Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 38(24): e102155, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31721250

RESUMEN

Translation fidelity is crucial for prokaryotes and eukaryotic nuclear-encoded proteins; however, little is known about the role of mistranslation in mitochondria and its potential effects on metabolism. We generated yeast and mouse models with error-prone and hyper-accurate mitochondrial translation, and found that translation rate is more important than translational accuracy for cell function in mammals. Specifically, we found that mitochondrial mistranslation causes reduced overall mitochondrial translation and respiratory complex assembly rates. In mammals, this effect is compensated for by increased mitochondrial protein stability and upregulation of the citric acid cycle. Moreover, this induced mitochondrial stress signaling, which enables the recovery of mitochondrial translation via mitochondrial biogenesis, telomerase expression, and cell proliferation, and thereby normalizes metabolism. Conversely, we show that increased fidelity of mitochondrial translation reduces the rate of protein synthesis without eliciting a mitochondrial stress response. Consequently, the rate of translation cannot be recovered and this leads to dilated cardiomyopathy in mice. In summary, our findings reveal mammalian-specific signaling pathways that respond to changes in the fidelity of mitochondrial protein synthesis and affect metabolism.


Asunto(s)
Proliferación Celular , Mitocondrias/metabolismo , Biogénesis de Organelos , Transducción de Señal , Animales , Ciclo del Ácido Cítrico/fisiología , Escherichia coli/metabolismo , Femenino , Metabolómica , Ratones , Ratones Transgénicos , Mitocondrias/genética , Enfermedades Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Biosíntesis de Proteínas , Proteómica , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(37): 23113-23124, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32859761

RESUMEN

Currently there is an unmet need for treatments that can prevent hypertrophic cardiomyopathy (HCM). Using a murine model we previously identified that HCM causing cardiac troponin I mutation Gly203Ser (cTnI-G203S) is associated with increased mitochondrial metabolic activity, consistent with the human condition. These alterations precede development of the cardiomyopathy. Here we examine the efficacy of in vivo treatment of cTnI-G203S mice with a peptide derived against the α-interaction domain of the cardiac L-type calcium channel (AID-TAT) on restoring mitochondrial metabolic activity, and preventing HCM. cTnI-G203S or age-matched wt mice were treated with active or inactive AID-TAT. Following treatment, targeted metabolomics was utilized to evaluate myocardial substrate metabolism. Cardiac myocyte mitochondrial metabolic activity was assessed as alterations in mitochondrial membrane potential and flavoprotein oxidation. Cardiac morphology and function were examined using echocardiography. Cardiac uptake was assessed using an in vivo multispectral imaging system. We identified alterations in six biochemical intermediates in cTnI-G203S hearts consistent with increased anaplerosis. We also reveal that AID-TAT treatment of precardiomyopathic cTnI-G203S mice, but not mice with established cardiomyopathy, restored cardiac myocyte mitochondrial membrane potential and flavoprotein oxidation, and prevented myocardial hypertrophy. Importantly, AID-TAT was rapidly targeted to the heart, and not retained by the liver or kidneys. Overall, we identify biomarkers of HCM resulting from the cTnI mutation Gly203Ser, and present a safe, preventative therapy for associated cardiomyopathy. Utilizing AID-TAT to modulate cardiac metabolic activity may be beneficial in preventing HCM in "at risk" patients with identified Gly203Ser gene mutations.


Asunto(s)
Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación/genética , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Péptidos/farmacología , Troponina I/metabolismo
3.
J Mol Cell Cardiol ; 172: 100-108, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36041287

RESUMEN

Cardiovascular disease continues to be the leading health burden worldwide and with the rising rates in obesity and type II diabetes and ongoing effects of long COVID, it is anticipated that the burden of cardiovascular morbidity and mortality will increase. Calcium is essential to cardiac excitation and contraction. The main route for Ca2+ influx is the L-type Ca2+ channel (Cav1.2) and embryos that are homozygous null for the Cav1.2 gene are lethal at day 14 postcoitum. Acute changes in Ca2+ influx through the channel contribute to arrhythmia and sudden death, and chronic increases in intracellular Ca2+ contribute to pathological hypertrophy and heart failure. We use a multidisciplinary approach to study the regulation of the channel from the molecular level through to in vivo CRISPR mutant animal models. Here we describe some examples of our work from over 2 decades studying the role of the channel under physiological and pathological conditions. Our single channel analysis of purified human Cav1.2 protein in proteoliposomes has contributed to understanding direct molecular regulation of the channel including identifying the critical serine involved in the "fight or flight" response. Using the same approach we identified the cysteine responsible for altered function during oxidative stress. Chronic activation of the L-type Ca2+ channel during oxidative stress occurs as a result of persistent glutathionylation of the channel that contributes to the development of hypertrophy. We describe for the first time that activation of the channel alters mitochondrial function (and energetics) on a beat-to-beat basis via movement of cytoskeletal proteins. In translational studies we have used this response to "report" mitochondrial function in models of cardiomyopathy and to test efficacy of novel therapies to prevent cardiomyopathy.


Asunto(s)
Canales de Calcio Tipo L , Cardiomiopatías , Animales , Humanos , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Cardiomiopatías/metabolismo , COVID-19 , Diabetes Mellitus Tipo 2/metabolismo , Hipertrofia/metabolismo , Miocitos Cardíacos/metabolismo , Síndrome Post Agudo de COVID-19
4.
J Physiol ; 599(14): 3449-3462, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32710561

RESUMEN

The evolutionary acquisition of mitochondria has given rise to the diversity of eukaryotic life. Mitochondria have retained their ancestral α-proteobacterial traits through the maintenance of double membranes and their own circular genome. Their genome varies in size from very large in plants to the smallest in animals and their parasites. The mitochondrial genome encodes essential genes for protein synthesis and has to coordinate its expression with the nuclear genome from which it sources most of the proteins required for mitochondrial biogenesis and function. The mitochondrial protein synthesis machinery is unique because it is encoded by both the nuclear and mitochondrial genomes thereby requiring tight regulation to produce the respiratory complexes that drive oxidative phosphorylation for energy production. The fidelity and coordination of mitochondrial protein synthesis are essential for ATP production. Here we compare and contrast the mitochondrial translation mechanisms in mammals and fungi to bacteria and reveal that their diverse regulation can have unusual impacts on the health and disease of these organisms. We highlight that in mammals the rate of protein synthesis is more important than the fidelity of translation, enabling coordinated biogenesis of the mitochondrial respiratory chain with respiratory chain proteins synthesised by cytoplasmic ribosomes. Changes in mitochondrial protein fidelity can trigger the activation of the diverse cellular signalling networks in fungi and mammals to combat dysfunction in energy conservation. The physiological consequences of altered fidelity of protein synthesis can range from liver regeneration to the onset and development of cardiomyopathy.


Asunto(s)
Genoma Mitocondrial , Biosíntesis de Proteínas , Animales , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Ribosomas/metabolismo
5.
Pflugers Arch ; 472(1): 61-74, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31822999

RESUMEN

Neuronal nitric oxide synthase (nNOS) is considered a regulator of Cav1.2 L-type Ca2+ channels and downstream Ca2+ cycling in the heart. The commonest view is that nitric oxide (NO), generated by nNOS activity in cardiomyocytes, reduces the currents through Cav1.2 channels. This gives rise to a diminished Ca2+ release from the sarcoplasmic reticulum, and finally reduced contractility. Here, we report that nNOS inhibitor substances significantly increase intracellular Ca2+ transients in ventricular cardiomyocytes derived from adult mouse and rat hearts. This is consistent with an inhibitory effect of nNOS/NO activity on Ca2+ cycling and contractility. Whole cell currents through L-type Ca2+ channels in rodent myocytes, on the other hand, were not substantially affected by the application of various NOS inhibitors, or application of a NO donor substance. Moreover, the presence of NO donors had no effect on the single-channel open probability of purified human Cav1.2 channel protein reconstituted in artificial liposomes. These results indicate that nNOS/NO activity does not directly modify Cav1.2 channel function. We conclude that-against the currently prevailing view-basal Cav1.2 channel activity in ventricular cardiomyocytes is not substantially regulated by nNOS activity and NO. Hence, nNOS/NO inhibition of Ca2+ cycling and contractility occurs independently of direct regulation of Cav1.2 channels by NO.


Asunto(s)
Potenciales de Acción , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Miocitos Cardíacos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Femenino , Ventrículos Cardíacos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ornitina/análogos & derivados , Ornitina/farmacología , Ratas , Ratas Sprague-Dawley
6.
EMBO Rep ; 19(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30126926

RESUMEN

The molecular roles of the dually targeted ElaC domain protein 2 (ELAC2) during nuclear and mitochondrial RNA processing in vivo have not been distinguished. We generated conditional knockout mice of ELAC2 to identify that it is essential for life and its activity is non-redundant. Heart and skeletal muscle-specific loss of ELAC2 causes dilated cardiomyopathy and premature death at 4 weeks. Transcriptome-wide analyses of total RNAs, small RNAs, mitochondrial RNAs, and miRNAs identified the molecular targets of ELAC2 in vivo We show that ELAC2 is required for processing of tRNAs and for the balanced maintenance of C/D box snoRNAs, miRNAs, and a new class of tRNA fragments. We identify that correct biogenesis of regulatory non-coding RNAs is essential for both cytoplasmic and mitochondrial protein synthesis and the assembly of mitochondrial ribosomes and cytoplasmic polysomes. We show that nuclear tRNA processing is required for the balanced production of snoRNAs and miRNAs for gene expression and that 3' tRNA processing is an essential step in the production of all mature mitochondrial RNAs and the majority of nuclear tRNAs.


Asunto(s)
Endorribonucleasas/genética , Proteínas de Neoplasias/genética , ARN Mitocondrial/genética , ARN no Traducido/genética , Animales , Núcleo Celular/genética , Perfilación de la Expresión Génica , Ratones , MicroARNs/genética , ARN Nucleolar Pequeño/genética , ARN de Transferencia/genética , ARN no Traducido/clasificación , ARN no Traducido/aislamiento & purificación
7.
Nanomedicine ; 29: 102264, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32659322

RESUMEN

Therapeutic approaches for myocardial ischemia-reperfusion injury (MI) have been ineffective due to limited bioavailability and poor specificity. We have previously shown that a peptide that targets the α-interaction domain of the cardiac L-type calcium channel (AID-peptide) attenuates MI when tethered to transactivator of transcription sequence (TAT) or spherical nanoparticles. However some reservations remain regarding use of these delivery platforms due to the relationship with human immunodeficiency virus, off-target effects and toxicity. Here we investigate the use of linear dendronized polymers (denpols) to deliver AID-peptide as a potential MI therapy using in vitro, ex vivo and in vivo models. Optimized denpol-complexed AID-peptide facilitated in vitro cardiac uptake of AID-peptide, and reduced MI. Maximal in vivo cardiac uptake was achieved within the 2 h therapeutic time window for acute myocardial infarction. Importantly, optimized denpol-complexed AID-peptide was not toxic. This platform may represent an alternative therapeutic approach for the prevention of MI.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/genética , Corazón/efectos de los fármacos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Nanopartículas/química , Animales , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo L/efectos de los fármacos , Modelos Animales de Enfermedad , Cobayas , Corazón/fisiopatología , Humanos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Péptidos/química , Péptidos/farmacología , Polímeros/química , Polímeros/farmacología
8.
Heart Lung Circ ; 29(11): 1588-1595, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32839116

RESUMEN

BACKGROUND: Cardiovascular disease is the leading cause of death in Australia. Investment in research solutions has been demonstrated to yield health and a 9.8-fold return economic benefit. The sector, however, is severely challenged with success rates of traditional peer-reviewed funding in decline. Here, we aimed to understand the perceived challenges faced by the cardiovascular workforce in Australia prior to the COVID-19 pandemic. METHODS: We used an online survey distributed across Australian cardiovascular societies/councils, universities and research institutes over a period of 6 months during 2019, with 548 completed responses. Inclusion criteria included being an Australian resident or an Australian citizen who lived overseas, and a current or past student or employee in the field of cardiovascular research. RESULTS: The mean age of respondents was 42±13 years, 47% were male, 85% had a full-time position, and 40% were a group leader or laboratory head. Twenty-three per cent (23%) had permanent employment, and 82% of full-time workers regularly worked >40 hours/week. Sixty-eight per cent (68%) said they had previously considered leaving the cardiovascular research sector. If their position could not be funded in the next few years, a staggering 91% of respondents would leave the sector. Compared to PhD- and age-matched men, women were less likely to be a laboratory head and to feel they had a long-term career path as a cardiovascular researcher, while more women were unsure about future employment and had considered leaving the sector (all p<0.05). Greater job security (76%) and government and philanthropic investment in cardiovascular research (72%) were highlighted by responders as the main changes to current practices that would encourage them to stay. CONCLUSION: Strategic solutions, such as diversification of career pathways and funding sources, and moving from a competitive to a collaborative culture, need to be a priority to decrease reliance on government funding and allow cardiovascular researchers to thrive.


Asunto(s)
Investigación Biomédica , Enfermedades Cardiovasculares , Infecciones por Coronavirus/epidemiología , Administración Financiera , Neumonía Viral/epidemiología , Investigadores , Apoyo a la Investigación como Asunto , Recursos Humanos , Adulto , Australia , Betacoronavirus , Investigación Biomédica/economía , Investigación Biomédica/organización & administración , Investigación Biomédica/tendencias , COVID-19 , Empleo/economía , Empleo/psicología , Femenino , Administración Financiera/métodos , Administración Financiera/organización & administración , Administración Financiera/estadística & datos numéricos , Financiación Gubernamental , Humanos , Masculino , Cultura Organizacional , Pandemias , Técnicas de Planificación , Investigadores/economía , Investigadores/psicología , Investigadores/estadística & datos numéricos , Apoyo a la Investigación como Asunto/organización & administración , Apoyo a la Investigación como Asunto/tendencias , SARS-CoV-2 , Encuestas y Cuestionarios , Recursos Humanos/estadística & datos numéricos
9.
Arch Biochem Biophys ; 665: 166-174, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30885674

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a primary myocardial disorder, characterised by myocyte remodeling, disorganisation of sarcomeric proteins, impaired energy metabolism and altered cardiac contractility. Gene mutations encoding cardiac contractile proteins account for 60% of HCM aetiology. Current drug therapy including L-type calcium channel antagonists, are used to manage symptoms in patients with overt HCM, but no treatment exists that can reverse or prevent the cardiomyopathy. Design of effective drug therapy will require a clear understanding of the early pathophysiological mechanisms of the disease. Numerous studies have investigated specific aspects of HCM pathophysiology. This review brings these findings together, in order to develop a holistic understanding of the early pathophysiological mechanisms of the disease. We focus on gene mutations in cardiac myosin binding protein-C, ß-cardiac myosin heavy chain, cardiac troponin I, and cardiac troponin T, that comprise the majority of all HCM sarcomeric gene mutations. We find that although some similarities exist, each mutation leads to mutation-specific alterations in calcium handling, myofilament calcium sensitivity and mitochondrial metabolic function. This may contribute to the observed clinical phenotypic variability in sarcomeric-related HCM. An understanding of early mutation-specific mechanisms of the disease may provide useful markers of disease progression, and inform therapeutic design.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Mitocondrias Cardíacas/metabolismo , Animales , Biomarcadores/metabolismo , Humanos , Troponina T/metabolismo
11.
PLoS Genet ; 11(3): e1005089, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25816300

RESUMEN

The evolutionary divergence of mitochondrial ribosomes from their bacterial and cytoplasmic ancestors has resulted in reduced RNA content and the acquisition of mitochondria-specific proteins. The mitochondrial ribosomal protein of the small subunit 34 (MRPS34) is a mitochondria-specific ribosomal protein found only in chordates, whose function we investigated in mice carrying a homozygous mutation in the nuclear gene encoding this protein. The Mrps34 mutation causes a significant decrease of this protein, which we show is required for the stability of the 12S rRNA, the small ribosomal subunit and actively translating ribosomes. The synthesis of all 13 mitochondrially-encoded polypeptides is compromised in the mutant mice, resulting in reduced levels of mitochondrial proteins and complexes, which leads to decreased oxygen consumption and respiratory complex activity. The Mrps34 mutation causes tissue-specific molecular changes that result in heterogeneous pathology involving alterations in fractional shortening of the heart and pronounced liver dysfunction that is exacerbated with age. The defects in mitochondrial protein synthesis in the mutant mice are caused by destabilization of the small ribosomal subunit that affects the stability of the mitochondrial ribosome with age.


Asunto(s)
Cardiopatías Congénitas/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Ribosomas Mitocondriales/metabolismo , Proteínas Ribosómicas/biosíntesis , Animales , ADN Mitocondrial/genética , Metabolismo Energético , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Humanos , Hepatopatías/genética , Hepatopatías/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Ribosomas Mitocondriales/patología , Mutación , ARN Ribosómico/genética , Proteínas Ribosómicas/genética , Saccharomyces cerevisiae/genética
12.
Clin Exp Pharmacol Physiol ; 44 Suppl 1: 46-54, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28306174

RESUMEN

Cardiovascular disease is the leading cause of death in the Western world. The incidence of cardiovascular disease is predicted to further rise with the increase in obesity and diabetes and with the aging population. Even though the survival rate from ischaemic heart disease has improved over the past 30 years, many patients progress to a chronic pathological condition, known as cardiac hypertrophy that is associated with an increase in morbidity and mortality. Reactive oxygen species (ROS) and calcium play an essential role in mediating cardiac hypertrophy. The L-type calcium channel is the main route for calcium influx into cardiac myocytes. There is now good evidence for a direct role for the L-type calcium channel in the development of cardiac hypertrophy. Cysteines on the channel are targets for redox modification and glutathionylation of the channel can modulate the function of the channel protein leading to the onset of pathology. The cysteine responsible for modification of L-type calcium channel function has now been identified. Detailed understanding of the role of cysteines as possible targets during oxidative stress may assist in designing therapy to prevent the development of hypertrophy and heart failure.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Cardiomegalia/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Remodelación Ventricular , Animales , Canales de Calcio Tipo L/química , Cardiomegalia/diagnóstico , Cardiomegalia/fisiopatología , Cisteína , Humanos , Miocitos Cardíacos/patología , Oxidación-Reducción , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
13.
Proc Natl Acad Sci U S A ; 111(28): E2905-14, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24969422

RESUMEN

Duchenne muscular dystrophy is a fatal X-linked disease characterized by the absence of dystrophin. Approximately 20% of boys will die of dilated cardiomyopathy that is associated with cytoskeletal protein disarray, contractile dysfunction, and reduced energy production. However, the mechanisms for altered energy metabolism are not yet fully clarified. Calcium influx through the L-type Ca(2+) channel is critical for maintaining cardiac excitation and contraction. The L-type Ca(2+) channel also regulates mitochondrial function and metabolic activity via transmission of movement of the auxiliary beta subunit through intermediate filament proteins. Here, we find that activation of the L-type Ca(2+) channel is unable to induce increases in mitochondrial membrane potential and metabolic activity in intact cardiac myocytes from the murine model of Duchenne muscular dystrophy (mdx) despite robust increases recorded in wt myocytes. Treatment of mdx mice with morpholino oligomers to induce exon skipping of dystrophin exon 23 (that results in functional dystrophin accumulation) or application of a peptide that resulted in block of voltage-dependent anion channel (VDAC) "rescued" mitochondrial membrane potential and metabolic activity in mdx myocytes. The mitochondrial VDAC coimmunoprecipitated with the L-type Ca(2+) channel. We conclude that the absence of dystrophin in the mdx ventricular myocyte leads to impaired functional communication between the L-type Ca(2+) channel and mitochondrial VDAC. This appears to contribute to metabolic inhibition. These findings provide new mechanistic and functional insight into cardiomyopathy associated with Duchenne muscular dystrophy.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Distrofina/metabolismo , Potencial de la Membrana Mitocondrial , Membranas Mitocondriales/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Calcio/metabolismo , Distrofina/antagonistas & inhibidores , Distrofina/genética , Exones , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Masculino , Ratones , Ratones Endogámicos mdx , Membranas Mitocondriales/patología , Morfolinos/genética , Morfolinos/farmacología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Miocardio , Miocitos Cardíacos/patología
14.
J Physiol ; 599(14): 3447-3448, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34263447

Asunto(s)
Mitocondrias , Humanos
15.
J Biol Chem ; 290(3): 1729-42, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25451916

RESUMEN

The maintenance of bone homeostasis requires tight coupling between bone-forming osteoblasts and bone-resorbing osteoclasts. However, the precise molecular mechanism(s) underlying the differentiation and activities of these specialized cells are still largely unknown. Here, we identify choline kinase ß (CHKB), a kinase involved in the biosynthesis of phosphatidylcholine, as a novel regulator of bone homeostasis. Choline kinase ß mutant mice (flp/flp) exhibit a systemic low bone mass phenotype. Consistently, osteoclast numbers and activity are elevated in flp/flp mice. Interestingly, osteoclasts derived from flp/flp mice exhibit reduced sensitivity to excessive levels of extracellular calcium, which could account for the increased bone resorption. Conversely, supplementation of cytidine 5'-diphosphocholine in vivo and in vitro, a regimen that bypasses CHKB deficiency, restores osteoclast numbers to physiological levels. Finally, we demonstrate that, in addition to modulating osteoclast formation and function, loss of CHKB corresponds with a reduction in bone formation by osteoblasts. Taken together, these data posit CHKB as a new modulator of bone homeostasis.


Asunto(s)
Colina Quinasa/genética , Mutación , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Fosforilcolina/metabolismo , Animales , Densidad Ósea , Resorción Ósea , Huesos/metabolismo , Calcio/metabolismo , Proliferación Celular , Homeostasis , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Mutagénesis , Osteoblastos/citología , Osteoclastos/citología , Fenotipo , Microtomografía por Rayos X
16.
Acta Derm Venereol ; 96(6): 774-8, 2016 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-26911400

RESUMEN

A double-blind randomized controlled trial with a paired split-scar design compared verapamil, an L-type Ca2+ channel antagonist, and triamcinolone for prevention of keloid recurrence after excision. Ca2+ channel blocking activity of verapamil in keloid cells was explored. One keloid was excised per subject and each wound half randomized to receive intralesional injections of triamcinolone (10 mg/ml) or verapamil (2.5 mg/ml) at monthly intervals (4 doses). Interim analysis was performed after 14 subjects were completed. Survival analysis demonstrated significantly higher keloid recurrence with verapamil compared to triamcinolone 12 months post-surgery (log-rank test, p = 0.01) and higher overall risk of recurrence with verapamil (hazard ratio 8.44, 95% CI 1.62-44.05). The study was terminated early according to the stopping guideline (p < 0.05). Verapamil is safe but not as effective as triamcinolone in preventing keloid recurrence after excision. Further study is necessary to determine if clinical response to verapamil is linked to modulation of intracellular Ca2+.


Asunto(s)
Cicatriz/prevención & control , Glucocorticoides/uso terapéutico , Queloide/tratamiento farmacológico , Queloide/cirugía , Triamcinolona Acetonida/uso terapéutico , Vasodilatadores/uso terapéutico , Verapamilo/uso terapéutico , Adolescente , Adulto , Método Doble Ciego , Femenino , Glucocorticoides/administración & dosificación , Humanos , Inyecciones Intralesiones , Masculino , Persona de Mediana Edad , Recurrencia , Resultado del Tratamiento , Triamcinolona Acetonida/administración & dosificación , Vasodilatadores/administración & dosificación , Verapamilo/administración & dosificación
18.
J Physiol ; 593(6): 1331-45, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25772289

RESUMEN

In February 2014, a group of scientists convened as part of the University of California Davis Cardiovascular Symposium to bring together experimental and mathematical modelling perspectives and discuss points of consensus and controversy on the topic of sodium in the heart. This paper summarizes the topics of presentation and discussion from the symposium, with a focus on the role of aberrant sodium channels and abnormal sodium homeostasis in cardiac arrhythmias and pharmacotherapy from the subcellular scale to the whole heart. Two following papers focus on Na(+) channel structure, function and regulation, and Na(+)/Ca(2+) exchange and Na(+)/K(+) ATPase. The UC Davis Cardiovascular Symposium is a biannual event that aims to bring together leading experts in subfields of cardiovascular biomedicine to focus on topics of importance to the field. The focus on Na(+) in the 2014 symposium stemmed from the multitude of recent studies that point to the importance of maintaining Na(+) homeostasis in the heart, as disruption of homeostatic processes are increasingly identified in cardiac disease states. Understanding how disruption in cardiac Na(+)-based processes leads to derangement in multiple cardiac components at the level of the cell and to then connect these perturbations to emergent behaviour in the heart to cause disease is a critical area of research. The ubiquity of disruption of Na(+) channels and Na(+) homeostasis in cardiac disorders of excitability and mechanics emphasizes the importance of a fundamental understanding of the associated mechanisms and disease processes to ultimately reveal new targets for human therapy.


Asunto(s)
Síndrome de Brugada/metabolismo , Paro Cardíaco/metabolismo , Sodio/metabolismo , Animales , Síndrome de Brugada/fisiopatología , Congresos como Asunto , Paro Cardíaco/fisiopatología , Humanos
19.
Circ Res ; 112(5): 781-91, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23303165

RESUMEN

RATIONALE: Regulator of G-protein signaling 5 (RGS5) modulates G-protein-coupled receptor signaling and is prominently expressed in arterial smooth muscle cells. Our group first reported that RGS5 is important in vascular remodeling during tumor angiogenesis. We hypothesized that RGS5 may play an important role in vessel wall remodeling and blood pressure regulation. OBJECTIVE: To demonstrate that RGS5 has a unique and nonredundant role in the pathogenesis of hypertension and to identify crucial RGS5-regulated signaling pathways. METHODS AND RESULTS: We observed that arterial RGS5 expression is downregulated with chronically elevated blood pressure after angiotensin II infusion. Using a knockout mouse model, radiotelemetry, and pharmacological inhibition, we subsequently showed that loss of RGS5 results in profound hypertension. RGS5 signaling is linked to the renin-angiotensin system and directly controls vascular resistance, vessel contractility, and remodeling. RGS5 deficiency aggravates pathophysiological features of hypertension, such as medial hypertrophy and fibrosis. Moreover, we demonstrate that protein kinase C, mitogen-activated protein kinase/extracellular signal-regulated kinase, and Rho kinase signaling pathways are major effectors of RGS5-mediated hypertension. CONCLUSIONS: Loss of RGS5 results in hypertension. Loss of RGS5 signaling also correlates with hyper-responsiveness to vasoconstrictors and vascular stiffening. This establishes a significant, distinct, and causal role of RGS5 in vascular homeostasis. RGS5 modulates signaling through the angiotensin II receptor 1 and major Gαq-coupled downstream pathways, including Rho kinase. So far, activation of RhoA/Rho kinase has not been associated with RGS molecules. Thus, RGS5 is a crucial regulator of blood pressure homeostasis with significant clinical implications for vascular pathologies, such as hypertension.


Asunto(s)
Presión Sanguínea/fisiología , Vasos Sanguíneos/fisiología , Homeostasis/fisiología , Músculo Liso Vascular/fisiología , Proteínas RGS/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Hipertensión/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa C/fisiología , Proteínas RGS/deficiencia , Proteínas RGS/genética , Transducción de Señal/fisiología , Vasoconstricción/fisiología , Quinasas Asociadas a rho/fisiología
20.
Int J Mol Sci ; 15(10): 19203-25, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25340983

RESUMEN

There is mounting evidence to suggest that protein glutathionylation is a key process contributing to the development of pathology. Glutathionylation occurs as a result of posttranslational modification of a protein and involves the addition of a glutathione moiety at cysteine residues. Such modification can occur on a number of proteins, and exerts a variety of functional consequences. The L-type Ca2+ channel has been identified as a glutathionylation target that participates in the development of cardiac pathology. Ca2+ influx via the L-type Ca2+ channel increases production of mitochondrial reactive oxygen species (ROS) in cardiomyocytes during periods of oxidative stress. This induces a persistent increase in channel open probability, and the resulting constitutive increase in Ca2+ influx amplifies the cross-talk between the mitochondria and the channel. Novel strategies utilising targeted peptide delivery to uncouple mitochondrial ROS and Ca2+ flux via the L-type Ca2+ channel following ischemia-reperfusion have delivered promising results, and have proven capable of restoring appropriate mitochondrial function in myocytes and in vivo.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Glutatión/metabolismo , Corazón/fisiopatología , Miocitos Cardíacos/patología , Estrés Oxidativo/fisiología , Animales , Humanos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA