Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Infect Dis ; 228(Suppl 6): S355-S358, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37849398

RESUMEN

Given the increased risk of pandemics driven by emerging and reemerging infectious diseases, it is imperative that the United States and global scientific community be better prepared for future threats by prioritizing and launching key research programs and strategies. In December 2021, the National Institute of Allergy and Infectious Diseases (NIAID) published its pandemic preparedness plan, which focuses on the prototype pathogen approach for medical countermeasure development. The plan was introduced before its release at a NIAID-hosted workshop in November 2021 that featured scientific experts from the extramural community, government, and the private sector and focused on selection of prototype pathogens from 10 viral families with pandemic risk and moderate resources. This article will serve as an introduction to this special issue and will briefly define the prototype pathogen approach, describe the workshop goals and process for outcomes, and provide an outline of the viral working group articles to follow and future directions for NIAID.


Asunto(s)
Enfermedades Transmisibles Emergentes , Vacunas , Estados Unidos , Humanos , National Institute of Allergy and Infectious Diseases (U.S.) , Pandemias/prevención & control
2.
J Infect Dis ; 228(Suppl 6): S460-S464, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37849396

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic demonstrated how rapidly vaccines and monoclonal antibodies (mAbs) could be deployed when the field is prepared to respond to a novel virus, serving as proof of concept that the prototype pathogen approach is feasible. This success was built upon decades of foundational research, including the characterization of protective antigens and coronavirus immunity leading to the development and validation of a generalizable vaccine approach for multiple coronaviruses. For other virus families of pandemic concern, the field is less prepared. The articles in this special issue have highlighted research gaps that need to be addressed to accelerate the development of effective vaccines and mAbs, to identify generalizable vaccine and mAb strategies, and to increase preparedness against other pandemic threats. Successful implementation of the prototype pathogen approach will require a systematic, multidisciplinary, coordinated approach with expertise and crosstalk among researchers of different virus families.


Asunto(s)
COVID-19 , Vacunas , Vacunas Virales , Humanos , Anticuerpos Antivirales , Pandemias/prevención & control , COVID-19/prevención & control , Anticuerpos Monoclonales
3.
J Infect Dis ; 227(12): 1433-1441, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35876700

RESUMEN

Severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) emerged 20 years ago, presaging a series of subsequent infectious disease epidemics of international concern. The recent emergence of SARS-CoV-2 has underscored the importance of targeted preparedness research to enable rapid countermeasure development during a crisis. In December 2021 the National Institute of Allergy and Infectious Diseases (NIAID), building upon the successful strategies developed during the SARS-CoV-2 response and to prepare for future pandemics, published a pandemic preparedness plan that outlined a research strategy focused on priority pathogens, technology platforms, and prototype pathogens. To accelerate the discovery, development, and evaluation of medical countermeasures against new or previously unknown pathogens of pandemic potential, we present here a strategy of research directed at select prototype pathogens. In this manner, leveraging a prototype pathogen approach may serve as a powerful cornerstone in biomedical research preparedness to protect public health from newly emerging and reemerging infectious diseases.


Asunto(s)
Pandemias , Vacunas , Brotes de Enfermedades , National Institute of Allergy and Infectious Diseases (U.S.) , Pandemias/prevención & control , Desarrollo de Vacunas , Enfermedades Transmisibles/epidemiología
4.
FASEB J ; 28(3): 1049-58, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24297696

RESUMEN

Physician scientists (researchers with either M.D. or M.D.-Ph.D. degrees) have the unique potential to combine clinical perspectives with scientific insight, and their participation in biomedical research has long been an important topic for policymakers and educators. Given the recent changes in the research environment, an update and extension of earlier studies of this population was needed. Our findings show that physician scientists are less likely to take a major role in biomedical research than they were in the past. The number of physician scientists receiving postdoctoral research training and career development awards is at an all-time low. Physician scientists today, on average, receive their first major research award (R01 equivalent) at a later age than in the 1980s. The number of first-time R01-equivalent awards to physicians is at the same level as it was 30 yr ago, but physicians now represent a smaller percentage of the grant recipients. The long-term decline in the number of physicians entering research careers was temporarily halted during the period of substantial U.S. National Institutes of Health (NIH) budget growth (1998-2003). These gains are lost, however, in the subsequent years when NIH budgets failed to keep pace with rising costs.


Asunto(s)
National Institutes of Health (U.S.)/economía , Médicos/economía , Apoyo a la Investigación como Asunto , Presupuestos , Historia del Siglo XXI , Estados Unidos
5.
Physiol Genomics ; 45(12): 493-8, 2013 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-23632416

RESUMEN

It is well recognized that there is sex-dimorphic expression of mRNA and protein in the heart; however, the underlying mechanism is poorly understood. Endothelial nitric oxide synthase (eNOS) is an important regulator of cardiac function, and the expression levels of eNOS differ between male and female hearts. The aim of this study was to examine whether expression of specific microRNA (miRNA, miR) in males and females contributes to changes in the expression of eNOS. miRNA was extracted from the myocardium of male and female C57BL/6 mice and subjected to an Affymetrix miRNA array. Decreased expression of miR-222 was discovered in females and confirmed by qRT-PCR from whole heart or isolated cardiomyocytes. The transcription factor V-ets erythroblastosis virus E26 oncogene homolog-1 (ets-1) was identified as a potential target of miR-222 using TargetScan, and fivefold increased ets-1 protein expression in females was confirmed by Western blot. Targeting of ets-1 by miR-222 was determined in HEK293 cells overexpressing luciferase under regulation of either the ets-1 3'-UTR, a null 3'-UTR control, or a scrambled ets-1 3'-UTR and treated with a small molecule miR-222 mimic or inhibitor. Additionally qRT-PCR confirmed that mRNA levels of the ets-1 transcriptional target, eNOS, were 25% higher in females. Compared with untreated myocyte controls, 50% inhibition of eNOS expression was achieved by treatment with a miR-222 mimic, compared with a 25% increase due to miR-222 inhibitor. Our findings indicate that sex-dependent miR-222 regulation alters the expression of the cardiac regulatory protein eNOS.


Asunto(s)
MicroARNs/metabolismo , Miocardio/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Caracteres Sexuales , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Óxido Nítrico Sintasa de Tipo III/genética , Proteína Proto-Oncogénica c-ets-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-ets-1/genética
7.
J Transl Med ; 10: 72, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22500917

RESUMEN

This report is based on the Federation of American Societies for Experimental Biology's symposium, "Engaging basic Scientists in Translational Research: Identifying Opportunities, Overcoming Obstacles," held in Chevy Chase, MD, March 24-25, 2011. Meeting participants examined the benefits of engaging basic scientists in translational research, the challenges to their participation in translational research, and the roles that research institutions, funding organizations, professional societies, and scientific publishers can play to address these challenges.


Asunto(s)
Investigadores , Investigación Biomédica Traslacional , Animales , Conducta Cooperativa , Directrices para la Planificación en Salud , Organizaciones de Planificación en Salud/economía , Humanos , Motivación , Cultura Organizacional , Investigadores/economía , Investigación Biomédica Traslacional/economía , Investigación Biomédica Traslacional/educación
8.
Am J Physiol Heart Circ Physiol ; 301(4): H1513-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21841012

RESUMEN

Methionine sulfoxide reductase A (MsrA) catalytically scavenges reactive oxygen species and also repairs oxidized methionines in proteins. Increasing MsrA protects cells and organs from a variety of oxidative stresses while decreasing MsrA enhances damage, but the mechanisms of action have not been elucidated. A single gene encodes MsrA of which ∼25% is targeted to the mitochondria, a major site of reactive oxygen species production. The other ∼75% is targeted to the cytosol and is posttranslationally modified by myristoylation. To determine the relative importance of MsrA in each compartment in protecting against ischemia-reperfusion damage, we created a series of transgenic mice overexpressing MsrA targeted to the mitochondria or the cytosol. We used a Langendorff model of ischemia-reperfusion and assayed both the rate pressure product and infarct size following ischemia and reperfusion as measures of injury. While the mitochondrially targeted MsrA was expected to be protective, it was not. Notably, the cytosolic form was protective but only if myristoylated. The nonmyristoylated, cytosolic form offered no protection against injury. We conclude that cytosolic MsrA protects the heart from ischemia-reperfusion damage. The requirement for myristoylation suggests that MsrA must interact with a hydrophobic domain to provide protection.


Asunto(s)
Metionina Sulfóxido Reductasas/fisiología , Daño por Reperfusión/prevención & control , Animales , Citosol/metabolismo , Citosol/fisiología , Femenino , Hemodinámica/fisiología , Inmunohistoquímica , Metionina Sulfóxido Reductasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/fisiología , Miocardio/metabolismo , Miocardio/patología , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/patología , Fracciones Subcelulares/metabolismo
9.
Trends Cardiovasc Med ; 20(3): 73-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21130949

RESUMEN

Premenopausal females have a comparably lower incidence of cardiovascular disease than their male counterparts. Although estrogen and activation of estrogen receptors (ERs) have been found to contribute to female protection, the complex mechanisms involved are unclear. Besides altering gene transcription, estrogen could elicit its cardioprotective effect via ER-mediated nongenomic signaling pathways. In addition to the two classic nuclear ER isoforms, ERα and ERß, a G-protein coupled ER (GPR30 or GPER) has been found to be expressed in cardiomyocytes and plays an acute cardioprotective role in ischemia reperfusion injury. By using isoform-specific ER knockout mouse models and/or their specific modulators, the mechanisms of the different ERs involved in cardioprotection have been explored. In this review, we will focus on the signaling pathways leading to cardioprotection in ischemia reperfusion injury after ER activation and discuss the possibility and promise of specific ER modulators to treat ischemic heart diseases.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Transducción de Señal , Animales , Fármacos Cardiovasculares/uso terapéutico , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Factores Sexuales , Transducción de Señal/efectos de los fármacos
10.
Am J Physiol Heart Circ Physiol ; 297(5): H1806-13, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19717735

RESUMEN

Premenopausal females have a lower incidence of cardiovascular disease than their male counterparts, but the mechanism is unclear. Estrogen has been thought to signal through two nuclear receptors: estrogen receptor-alpha or estrogen receptor-beta; however, a third, membrane-bound receptor G protein-coupled estrogen receptor (GPER), has been identified and shown to bind estrogen with high affinity. To date, there is little information on GPER in the heart and no study has looked at the effect of GPER activation during myocardial ischemia-reperfusion (I/R). Therefore, the goal of this study was to determine whether activation of GPER is cardioprotective in rats. A highly specific GPER agonist, G-1, was administered to Sprague-Dawley (200-350 g) rat hearts 10 min before 20 min of ischemic followed by 120 min of reperfusion using a Langendorff model. Similar levels of GPER were found in both male and female rat hearts. With administration of 110 nM of G-1, postischemic contractile dysfunction was significantly reduced compared with untreated controls (43.8 + or - 4.3% vs. 26.9 + or - 2.1% of preischemic rate pressure product; P < 0.05). Additionally, infarct size was reduced in the G-1-treated animals when compared with control (18.8 + or - 2.7% vs. 32.4 + or - 2.1%; P < 0.05). These observations were demonstrated in both male and intact female rat hearts. Through Western blot analysis, it was demonstrated that G-1 induces the activation of both Akt and ERK1/2. Furthermore, the protection afforded by G-1 was blocked by coadministration of a phosphatidylinositol 3-kinase (PI3K) inhibitor (wortmannin, 100 nM). Taken together, the data show that G-1 activation of GPER improves functional recovery and reduces infarct size in isolated rat hearts following I/R through a PI3K-dependent, gender-independent mechanism.


Asunto(s)
Cardiotónicos/farmacología , Ciclopentanos/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Quinolinas/farmacología , Receptores de Estrógenos/agonistas , Receptores Acoplados a Proteínas G/agonistas , Androstadienos/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas In Vitro , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Perfusión , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular/efectos de los fármacos , Wortmanina
11.
Circulation ; 118(14 Suppl): S16-23, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18824748

RESUMEN

BACKGROUND: Past studies have clearly established that matrix metalloproteinases (MMPs) contribute to adverse myocardial remodeling with ischemia and reperfusion. However, these studies measured MMP levels in extracted samples, and therefore whether and to what degree actual changes in interstitial MMP activity occur within the human myocardium in the context of ischemia/reperfusion remained unknown. METHODS AND RESULTS: The present study directly quantified MMP interstitial activity within the myocardium of patients (n=14) undergoing elective cardiac surgery during steady-state conditions, as well as during and following an obligatory period of myocardial arrest and reperfusion achieved by cardiopulmonary bypass. Interstitial MMP activity was continuously monitored using a validated MMP fluorogenic substrate, a microdialysis system placed within the myocardium, and in-line fluorescent detection system. MMP activity, as measured by fluorescent emission, reached a stable steady state level by 10 minutes after deployment of the microdialysis system. During initiation of cardiopulmonary bypass, MMP activity increased by 20% from baseline values (P<0.05), and then rapidly fell with cardiac arrest and longer periods of cardiopulmonary bypass. However, with restoration of myocardial blood flow and separation from cardiopulmonary bypass, MMP interstitial activity increased by over 30% from baseline (P<0.05). CONCLUSIONS: The present study directly demonstrated that MMP proteolytic activity exists within the human myocardial interstitium and is a dynamic process under conditions such as myocardial arrest and reperfusion.


Asunto(s)
Puente de Arteria Coronaria , Paro Cardíaco Inducido , Metaloproteinasas de la Matriz/metabolismo , Reperfusión Miocárdica , Miocardio/enzimología , Anciano , Puente Cardiopulmonar , Homeostasis , Humanos , Masculino , Metaloproteinasas de la Matriz/sangre , Microdiálisis , Persona de Mediana Edad , Reperfusión Miocárdica/métodos
12.
Am J Physiol Heart Circ Physiol ; 294(2): H875-83, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18065523

RESUMEN

The matrix metalloproteinases (MMPs), in particular, membrane type 1 MMP (MT1-MMP), are increased in the context of myocardial ischemia and reperfusion (I/R) and likely contribute to myocardial dysfunction. One potential upstream induction mechanism for MT1-MMP is endothelin (ET) release and subsequent protein kinase C (PKC) activation. Modulation of ET and PKC signaling with respect to MT1-MMP activity with I/R has yet to be explored. Accordingly, this study examined in vivo MT1-MMP activation during I/R following modification of ET signaling and PKC activation. With the use of a novel fluorogenic microdialysis system, myocardial interstitial MT1-MMP activity was measured in pigs (30 kg; n = 9) during I/R (90 min I/120 min R). Local ET(A) receptor antagonism (BQ-123, 1 microM) and PKC inhibition (chelerythrine, 1 microM) were performed in parallel microdialysis probes. MT1-MMP activity was increased during I/R by 122 +/- 10% (P < 0.05) and was unchanged from baseline with ET antagonism and/or PKC inhibition. Selective PKC isoform induction occurred such that PKC-betaII increased by 198 +/- 31% (P < 0.05). MT1-MMP phosphothreonine, a putative PKC phosphorylation site, was increased by 121 +/- 8% (P < 0.05) in the I/R region. These studies demonstrate for the first time that increased interstitial MT1-MMP activity during I/R is a result of the ET/PKC pathway and may be due to enhanced phosphorylation of MT1-MMP. These findings identify multiple potential targets for modulating a local proteolytic pathway operative during I/R.


Asunto(s)
Endotelinas/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Transducción de Señal/fisiología , Animales , Antagonistas de los Receptores de la Endotelina B , Colorantes Fluorescentes , Inmunoprecipitación , Isoenzimas/metabolismo , Microdiálisis , Daño por Reperfusión Miocárdica/enzimología , Péptidos Cíclicos/farmacología , Fosforilación , Proteína Quinasa C/metabolismo , Receptor de Endotelina B/metabolismo , Volumen Sistólico/fisiología , Porcinos , Treonina/metabolismo
13.
Ann Thorac Surg ; 82(2): 664-71, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16863782

RESUMEN

BACKGROUND: Increased myocardial interstitial levels of endothelin (ET) occur during cardioplegic arrest (CA) and may contribute to contractile dysfunction. Endothelin receptor transduction involves the protein kinase-C (PKC) family comprised of multiple isoforms with diverse functions. Which PKC isoforms may be involved in ET-induced contractile dysfunction after CA remains unknown. METHODS: Shortening velocity was measured in isolated left ventricular porcine myocytes and randomized (minimum of 30 per group): normothermia (cell culture media for 2 hours at 37 degrees C); CA (2 hours in CA solution [4 degrees C, 24 mEq K+] followed by reperfusion in cell media); ET/CA (100 pM ET incubated during CA and reperfusion). These studies were carried out in the presence and absence of PKC inhibitors (500 nM) and directed against members of the classical PKC subfamily (beta I, beta II, gamma) and the novel subfamily (epsilon, eta). RESULTS: Cardiac arrest reduced shortening velocity by approximately 50%, which was further reduced in the presence of ET. Inhibition of either the beta II or gamma PKC isoform significantly increased shortening velocity from ET/CA as well as CA only values. In separate studies (n = 3), total beta II and phosphorylated beta II increased by over 150% with ET/CA (p < 0.05). Taken together, these results suggest that a predominant intracellular effector for the negative contractile effects mediated by ET in the context of CA is the PKC isoform beta II. CONCLUSIONS: Targeted inhibition of specific PKC isoforms relieves the negative inotropic effects of ET after simulated CA. These findings provide important mechanistic support for the development of targeted inhibitory strategies with respect to ET signaling and myocyte contractile dysfunction in the context of CA and reperfusion.


Asunto(s)
Endotelinas/farmacología , Paro Cardíaco Inducido , Isoenzimas/fisiología , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/fisiología , Proteína Quinasa C/fisiología , Animales , Activación Enzimática , Reperfusión Miocárdica , Miocitos Cardíacos/enzimología , Receptores de Cinasa C Activada , Receptores de Superficie Celular/fisiología , Porcinos
14.
J Cardiovasc Pharmacol ; 47(2): 228-35, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16495760

RESUMEN

BACKGROUND: A cause-effect relationship has been established between MMP activation and left ventricular (LV) remodeling following myocardial infarction. The goal of the present study was to examine a selective MMP inhibitor (sMMPi) strategy that effectively spared MMP-1, -3, and -7 with effect to regional and global left ventricular remodeling in a pig model of myocardial infarction. METHODS AND RESULTS: Pigs instrumented with coronary snares and radiopaque markers within the area at risk were randomized to myocardial infarction-only (n = 10) or sMMPi (PGE-530742, 1 mg/kg TID) begun 3 days prior to myocardial infarction. Ten weight-matched noninstrumented pigs served as reference controls. Left ventricular end-diastolic volume in the myocardial infarction-only group was increased from baseline (81 +/- 3 mL versus 55 +/- 4 mL, respectively, P < 0.05) but was attenuated with sMMPi (67 +/- 3 mL, P < 0.05). Fractional area of shortening of marker area was decreased in the myocardial infarction-only group (change from baseline -63 +/- 10%, P < 0.05) but this effect was attenuated with sMMPi (-28 +/- 14%, P < 0.05), indicative of less dyskinesis of the infarct region with sMMPi. Wall stress was reduced within both the septal and posterior wall regions with sMMPi. Myocardial MMP-2 activity was decreased in both remote and border areas of sMMPi-treated samples compared with myocardial infarction-only values, consistent with pharmacologic MMP inhibition. CONCLUSIONS: Selective MMP inhibition favorably affected regional myocardial geometry and decreased left ventricular dilation post-myocardial infarction. This study suggests that a strategy of selective MMP inhibition of a limited array of MMPs may be an achievable goal in preventing pathologic left ventricular remodeling post-myocardial infarction.


Asunto(s)
Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/fisiología , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Inhibidores Tisulares de Metaloproteinasas/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Fluoroscopía , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Inhibidores de Proteasas/administración & dosificación , Distribución Aleatoria , Porcinos , Factores de Tiempo , Inhibidores Tisulares de Metaloproteinasas/farmacocinética , Inhibidores Tisulares de Metaloproteinasas/farmacología , Remodelación Ventricular/fisiología
15.
Cardiovasc Res ; 69(3): 666-76, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16426590

RESUMEN

The structural basis for the development of congestive heart failure (CHF) is a maladaptive myocardial remodeling process which occurs secondarily to post-myocardial infarction (MI), hypertensive hypertrophy, or cardiomyopathy. Both cellular and extracellular factors are involved in the remodeling process and it is the combined action of these factors giving rise to changes in myocardial structure which eventually affects function. One component in this remodeling process is a family of extracellular matrix degrading enzymes, the matrix metalloproteinases or MMPs. Many bioactive molecules such as cytokines/chemokines, bioactive peptides, and neurohormones which are operative in CHF likely contribute to the induction of MMPs. For example, a specific cassette of transcription factors is likely induced with extracellular stimuli in the context of CHF which in turn induces MMPs and contributes to the maladaptive remodeling process. This review will briefly discuss the biology of the MMP family, but will more importantly identify how biological factors active in CHF result in the modulation of the MMP family. Understanding how upstream molecules are involved in MMP regulation/dysregulation may provide an avenue to develop important therapeutic interventions.


Asunto(s)
Insuficiencia Cardíaca/enzimología , Metaloproteinasas de la Matriz/metabolismo , Miocardio/enzimología , Transducción de Señal , Animales , Membrana Celular/metabolismo , Citocinas/metabolismo , Activación Enzimática , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Insuficiencia Cardíaca/genética , Humanos , Metaloproteinasas de la Matriz/genética , Sistema Renina-Angiotensina/fisiología
16.
Curr Heart Fail Rep ; 2(1): 10-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16036046

RESUMEN

Myocardial remodeling invariably occurs in congestive heart failure (CHF) and is a response to a prolonged cardiovascular stress, which is characterized by a cascade of compensatory structural events. Remodeling of the myocardial interstitium occurs in CHF and likely contributes to the progression of the remodeling process. The myocardial matrix can be considered a biological highway in which a large amount of signaling proteins and structural proteins are being moved within the interstitium, entering and exiting the interstitial space, and docking to cellular components. The rates at which these events occur can accelerate and decelerate depending on the particular cardiac disease state and thereby can alter the course of myocardial remodeling. Once considered merely a scaffolding to align cells, the matrix plays a complex and divergent role in influencing cell behavior. For example, the matrix has a functional role in cell migration, proliferation, adhesion, and cell-to-cell signaling. In light of this, the myocardial matrix should not be regarded as merely a static structure, but rather, as a complex system of dynamic interactions between matrix molecules, signaling proteins, and transmembrane proteins. Specific strategies that are targeted at modifying activity along this matrix highway will likely alter the course of myocardial remodeling and heart failure.


Asunto(s)
Matriz Extracelular/fisiología , Insuficiencia Cardíaca/fisiopatología , Metaloproteinasas de la Matriz/metabolismo , Miocardio/metabolismo , Angiotensina II/fisiología , Progresión de la Enfermedad , Endotelina-1/fisiología , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Humanos , Hipertrofia Ventricular Izquierda/fisiopatología , Integrinas/fisiología , Infarto del Miocardio/metabolismo , Miocardio/química , Factor de Crecimiento Transformador beta/fisiología , Remodelación Ventricular/fisiología
17.
Circ Res ; 96(10): 1110-8, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15860759

RESUMEN

LV myocardial remodeling is a structural hallmark of hypertensive hypertrophy, but molecular mechanisms driving this process are not well understood. The matrix metalloproteinases (MMPs) can cause myocardial remodeling in chronic disease states, but how MMP activity is altered with a mechanical load remains unknown. The present study quantified interstitial MMP activity after a discrete increase in LV load and dissected out the contributory role of the angiotensin II Type 1 receptor (AT1R). Pigs (38 kg) were randomized to undergo (1) increased LV load by insertion of an intra-aortic balloon pump (IABP) triggered at systole for 3 hours, then deactivated (n=11); (2) IABP and AT1R blockade (AT1RB; valsartan, 3 ng/kg/hr; n=6). MMP activity was directly measured in the myocardial interstitium using a validated inline digital fluorogenic microdialysis system. IABP engagement increased LV peak pressure from 92+/-3 to 113+/-5 and 123+/-7 mm Hg in the vehicle and AR1RB group, respectively, and remained elevated throughout the IABP period (P<0.05). With IABP disengagement, segmental shortening (% change from baseline of 0) remained depressed in the vehicle group (-32.2+/-11.8%, P<0.05) but returned to baseline in the AT1RB group (2.3+/-12.5%). MMP activity decreased with IABP in both groups. At IABP disengagement, a surge in MMP activity occurred in the vehicle group that was abrogated with AT1RB (3.03+/-0.85 versus 0.07+/-1.55 MMP units/hr, P<0.05). A transient increase in LV load caused a cyclic variation in interstitial MMP activity that is regulated in part by the AT1R. These temporally dynamic changes in MMP activity likely influence myocardial function and structure with increased LV load.


Asunto(s)
Hipertensión/enzimología , Metaloproteinasas de la Matriz/metabolismo , Miocardio/enzimología , Receptor de Angiotensina Tipo 1/fisiología , Función Ventricular Izquierda , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Hipertensión/fisiopatología , Contrapulsador Intraaórtico , Porcinos , Remodelación Ventricular
18.
Curr Opin Cardiol ; 20(3): 211-6, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15861009

RESUMEN

PURPOSE OF REVIEW: Myocardial remodeling is a complex process involving several molecular and cellular factors. Extracellular matrix has been implicated in the remodeling process. Historically, the myocardial extracellular matrix was thought to serve solely as a means to align cells and provide structure to the tissue. Although this is one of its important functions, evidence suggests that the extracellular matrix plays a complex and divergent role in influencing cell behavior. This paper characterizes some of the notable studies on this dynamic entity and on adverse myocardial remodeling that have been published over the past year, which further question the belief that the extracellular matrix is a static structure. RECENT FINDINGS: Progress has been made in understanding how the extracellular matrix is operative in the three major conditions (myocardial infarction, left ventricular hypertrophy due to overload, and dilated cardiomyopathy) that involve myocardial remodeling. Several studies have examined plasma profiles of matrix metalloproteinases and tissue inhibitors of matrix metalloproteinases following myocardial infarction and during left ventricular hypertrophy as surrogate markers of remodeling/remodeled myocardium. It has been demonstrated that bioactive signaling molecules and growth factors, proteases, and structural proteins influence cell-matrix interactions in the context of left ventricular hypertrophy. Finally, studies that either removed or added tissue inhibitor of metalloproteinases species in the myocardium demonstrated the importance of this regulatory protein in the remodeling process. SUMMARY: Understanding the cellular and molecular triggers that in turn give rise to changes in the extracellular matrix could provide opportunities to modify the remodeling process.


Asunto(s)
Matriz Extracelular/patología , Insuficiencia Cardíaca/patología , Miocardio/patología , Remodelación Ventricular , Animales , Progresión de la Enfermedad , Matriz Extracelular/enzimología , Insuficiencia Cardíaca/enzimología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Miocardio/enzimología
19.
J Card Fail ; 11(2): 124-30, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15732032

RESUMEN

BACKGROUND: The overall goal of this study was to develop an assay procedure for measuring the relative abundance of tissue inhibitor of metalloproteinase (TIMP)-4 in plasma, and then use this approach to determine dynamic changes of TIMP-4 levels in hypertrophic obstructive cardiomyopathic (HOCM) patients after an acute myocardial infarction (MI). Matrix metalloproteinases (MMPs) contribute to tissue remodeling and are regulated by the endogenous TIMPs. TIMP-4 is observed to be expressed in higher abundance in the myocardium when compared with other types of tissues. Recent clinical studies have measured changes in TIMP-4 levels; however, these studies have been limited to measuring this protein from myocardial tissue samples. To date, no studies have monitored TIMP-4 levels in plasma samples. METHODS AND RESULTS: Plasma TIMP-4 levels were examined (by semiquantitative immunoblotting) in normal (n=18) and HOCM (n=16) patients after alcohol-induced MI. Serial measurements of plasma TIMP-4 levels were examined up to 60 hours after alcohol-induced MI in patients with HOCM. Unglycosylated plasma TIMP-4 levels increased 250% in the HOCM patients when compared with normal controls. Total plasma TIMP-4 levels decreased by 20% at 30 hours after alcohol-induced MI. CONCLUSIONS: The unique results demonstrated that an induction of a controlled MI, specifically through alcohol ablation, caused a reduction in plasma TIMP-4 levels in HOCM patients after alcohol-induced MI that would facilitate myocardial remodeling in the early post-MI setting.


Asunto(s)
Cardiomiopatía Hipertrófica/terapia , Ablación por Catéter/métodos , Etanol/administración & dosificación , Inhibidores Tisulares de Metaloproteinasas/sangre , Cardiomiopatía Hipertrófica/sangre , Estudios de Casos y Controles , Inhibidores Enzimáticos/sangre , Femenino , Tabiques Cardíacos , Humanos , Immunoblotting , Masculino , Metaloendopeptidasas/antagonistas & inhibidores , Persona de Mediana Edad , Infarto del Miocardio/inducido químicamente , Miocardio/metabolismo , Remodelación Ventricular , Inhibidor Tisular de Metaloproteinasa-4
20.
Circulation ; 111(9): 1166-74, 2005 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-15723986

RESUMEN

BACKGROUND: The matrix metalloproteinases (MMPs) contribute to regional remodeling after prolonged periods of ischemia and reperfusion (I/R), but specific MMP types activated during this process remain poorly understood. A novel class, the membrane-type MMPs (MT-MMPs), has been identified in the myocardium, but activity of these MMP types has not been assessed in vivo, particularly during I/R. METHODS AND RESULTS: Pigs (30 kg, n=8) were instrumented with microdialysis catheters to measure MT1-MMP activity in both ischemic and nonischemic (remote) myocardium. A validated MT1-MMP fluorogenic substrate was infused through the microdialysis system, and changes in fluorescence were reflective of MT1-MMP activity at steady state, during ischemia (90 minutes), and during reperfusion (120 minutes). At peak ischemia, MT1-MMP activity was increased by >40% in the ischemic region, with no change in the remote region, which persisted with reperfusion (P<0.05). After I/R, MT1-MMP abundance was increased by >50% (P<0.05). Differential centrifugation revealed that the endosomal fraction (which contains subcellular organelles) within the ischemic myocardium was associated with a >135% increase in MT1-MMP (P<0.05). Furthermore, in an isolated left ventricular myocyte model of I/R, hypoxia (simulated ischemia) induced a >70% increase in MT1-MMP abundance in myocytes, and confocal microscopy revealed MT1-MMP internalization during this time period and reemergence to the membrane with reperfusion. CONCLUSIONS: These unique results demonstrate that a specific MMP type, MT1-MMP, is increased in abundance and activity with I/R and is likely attributed, at least in part, to changes in intracellular trafficking.


Asunto(s)
Metaloendopeptidasas/metabolismo , Isquemia Miocárdica/enzimología , Daño por Reperfusión Miocárdica/enzimología , Miocitos Cardíacos/enzimología , Transporte de Proteínas , Animales , Hipoxia de la Célula , Endosomas/enzimología , Ventrículos Cardíacos , Metaloproteinasas de la Matriz Asociadas a la Membrana , Metaloendopeptidasas/análisis , Microdiálisis , Microscopía Confocal , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Miocitos Cardíacos/ultraestructura , Fracciones Subcelulares/enzimología , Sus scrofa , Inhibidor Tisular de Metaloproteinasa-1/análisis , Inhibidor Tisular de Metaloproteinasa-2/análisis , Inhibidor Tisular de Metaloproteinasa-3/análisis , Inhibidores Tisulares de Metaloproteinasas/análisis , Inhibidor Tisular de Metaloproteinasa-4
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA