Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 5(10): 1787-1802, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37679556

RESUMO

Neuroinflammatory microglia secrete cytokines to induce neurotoxic reactive astrocytes, which are one of the major causes of neuronal death. However, the intrinsic key regulators underlying neurotoxic reactive astrocytes induction are unknown. Here we show that the transmembrane protein 164 (TMEM164) is an early-response intrinsic factor that regulates neurotoxic astrocyte reactivity. TMEM164 overexpression inhibits the induction of neurotoxic reactive astrocytes, maintains normal astrocytic functions and suppresses neurotoxic reactive astrocyte-mediated neuronal death by decreasing the secretion of neurotoxic saturated lipids. Adeno-associated virus-mediated, astrocyte-specific TMEM164 overexpression in male and female mice prevents the induction of neurotoxic reactive astrocytes, dopaminergic neuronal loss and motor deficits in a Parkinson's disease model. Notably, brain-wide astrocyte-specific TMEM164 overexpression prevents the induction of neurotoxic reactive astrocytes, amyloid ß deposition, neurodegeneration and memory decline in the 5XFAD Alzheimer's disease mouse model, suggesting that TMEM164 could serve as a potential therapeutic target for neurodegenerative disorders.


Assuntos
Doença de Alzheimer , Astrócitos , Feminino , Camundongos , Animais , Masculino , Astrócitos/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/metabolismo , Microglia/metabolismo , Neurônios/metabolismo
2.
Cancer Discov ; 8(2): 196-215, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29101162

RESUMO

Ex vivo systems that incorporate features of the tumor microenvironment and model the dynamic response to immune checkpoint blockade (ICB) may facilitate efforts in precision immuno-oncology and the development of effective combination therapies. Here, we demonstrate the ability to interrogate ex vivo response to ICB using murine- and patient-derived organotypic tumor spheroids (MDOTS/PDOTS). MDOTS/PDOTS isolated from mouse and human tumors retain autologous lymphoid and myeloid cell populations and respond to ICB in short-term three-dimensional microfluidic culture. Response and resistance to ICB was recapitulated using MDOTS derived from established immunocompetent mouse tumor models. MDOTS profiling demonstrated that TBK1/IKKε inhibition enhanced response to PD-1 blockade, which effectively predicted tumor response in vivo Systematic profiling of secreted cytokines in PDOTS captured key features associated with response and resistance to PD-1 blockade. Thus, MDOTS/PDOTS profiling represents a novel platform to evaluate ICB using established murine models as well as clinically relevant patient specimens.Significance: Resistance to PD-1 blockade remains a challenge for many patients, and biomarkers to guide treatment are lacking. Here, we demonstrate feasibility of ex vivo profiling of PD-1 blockade to interrogate the tumor immune microenvironment, develop therapeutic combinations, and facilitate precision immuno-oncology efforts. Cancer Discov; 8(2); 196-215. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Deng et al., p. 216This article is highlighted in the In This Issue feature, p. 127.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Citocinas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Imunofenotipagem , Camundongos , Técnicas Analíticas Microfluídicas , Receptor de Morte Celular Programada 1/metabolismo , Esferoides Celulares , Imagem com Lapso de Tempo , Células Tumorais Cultivadas
3.
J Am Heart Assoc ; 3(3): e000527, 2014 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-24958777

RESUMO

BACKGROUND: A highly organized transverse tubule (T-tubule) network is necessary for efficient Ca(2+)-induced Ca(2+) release and synchronized contraction of ventricular myocytes. Increasing evidence suggests that T-tubule remodeling due to junctophilin-2 (JP-2) downregulation plays a critical role in the progression of heart failure. However, the mechanisms underlying JP-2 dysregulation remain incompletely understood. METHODS AND RESULTS: A mouse model of reversible heart failure that is driven by conditional activation of the heterotrimeric G protein Gαq in cardiac myocytes was used in this study. Mice with activated Gαq exhibited disruption of the T-tubule network and defects in Ca(2+) handling that culminated in heart failure compared with wild-type mice. Activation of Gαq/phospholipase Cß signaling increased the activity of the Ca(2+)-dependent protease calpain, leading to the proteolytic cleavage of JP-2. A novel calpain cleavage fragment of JP-2 is detected only in hearts with constitutive Gαq signaling to phospholipase Cß. Termination of the Gαq signal was followed by normalization of the JP-2 protein level, repair of the T-tubule network, improvements in Ca(2+) handling, and reversal of heart failure. Treatment of mice with a calpain inhibitor prevented Gαq-dependent JP-2 cleavage, T-tubule disruption, and the development of heart failure. CONCLUSIONS: Disruption of the T-tubule network in heart failure is a reversible process. Gαq-dependent activation of calpain and subsequent proteolysis of JP-2 appear to be the molecular mechanism that leads to T-tubule remodeling, Ca(2+) handling dysfunction, and progression to heart failure in this mouse model.


Assuntos
Calpaína/fisiologia , Insuficiência Cardíaca/fisiopatologia , Proteínas de Membrana/fisiologia , Proteínas Musculares/fisiologia , Animais , Cálcio/metabolismo , Notificação de Doenças , Regulação para Baixo/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Musculares/fisiologia , Proteínas Musculares/metabolismo , Proteólise , Transdução de Sinais/fisiologia
4.
PLoS One ; 9(5): e96921, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24805850

RESUMO

Amyotrophic Lateral Sclerosis (ALS) is a motor neuron disease characterized by progressive motor neuron loss resulting in muscle atrophy, declining muscle function, and eventual paralysis. Patients typically die from respiratory failure 3 to 5 years from the onset of symptoms. Tirasemtiv is a fast skeletal troponin activator that sensitizes the sarcomere to calcium; this mechanism of action amplifies the response of muscle to neuromuscular input producing greater force when nerve input is reduced. Here, we demonstrate that a single dose of tirasemtiv significantly increases submaximal isometric force, forelimb grip strength, grid hang time, and rotarod performance in a female transgenic mouse model (B6SJL-SOD1 G93A) of ALS with functional deficits. Additionally, diaphragm force and tidal volume are significantly higher in tirasemtiv-treated female B6SJL-SOD1 G93A mice. These results support the potential of fast skeletal troponin activators to improve muscle function in neuromuscular diseases.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Imidazóis/administração & dosagem , Neurônios Motores/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Pirazinas/administração & dosagem , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Neurônios Motores/patologia , Força Muscular/genética , Músculo Esquelético/efeitos dos fármacos , Troponina/genética , Troponina/metabolismo
5.
Circ Arrhythm Electrophysiol ; 5(4): 831-40, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22722661

RESUMO

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel is largely inactivated, contributing to slow conduction and reentry. We have demonstrated that adenoviral delivery of the skeletal muscle Na(+) channel (SkM1) to epicardial border zones normalizes conduction and reduces induction of ventricular tachycardia/ventricular fibrillation. We now studied the impact of canine mesenchymal stem cells (cMSCs) in delivering SkM1. METHODS AND RESULTS: cMSCs were isolated and transfected with SkM1. Coculture experiments showed cMSC/SkM1 but not cMSC alone and maintained fast conduction at depolarized potentials. We studied 3 groups in the canine 7d infarct: sham, cMSC, and cMSC/SkM1. In vivo epicardial border zones electrograms were broad and fragmented in sham, narrower in cMSCs, and narrow and unfragmented in cMSC/SkM1 (P<0.05). During programmed electrical stimulation of epicardial border zones, QRS duration in cMSC/SkM1 was shorter than in cMSC and sham (P<0.05). Programmed electrical stimulation-induced ventricular tachycardia/ventricular fibrillation was equivalent in all groups (P>0.05). CONCLUSION: cMSCs provide efficient delivery of SkM1 current. The interventions performed (cMSCs or cMSC/SkM1) were neither antiarrhythmic nor proarrhythmic. Comparing outcomes with cMSC/SkM1 and viral gene delivery highlights the criticality of the delivery platform to SkM1 antiarrhythmic efficacy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Proteínas Musculares/metabolismo , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Taquicardia Ventricular/prevenção & controle , Fibrilação Ventricular/prevenção & controle , Potenciais de Ação , Animais , Animais Recém-Nascidos , Estimulação Cardíaca Artificial , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Cães , Técnicas Eletrofisiológicas Cardíacas , Humanos , Proteínas Musculares/genética , Infarto do Miocárdio/complicações , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5 , Ratos , Ratos Sprague-Dawley , Canais de Sódio/genética , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Transfecção , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/genética , Fibrilação Ventricular/metabolismo , Fibrilação Ventricular/fisiopatologia
6.
J Cardiovasc Pharmacol ; 60(1): 88-99, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22526298

RESUMO

The voltage-gated Na+ channel is a critical determinant of the action potential (AP) upstroke. Increasing Na+ conductance may speed AP propagation. In this study, we propose use of the skeletal muscle Na+ channel SkM1 as a more favorable gene than the cardiac isoform SCN5A to enhance conduction velocity in depolarized cardiac tissue. We used cells that electrically coupled with cardiac myocytes as a delivery platform to introduce the Na+ channels. Human embryonic kidney 293 cells were stably transfected with SkM1 or SCN5A. SkM1 had a more depolarized (18 mV shift) inactivation curve than SCN5A. We also found that SkM1 recovered faster from inactivation than SCN5A. When coupled with SkM1 expressing cells, cultured myocytes showed an increase in the dV/dtmax of the AP. Expression of SCN5A had no such effect. In an in vitro cardiac syncytium, coculture of neonatal cardiac myocytes with SkM1 expressing but not SCN5A expressing cells significantly increased the conduction velocity under both normal and depolarized conditions. In an in vitro reentry model induced by high-frequency stimulation, expression of SkM1 also enhanced angular velocity of the induced reentry. These results suggest that cells carrying a Na+ channel with a more depolarized inactivation curve can improve cardiac excitability and conduction in depolarized tissues.


Assuntos
Músculo Esquelético/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Potenciais de Ação , Animais , Animais Recém-Nascidos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Cães , Feminino , Terapia Genética/métodos , Células HEK293 , Sistema de Condução Cardíaco/metabolismo , Humanos , Masculino , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção
7.
Biophys J ; 102(6): 1294-302, 2012 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-22455912

RESUMO

Recent experimental and modeling studies demonstrate the fine spatial scale, complex nature, and independent contribution of Ca(2+) dynamics as a proarrhythmic factor in the heart. The mechanism of progression of cell-level Ca(2+) instabilities, known as alternans, to tissue-level arrhythmias is not well understood. Because gap junction coupling dictates cardiac syncytial properties, we set out to elucidate its role in the spatiotemporal evolution of Ca(2+) instabilities. We experimentally perturbed cellular coupling in cardiac syncytium in vitro. Coupling was quantified by fluorescence recovery after photobleaching, and related to function, including subtle fine-scale Ca(2+) alternans, captured by optical mapping. Conduction velocity and threshold for alternans monotonically increased with coupling. Lower coupling enhanced Ca(2+) alternans amplitude, but the spatial spread of early (<2 Hz) alternation was the greatest under intermediate (not low) coupling. This nonmonotonic relationship was closely matched by the percent of samples exhibiting large-scale alternans at higher pacing rates. Computer modeling corroborated these experimental findings for strong but not weak electromechanical (voltage-Ca(2+)) coupling, and offered mechanistic insight. In conclusion, using experimental and modeling approaches, we reveal a general mechanism for the spatial spread of subtle cellular Ca(2+) alternans that relies on a combination of gap-junctional and voltage-Ca(2+) coupling.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Espaço Intracelular/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Animais , Difusão , Recuperação de Fluorescência Após Fotodegradação , Células Gigantes/citologia , Células Gigantes/metabolismo , Cinética , Ratos , Ratos Sprague-Dawley
8.
Nat Med ; 18(3): 452-5, 2012 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-22344294

RESUMO

Limited neural input results in muscle weakness in neuromuscular disease because of a reduction in the density of muscle innervation, the rate of neuromuscular junction activation or the efficiency of synaptic transmission. We developed a small-molecule fast-skeletal-troponin activator, CK-2017357, as a means to increase muscle strength by amplifying the response of muscle when neural input is otherwise diminished secondary to neuromuscular disease. Binding selectively to the fast-skeletal-troponin complex, CK-2017357 slows the rate of calcium release from troponin C and sensitizes muscle to calcium. As a consequence, the force-calcium relationship of muscle fibers shifts leftwards, as does the force-frequency relationship of a nerve-muscle pair, so that CK-2017357 increases the production of muscle force in situ at sub-maximal nerve stimulation rates. Notably, we show that sensitization of the fast-skeletal-troponin complex to calcium improves muscle force and grip strength immediately after administration of single doses of CK-2017357 in a model of the neuromuscular disease myasthenia gravis. Troponin activation may provide a new therapeutic approach to improve physical activity in diseases where neuromuscular function is compromised.


Assuntos
Cálcio/metabolismo , Músculo Esquelético/metabolismo , Doenças Neuromusculares/metabolismo , Troponina C/agonistas , Troponina C/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Bovinos , Humanos , Imidazóis/química , Imidazóis/uso terapêutico , Terapia de Alvo Molecular , Contração Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/patologia , Miastenia Gravis/tratamento farmacológico , Miastenia Gravis/metabolismo , Miastenia Gravis/patologia , Miosinas/isolamento & purificação , Miosinas/metabolismo , Doenças Neuromusculares/tratamento farmacológico , Doenças Neuromusculares/patologia , Pirazinas/química , Pirazinas/uso terapêutico , Coelhos , Ratos , Troponina/metabolismo , Troponina/fisiologia
9.
PLoS One ; 6(9): e24404, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21912691

RESUMO

BACKGROUND: Phosphoinositide 3-kinases (PI3Ks) regulate numerous physiological processes including some aspects of cardiac function. Although regulation of cardiac contraction by individual PI3K isoforms has been studied, little is known about the cardiac consequences of downregulating multiple PI3Ks concurrently. METHODS AND RESULTS: Genetic ablation of both p110α and p110ß in cardiac myocytes throughout development or in adult mice caused heart failure and death. Ventricular myocytes from double knockout animals showed transverse tubule (T-tubule) loss and disorganization, misalignment of L-type Ca(2+) channels in the T-tubules with ryanodine receptors in the sarcoplasmic reticulum, and reduced Ca(2+) transients and contractility. Junctophilin-2, which is thought to tether T-tubules to the sarcoplasmic reticulum, was mislocalized in the double PI3K-null myocytes without a change in expression level. CONCLUSIONS: PI3K p110α and p110ß are required to maintain the organized network of T-tubules that is vital for efficient Ca(2+)-induced Ca(2+) release and ventricular contraction. PI3Ks maintain T-tubule organization by regulating junctophilin-2 localization. These results could have important medical implications because several PI3K inhibitors that target both isoforms are being used to treat cancer patients in clinical trials.


Assuntos
Miócitos Cardíacos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Sarcolema/metabolismo , Animais , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/genética , Deleção de Genes , Técnicas de Inativação de Genes , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Contração Muscular/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Transporte Proteico , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcolema/patologia
10.
Circ Arrhythm Electrophysiol ; 4(5): 753-60, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21828312

RESUMO

BACKGROUND: After the recent cloning of light-sensitive ion channels and their expression in mammalian cells, a new field, optogenetics, emerged in neuroscience, allowing for precise perturbations of neural circuits by light. However, functionality of optogenetic tools has not been fully explored outside neuroscience, and a nonviral, nonembryogenesis-based strategy for optogenetics has not been shown before. METHODS AND RESULTS: We demonstrate the utility of optogenetics to cardiac muscle by a tandem cell unit (TCU) strategy, in which nonexcitable cells carry exogenous light-sensitive ion channels, and, when electrically coupled to cardiomyocytes, produce optically excitable heart tissue. A stable channelrhodopsin2 (ChR2)-expressing cell line was developed, characterized, and used as a cell delivery system. The TCU strategy was validated in vitro in cell pairs with adult canine myocytes (for a wide range of coupling strengths) and in cardiac syncytium with neonatal rat cardiomyocytes. For the first time, we combined optical excitation and optical imaging to capture light-triggered muscle contractions and high-resolution propagation maps of light-triggered electric waves, found to be quantitatively indistinguishable from electrically triggered waves. CONCLUSIONS: Our results demonstrate feasibility to control excitation and contraction in cardiac muscle by light, using the TCU approach. Optical pacing in this case uses less energy, offers superior spatiotemporal control and remote access and can serve not only as an elegant tool in arrhythmia research but may form the basis for a new generation of light-driven cardiac pacemakers and muscle actuators. The TCU strategy is extendable to (nonviral) stem cell therapy and is directly relevant to in vivo applications.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Luz , Contração Muscular/fisiologia , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Comunicação Celular/fisiologia , Channelrhodopsins , Técnicas de Cocultura , Cães , Estimulação Elétrica , Estudos de Viabilidade , Células HEK293 , Humanos , Rim/citologia , Rim/metabolismo , Miócitos Cardíacos/citologia , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Transfecção
11.
J Pharmacol Exp Ther ; 339(1): 307-12, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21784887

RESUMO

We examined a novel therapeutic approach for hypertension, a small-molecule direct inhibitor of smooth muscle myosin, CK-2018448 (CK-448), which is an N,N'-alkylurea (U.S. Patent Publication 2009-0275537 A1) in conscious dogs with renal hypertension and compared its efficacy with that of a calcium channel blocker, amlodipine. Dogs were instrumented with a miniature left ventricular pressure gauge, an aortic pressure catheter, and ultrasonic flow probes in the ascending aorta and renal and iliac arteries for measurement of cardiac output and regional blood flow. In the hypertensive state, mean arterial pressure increased from 101 ± 3.8 to 142 ± 1.9 mm Hg. At the doses selected, CK-448 and amlodipine increased cardiac output similarly (30 ± 11% versus 33 ± 6.4%) and similarly reduced mean arterial pressure (-22 ± 3.6% versus -16 ± 3.4%) and total peripheral resistance (-36 ± 5.9% versus -37 ± 5.8%). CK-448 had the greatest vasodilator effect in the renal bed, where renal blood flow increased by 46 ± 9.0%, versus 11 ± 3.4% for amlodipine (p < 0.01). CK-488 produced significantly less vasodilation in the limb, where iliac blood flow did not change; in contrast, it rose by 48 ± 12% with amlodipine (p < 0.01). The minimal effects on limb blood flow could limit the development of peripheral edema, an adverse side effect of Ca(2+) channel blockers. In addition, in a rodent model of hypertension, oral administration of a smooth muscle myosin inhibitor resulted in a sustained antihypertensive effect. Thus, the smooth muscle myosin inhibitor's preferential effect on renal blood flow makes this drug mechanism particularly appealing, because many patients with hypertension have renal insufficiency, and patients with heart failure could benefit from afterload reduction coupled with enhanced renal blood flow.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão/tratamento farmacológico , Músculo Liso/efeitos dos fármacos , Miosinas/antagonistas & inibidores , Ureia/análogos & derivados , Anlodipino/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Cães , Feminino , Hemodinâmica/efeitos dos fármacos , Artéria Ilíaca/efeitos dos fármacos , Injeções Intravenosas , Ratos , Ratos Endogâmicos SHR , Fluxo Sanguíneo Regional/efeitos dos fármacos , Circulação Renal/efeitos dos fármacos , Ureia/farmacologia , Resistência Vascular/efeitos dos fármacos
12.
Phys Rev E Stat Nonlin Soft Matter Phys ; 81(5 Pt 2): 056202, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20866302

RESUMO

Obstacle-anchored vortices can be terminated by the application of high-frequency wave trains in excitable media. We theoretically derived the dependency between the obstacle radius and the maximum unpinning period through reinterpretation of the well-known eikonal equation. Our theoretical result was confirmed by experiments with cardiomyocyte monolayers. This result may be useful for improving the stimulation protocol of implantable cardiac pacemakers.

13.
IEEE Trans Biomed Eng ; 57(2): 316-24, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19695992

RESUMO

Time-alternating biological signals, i.e., alternans, arise in variety of physiological states marked by dynamic instabilities, e.g., period doubling. Normally, a sequence of large-small-large transients, they can exhibit variable patterns over time and space, including spatial discordance. Capture of the early formation of such alternating regions is challenging because of the spatiotemporal similarities between noise and the small-amplitude alternating signals close to the bifurcation point. We present a new approach for automatic detection of alternating signals in large noisy spatiotemporal datasets by exploiting quantitative measures of alternans evolution, e.g., temporal persistence, and by preserving phase information. The technique specifically targets low amplitude, relatively short alternating sequences and is validated by combinatorics-derived probabilities and empirical datasets with white noise. Using high-resolution optical mapping in live cardiomyocyte networks, exhibiting calcium alternans, we reveal for the first time early fine-scale alternans, close to the noise level, which are linked to the later formation of larger regions and evolution of spatially discordant alternans. This robust method aims at quantification and better understanding of the onset of cardiac arrhythmias and can be applied to general analysis of space-time alternating signals, including the vicinity of the bifurcation point.


Assuntos
Algoritmos , Modelos Biológicos , Processamento de Sinais Assistido por Computador , Bases de Dados Factuais , Eletrocardiografia/métodos , Miócitos Cardíacos/fisiologia , Reprodutibilidade dos Testes
14.
Cardiovasc Res ; 81(3): 528-35, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18977767

RESUMO

AIMS: Reentrant arrhythmias often develop in the setting of myocardial infarction and ensuing slow propagation. Increased Na(+) channel expression could prevent or disrupt reentrant circuits by speeding conduction if channel availability is not limited by membrane depolarization within the diseased myocardium. We therefore asked if, in the setting of membrane depolarization, action potential (AP) upstroke and normal conduction can be better preserved by the expression of a Na(+) channel isoform with altered biophysical properties compared to the native cardiac Na(+) channel isoform, namely having a positively shifted, voltage-dependent inactivation. METHODS AND RESULTS: The skeletal Na(+) channel isoform (SkM1) and the cardiac Na(+) channel isoform (Nav1.5) were expressed in newborn rat ventricular myocyte cultures with a point mutation introduced in Nav1.5 to increase tetrodotoxin (TTX) sensitivity so native and expressed currents could be distinguished. External K(+) was increased from 5.4 to 10 mmol/L to induce membrane depolarization. APs, Na(+) currents, and conduction velocity (CV) were measured. In control cultures, elevated K(+) significantly reduced AP upstroke ( approximately 75%) and CV ( approximately 25%). Expression of Nav1.5 did not protect AP upstroke from K(+) depolarization. In contrast, in SkM1 expressing cultures, high K(+) reduced AP upstroke <50% and conduction was not significantly reduced. In a simulated anatomical reentry setting (using a void), the angular velocity (AV) of induced reentry was faster and the excitable gap shorter in SkM1 cultures compared to control for both normal and high K(+). CONCLUSION: Expression of SkM1 but not Nav1.5 preserves AP upstroke and CV in a K(+)-depolarized syncytium. The higher AV and shorter excitable gap observed during reentry excitation around a void in SkM1 cultures would be expected to facilitate reentry self-termination. SkM1 Na(+) channel expression represents a novel gene therapy for the treatment of reentrant arrhythmias.


Assuntos
Arritmias Cardíacas/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Sódio/metabolismo , Potenciais de Ação , Animais , Animais Recém-Nascidos , Arritmias Cardíacas/genética , Arritmias Cardíacas/terapia , Células Cultivadas , Técnicas de Transferência de Genes , Terapia Genética , Ventrículos do Coração/metabolismo , Humanos , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5 , Mutação Puntual , Potássio/metabolismo , Ratos , Bloqueadores dos Canais de Sódio/farmacologia , Canais de Sódio/genética , Tetrodotoxina/farmacologia , Fatores de Tempo
15.
Artigo em Inglês | MEDLINE | ID: mdl-19162616

RESUMO

Alternans, a beat-to-beat alternation in cardiac signals, may serve as a precursor to lethal cardiac arrhythmias, including ventricular tachycardia and ventricular fibrillation. Therefore, alternans is a desirable target of early arrhythmia prediction/detection. For long-term records and in the presence of noise, the definition of alternans is qualitative and ambiguous. This makes their automatic detection in large spatiotemporal data sets almost impossible. We present here a quantitative combinatorics-derived definition of alternans in the presence of random noise and a novel algorithm for automatic alternans detection using criteria like temporal persistence (TP), representative phase (RP) and alternans ratio (AR). This technique is validated by comparison to theoretically-derived probabilities and by test data sets with white noise. Finally, the algorithm is applied to ultra-high resolution optical mapping data from cultured cell monolayers, exhibiting calcium alternans. Early fine-scale alternans, close to the noise level, were revealed and linked to the later formation of larger regions and evolution of spatially discordant alternans (SDA). This robust new technique can be useful in quantification and better understanding of the onset of arrhythmias and in general analysis of space-time alternating signals.


Assuntos
Algoritmos , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/fisiopatologia , Diagnóstico por Computador/métodos , Eletroencefalografia/métodos , Miócitos Cardíacos , Reconhecimento Automatizado de Padrão/métodos , Animais , Animais Recém-Nascidos , Relógios Biológicos , Mapeamento Potencial de Superfície Corporal/métodos , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
16.
Biochemistry ; 46(11): 3494-502, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17302440

RESUMO

The Wiskott-Aldrich syndrome protein (WASP) and neural WASP (N-WASP) are key players in regulating actin cytoskeleton via the Arp2/3 complex. It has been widely reported that the WASP proteins are activated by Rho family small GTPase Cdc42 and that Rac1 acts through SCAR/WAVE proteins. However, a systematic study of the specificity of different GTPases for different Arp2/3 activators has not been conducted. In this study, we have expressed, purified, and characterized completely soluble, highly active, and autoinhibited full-length human WASP and N-WASP from mammalian cells. We show a novel N-WASP activation by Rho family small GTPase Rac1. This GTPase exclusively stimulates N-WASP and has no effects on WASP. Rac1 is a significantly more potent N-WASP activator than Cdc42. In contrast, Cdc42 is a more effective activator of WASP than N-WASP. Lipid vesicles containing PIP2 significantly improve actin nucleation by the Arp2/3 complex and N-WASP in the presence of Rac1 or Cdc42. PIP2 vesicles have no effect on WASP activity alone. Moreover, the inhibition of WASP-stimulated actin nucleation in the presence of Cdc42 and PIP2 vesicles has been observed. We found that adaptor proteins Nck1 or Nck2 are the most potent WASP and N-WASP activators with distinct effects on the WASP family members. Our in vitro data demonstrates differential regulation of full-length WASP and N-WASP by cellular activators that highlights fundamental differences of response at the protein-protein level.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/fisiologia , Actinas/metabolismo , Proteínas Oncogênicas/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Humanos , Proteínas Recombinantes/biossíntese , Transdução de Sinais
17.
Conf Proc IEEE Eng Med Biol Soc ; 2006: 1735-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17945663

RESUMO

To build a biomechanical human model can make much sense for surgical training and surgical rehearse. Especially, it will be more meaningful to develop a biomechanical model to guide the control strategy for the medical robots in HIT-Robot Assisted Orthopedic Surgery System (HIT-RAOS). In this paper, based the successful work of others, a novel reliable finite element method based biomechanical model for HIT-RAOS was developed to simulate the force needed in reposition procedure. Geometrical model was obtained from 3D reconstruction from CT images of a just died man. Using this boundary information, the finite element model of the leg including part of femur, broken upper tibia, broken lower tibia, talus, calcaneus, Kirschner nail, muscles and other soft tissues was created in ANSYS. Furthermore, as it was too difficult to reconstruct the accurate geometry model from CT images, a new simplified muscle model was presented. The bony structures and tendons were defined as linearly elastic, while soft tissues and muscle fibers were assumed to be hyper elastic. To validate this model, the same dead man was involved to simulate the patient, and a set of data of the force needed to separate the two broken bones and the distance between them in reposition procedure was recorded. Then, another set of data was acquired from the finite element analysis. After comparison, the two sets of data matched well. The Finite Element model was proved to be acceptable.


Assuntos
Fenômenos Biomecânicos/métodos , Fixação de Fratura/métodos , Modelos Biológicos , Procedimentos Ortopédicos/métodos , Robótica/métodos , Cirurgia Assistida por Computador/métodos , Fraturas da Tíbia/fisiopatologia , Fraturas da Tíbia/cirurgia , Fenômenos Biomecânicos/instrumentação , Análise de Elementos Finitos , Fixação de Fratura/instrumentação , Humanos , Procedimentos Ortopédicos/instrumentação , Robótica/instrumentação , Cirurgia Assistida por Computador/instrumentação
18.
Conf Proc IEEE Eng Med Biol Soc ; Suppl: 6505-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17959437

RESUMO

To build a biomechanical human model can make much sense for surgical training and surgical rehearse. Especially, it will be more meaningful to develop a biomechanical model to guide the control strategy for the medical robots in HIT-Robot Assisted Orthopedic Surgery System (HIT-RAOS). In this paper, based the successful work of others, a novel reliable finite element method based biomechanical model for HIT-RAOS was developed to simulate the force needed in reposition procedure. Geometrical model was obtained from 3D reconstruction from CT images of a just died man. Using this boundary information, the finite element model of the leg including part of femur, broken upper tibia, broken lower tibia, talus, calcaneus, Kirschner nail, muscles and other soft tissues was created in ANSYS. Furthermore, as it was too difficult to reconstruct the accurate geometry model from CT images, a new simplified muscle model was presented. The bony structures and tendons were defined as linearly elastic, while soft tissues and muscle fibers were assumed to be hyper elastic. To validate this model, the same dead man was involved to simulate the patient, and a set of data of the force needed to separate the two broken bones and the distance between them in reposition procedure was recorded. Then, another set of data was acquired from the finite element analysis. After comparison, the two sets of data matched well. The Finite Element model was proved to be acceptable.


Assuntos
Análise de Elementos Finitos , Procedimentos Ortopédicos/métodos , Robótica , Osso e Ossos/cirurgia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...