrial potassium channels was measured after reconstitution of the purified inner membrane from rat brain mitochondria into a planar lipid bilayer. In addition to a large conductance potassium channel that was described previously, we identified a potassium channel that has a mean conductance of 219 +/- 15 pS. The activity of this channel was inhibited by ATP/Mg(2+) and activated by the potassium channel opener BMS191095. Channel activity was not influenced either by 5-hydroxydecanoic acid, an inhibitor of mitochondrial ATP-regulated potassium channels, or by the plasma membrane ATP-regulated potassium channel blocker HMR1098. Likewise, this mitochondrial potassium channel was unaffected by the large conductance potassium channel inhibitor iberiotoxin or by the voltage-dependent potassium channel inhibitor margatoxin. The amplitude of the conductance was lowered by magnesium ions, but the opening ability was unaffected. Immunological studies identified the Kir6.1 channel subunit in the inner membrane from rat brain mitochondria. Taken together, our results demonstrate for the first time the single channel activity and properties of an ATP-regulated potassium channel from rat brain mitochondria.


Asunto(s)
Adenosina Trifosfato/administración & dosificación , Encéfalo/metabolismo , Activación del Canal Iónico/fisiología , Membrana Dobles de Lípidos/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/ultraestructura , Relación Dosis-Respuesta a Droga , Activación del Canal Iónico/efectos de los fármacos , Canales KATP , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Ratas , Ratas Wistar , Teofilina/análogos & derivados
20.
Arterioscler Thromb Vasc Biol ; 29(12): 2093-101, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19778941

RESUMEN

OBJECTIVE: Arteriogenesis, the development of a collateral circulation, is important for tissue survival but remains functionally defective because of early normalization of fluid shear stress (FSS). Using a surgical model of chronically elevated FSS we showed that rabbits exhibited normal blood flow reserve after femoral artery ligature (FAL). Inhibition of the Rho pathway by Fasudil completely blocked the beneficial effect of FSS. In a genome-wide gene profiling we identified actin-binding Rho activating protein (Abra), which was highly upregulated in growing collaterals. METHODS AND RESULTS: qRT-PCR and Western blot confirmed highly increased FSS-dependent expression of Abra in growing collaterals. NO blockage by L-NAME abolished FSS-generated Abra expression as well as the whole arteriogenic process. Cell culture studies demonstrated an Abra-triggered proliferation of smooth muscle cells through a mechanism that requires Rho signaling. Local intracollateral adenoviral overexpression of Abra improved collateral conductance by 60% in rabbits compared to the natural response after FAL. In contrast, targeted deletion of Abra in CL57BL/6 mice led to impaired arteriogenesis. CONCLUSIONS: FSS-induced Abra expression during arteriogenesis is triggered by NO and leads to stimulation of collateral growth by smooth muscle cell proliferation.


Asunto(s)
Arterias/crecimiento & desarrollo , Arterias/fisiología , Proteínas de Microfilamentos/metabolismo , Neovascularización Fisiológica , Actinas/metabolismo , Adenoviridae/genética , Animales , Proliferación Celular , Células Cultivadas , Circulación Colateral/fisiología , Técnicas de Transferencia de Gen , Hemorreología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Porcinos , Regulación hacia Arriba , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA