Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Physiol Rep ; 9(21): e15081, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34755490

RESUMEN

Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.


Asunto(s)
Angiopoyetina 2/antagonistas & inhibidores , Orthomyxoviridae/patogenicidad , Neumonía Viral/tratamiento farmacológico , Angiopoyetina 2/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Células Cultivadas , Citocinas/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Neumonía Viral/metabolismo , Neumonía Viral/virología , Receptor TIE-2/metabolismo , Carga Viral
2.
Circulation ; 143(2): 120-134, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33118835

RESUMEN

BACKGROUND: Heart failure (HF) with preserved ejection fraction (HFpEF) constitutes half of all HF but lacks effective therapy. Understanding of its myocardial biology remains limited because of a paucity of heart tissue molecular analysis. METHODS: We performed RNA sequencing on right ventricular septal endomyocardial biopsies prospectively obtained from patients meeting consensus criteria for HFpEF (n=41) contrasted with right ventricular septal tissue from patients with HF with reduced ejection fraction (HFrEF, n=30) and donor controls (n=24). Principal component analysis and hierarchical clustering tested for transcriptomic distinctiveness between groups, effect of comorbidities, and differential gene expression with pathway enrichment contrasted HF groups and donor controls. Within HFpEF, non-negative matrix factorization and weighted gene coexpression analysis identified molecular subgroups, and the resulting clusters were correlated with hemodynamic and clinical data. RESULTS: Patients with HFpEF were more often women (59%), African American (68%), obese (median body mass index 41), and hypertensive (98%), with clinical HF characterized by 65% New York Heart Association Class III or IV, nearly all on a loop diuretic, and 70% with a HF hospitalization in the previous year. Principal component analysis separated HFpEF from HFrEF and donor controls with minimal overlap, and this persisted after adjusting for primary comorbidities: body mass index, sex, age, diabetes, and renal function. Hierarchical clustering confirmed group separation. Nearly half the significantly altered genes in HFpEF versus donor controls (1882 up, 2593 down) changed in the same direction in HFrEF; however, 5745 genes were uniquely altered between HF groups. Compared with controls, uniquely upregulated genes in HFpEF were enriched in mitochondrial adenosine triphosphate synthesis/electron transport, pathways downregulated in HFrEF. HFpEF-specific downregulated genes engaged endoplasmic reticulum stress, autophagy, and angiogenesis. Body mass index differences largely accounted for HFpEF upregulated genes, whereas neither this nor broader comorbidity adjustment altered pathways enriched in downregulated genes. Non-negative matrix factorization identified 3 HFpEF transcriptomic subgroups with distinctive pathways and clinical correlates, including a group closest to HFrEF with higher mortality, and a mostly female group with smaller hearts and proinflammatory signaling. These groupings remained after sex adjustment. Weighted gene coexpression analysis yielded analogous gene clusters and clinical groupings. CONCLUSIONS: HFpEF exhibits distinctive broad transcriptomic signatures and molecular subgroupings with particular clinical features and outcomes. The data reveal new signaling targets to consider for precision therapeutics.


Asunto(s)
Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Volumen Sistólico/fisiología , Transcriptoma/fisiología , Anciano , Cateterismo Cardíaco/métodos , Femenino , Insuficiencia Cardíaca/patología , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , Estudios Prospectivos
3.
FEBS Open Bio ; 11(1): 207-225, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33135334

RESUMEN

Pericytes (PCs)-mural cells that envelop endothelial cells (ECs) of microvessels-regulate tissue-specific vasculature development as well as maturation and maintenance of endothelial barrier integrity. However, little is known about their tissue-specific function in the heart. Specifically, the mechanism by which cardiac PCs constrict coronary capillaries remains undetermined. To gain insights into the function of cardiac PCs at the cellular level, we isolated NG2+ PDGFRß+ CD146+ CD34- CD31- CD45- PCs for detailed characterization. Functionally, we provide evidence that these PCs increased transepithelial electrical resistance and decreased endothelial permeability. We show for the first time that this population of PCs express contractile proteins, are stimulated by adrenergic signaling, and demonstrate stereotypical contraction and relaxation. Furthermore, we also studied for the first time, the PCs in in vitro models of disease. PCs in hypoxia activated the hypoxia-inducible factor 1 alpha pathway, increased secretion of angiogenic factors, and caused cellular apoptosis. Supraphysiological levels of low-density lipoprotein decreased PC proliferation and induced lipid droplet accumulation. Elevated glucose levels triggered a proinflammatory response. Taken together, our study characterizes cardiac PCs under in vitro disease conditions and supports the hypothesis that cardiac PCs are key vasoactive cells that can regulate blood flow in the heart.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/citología , Fenómeno de no Reflujo/fisiopatología , Pericitos/fisiología , Animales , Permeabilidad Capilar , Hipoxia de la Célula/fisiología , Células Cultivadas , Circulación Coronaria/fisiología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Masculino , Ratones , Contracción Miocárdica/fisiología , Cultivo Primario de Células
4.
J Pharmacol Exp Ther ; 375(2): 296-307, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958629

RESUMEN

Infarct expansion can occur after myocardial infarction (MI), which leads to adverse left ventricular (LV) remodeling and failure. An imbalance between matrix metalloproteinase (MMP) induction and tissue inhibitors of MMPs (TIMPs) can accelerate this process. Past studies have shown different biologic effects of TIMP-3, which may depend upon specific domains within the TIMP-3 molecule. This study tested the hypothesis that differential effects of direct myocardial injections of either a full-length recombinant TIMP-3 (F-TIMP-3) or a truncated form encompassing the N-terminal region (N-TIMP-3) could be identified post-MI. MI was induced in pigs that were randomized for MI injections (30 mg) and received targeted injections within the MI region of F-TIMP-3 (n = 8), N-TIMP-3 (n = 9), or saline injection (MI-only, n = 11). At 14 days post-MI, LV ejection fraction fell post-MI but remained higher in both TIMP-3 groups. Tumor necrosis factor and interleukin-10 mRNA increased by over 10-fold in the MI-only and N-TIMP-3 groups but were reduced with F-TIMP-3 at this post-MI time point. Direct MI injection of either a full-length or truncated form of TIMP-3 is sufficient to favorably alter the course of post-MI remodeling. The functional and differential relevance of TIMP-3 domains has been established in vivo since the TIMP-3 constructs demonstrated different MMP/cytokine expression profiles. These translational studies identify a unique and more specific therapeutic strategy to alter the course of LV remodeling and dysfunction after MI. SIGNIFICANCE STATEMENT: Using different formulations of tissue inhibitor of matrix metalloproteinase-3 (TIMP-3), when injected into the myocardial infarction (MI) region, slowed the progression of indices of left ventricular (LV) failure, suggesting that the N terminus of TIMP-3 is sufficient to attenuate early adverse functional events post-MI. Injections of full-length recombinant TIMP-3, but not of the N-terminal region of TIMP-3, reduced relative indices of inflammation at the mRNA level, suggesting that the C-terminal region affects other biological pathways. These unique proof-of-concept studies demonstrate the feasibility of using recombinant small molecules to selectively interrupt adverse LV remodeling post-MI.


Asunto(s)
Infarto del Miocardio/patología , Fragmentos de Péptidos/farmacología , Inhibidor Tisular de Metaloproteinasa-3/química , Remodelación Ventricular/efectos de los fármacos , Secuencia de Aminoácidos , Colágeno/genética , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones , Metaloproteinasas de la Matriz/genética , Fragmentos de Péptidos/química , Dominios Proteicos , ARN Mensajero/genética , Inhibidor Tisular de Metaloproteinasa-3/genética
5.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32208384

RESUMEN

Heart failure (HF) remains a grievous illness with poor prognosis even with optimal care. The apelin receptor (APJ) counteracts the pressor effect of angiotensin II, attenuates ischemic injury, and has the potential to be a novel target to treat HF. Intravenous administration of apelin improves cardiac function acutely in patients with HF. However, its short half-life restricts its use to infusion therapy. To identify a longer acting APJ agonist, we conducted a medicinal chemistry campaign, leading to the discovery of potent small-molecule APJ agonists with comparable activity to apelin by mimicking the C-terminal portion of apelin-13. Acute infusion increased systolic function and reduced systemic vascular resistance in 2 rat models of impaired cardiac function. Similar results were obtained in an anesthetized but not a conscious canine HF model. Chronic oral dosing in a rat myocardial infarction model reduced myocardial collagen content and improved diastolic function to a similar extent as losartan, a RAS antagonist standard-of-care therapy, but lacked additivity with coadministration. Collectively, this work demonstrates the feasibility of developing clinical, viable, potent small-molecule agonists that mimic the endogenous APJ ligand with more favorable drug-like properties and highlights potential limitations for APJ agonism for this indication.


Asunto(s)
Receptores de Apelina/agonistas , Corazón/efectos de los fármacos , Animales , Perros , Descubrimiento de Drogas , Insuficiencia Cardíaca , Péptidos y Proteínas de Señalización Intercelular , Ratas
6.
Circ Cardiovasc Imaging ; 12(11): e009055, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31707811

RESUMEN

BACKGROUND: The induction of matrix metalloproteinases (MMPs) and reduction in tissue inhibitors of MMPs (TIMPs) plays a role in ischemia/reperfusion (I/R) injury post-myocardial infarction (MI) and subsequent left ventricular remodeling. We developed a hybrid dual isotope single-photon emission computed tomography/computed tomography approach for noninvasive evaluation of regional myocardial MMP activation with 99mTc-RP805 and dynamic 201Tl for determination of myocardial blood flow, to quantify the effects of intracoronary delivery of recombinant TIMP-3 (rTIMP-3) on I/R injury. METHODS: Studies were performed in control pigs (n=5) and pigs following 90-minute balloon occlusion-induced ischemia/reperfusion (I/R) of left anterior descending artery (n=9). Before reperfusion, pigs with I/R were randomly assigned to intracoronary infusion of rTIMP-3 (1.0 mg/kg; n=5) or saline (n=4). Three days post-I/R, dual isotope imaging was performed with 99mTc-RP805 and 201Tl along with contrast cineCT to assess left ventricular function. RESULTS: The ischemic to nonischemic ratio of 99mTc-RP805 was significantly increased following I/R in saline group (4.03±1.40), and this ratio was significantly reduced with rTIMP-3 treatment (2.22±0.57; P=0.03). This reduction in MMP activity in the MI-rTIMP-3 treatment group was associated with an improvement in relative MI region myocardial blood flow compared with the MI-saline group and improved myocardial strain in the MI region. CONCLUSIONS: We have established a novel hybrid single-photon emission computed tomography/computed tomography imaging approach for the quantitative assessment of regional MMP activation, myocardial blood flow, and cardiac function post-I/R that can be used to evaluate therapeutic interventions such as intracoronary delivery of rTIMP-3 for reduction of I/R injury in the early phases of post-MI remodeling.


Asunto(s)
Ventrículos Cardíacos , Metaloproteinasas de la Matriz , Infarto del Miocardio , Miocardio , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Masculino , Circulación Coronaria/fisiología , Modelos Animales de Enfermedad , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único/métodos , Porcinos , Función Ventricular Izquierda/fisiología , Remodelación Ventricular/fisiología
7.
J Vis Exp ; (150)2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31475977

RESUMEN

Pericytes, perivascular cells of microvessels and capillaries, are known to play a part in angiogenesis, vessel stabilization, and endothelial barrier integrity. However, their tissue-specific functions in the heart are not well understood. Moreover, there is currently no protocol utilizing readily accessible materials to isolate and purify pericytes of cardiac origin. Our protocol focuses on using the widely used mammalian model, the mouse, as our source of cells. Using the enzymatic digestion and mechanical dissociation of heart tissue, we obtained a crude cell mixture that was further purified by fluorescence activating cell sorting (FACS) by a plethora of markers. Because there is no single unequivocal marker for pericytes, we gated for cells that were CD31-CD34-CD45-CD140b+NG2+CD146+. Following purification, these primary cells were cultured and passaged multiple times without any changes in morphology and marker expression. With the ability to regularly obtain primary murine cardiac pericytes using our protocol, we hope to further understand the role of pericytes in cardiovascular physiology and their therapeutic potential.


Asunto(s)
Separación Celular/métodos , Miocardio/citología , Pericitos/fisiología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Ratones
8.
JACC Basic Transl Sci ; 4(2): 269-274, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31061928

RESUMEN

Despite the fact that cardiovascular disease (CVD) is the number 1 cause of death globally, investment in drug development and new drug approvals for CVD are precipitously declining. In contrast, the trajectory of both investment in development as well as new drug approvals for oncology have been increasing steadily over the same time frame. The factors that have spurred drug development in oncology may be applicable to new efforts to overcome barriers to drug development for CVD. Greater investment in basic research and application of expedited regulatory pathways have contributed to a lowering of development barriers in oncology. Barriers in implementation are also critical. More rapid adoption of guideline-based therapies and lower access barriers by payers have contributed to fewer implementation barriers for oncology therapeutics. There is substantially greater advocacy among patients and physicians for new oncology therapeutics, and such advocacy efforts are likely to have had a meaningful impact on lowering barriers to develop new oncology therapeutics. Broad support of patient and physician advocacy efforts directed towards CVD may help overcome existing development and implementation barriers to new drug development, thereby spurring more rapid progress in the fight to eradicate cardiovascular disease.

9.
Pharmacol Res Perspect ; 6(6): e00442, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30459952

RESUMEN

Tissue Inhibitor of Metalloproteinase 3 (TIMP3) is a secreted protein that has a great utility to inhibit elevated metalloproteinase (MMP) activity in injured tissues including infarcted cardiac tissue, inflamed vessels, and joint cartilages. An imbalance between TIMP3 and active MMP levels in the local tissue area may cause worsening of disease progression. To counter balance elevated MMP levels, exogenous administration of TIMP3 appeared to be beneficial in preclinical studies. However, the current form of WT-TIMP3 molecule has a limitation to be a therapeutic candidate due to low production yield, short plasma half-life, injection site retention, and difficulty in delivery, etc. We have engineered TIMP3 molecules by adding extra glycosylation sites or fusing with albumin, Fc, and antibody to improve pharmacokinetic properties. In general, the C-terminal fusion of TIMP3 improved expression and production in mammalian cells and extended half-lives dramatically 5-20 folds. Of note, a site-specific glycosylation at K22S/F34N resulted in a higher level of expression and better cardiac function compared to other fusion proteins in the context of left ventricle ejection fraction (LVEF) changes in a rat myocardial infarction model. It appeared that cardiac efficacy depends on a high ECM binding affinity, in which K22S/F34N and N-TIMP3 showed a higher binding to the ECM compared to other engineered molecules. In conclusion, we found that the ECM binding and sustained residence of injected TIMP3 molecules are important for cardiac tissue localization and inhibition of adverse remodeling activity.


Asunto(s)
Proteína ADAM17/antagonistas & inhibidores , Metaloproteinasas de la Matriz/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Proteína ADAM17/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos , Glicosilación , Humanos , Infusiones Intravenosas , Inyecciones Intralesiones , Masculino , Mutación , Infarto del Miocardio/etiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Inhibidor Tisular de Metaloproteinasa-3/química , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/uso terapéutico , Resultado del Tratamiento
10.
Am J Physiol Heart Circ Physiol ; 315(4): H814-H825, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29979624

RESUMEN

Although improvements in timing and approach for early reperfusion with acute coronary syndromes have occurred, myocardial injury culminating in a myocardial infarction (MI) remains a common event. Although a multifactorial process, an imbalance between the induction of proteolytic pathways, such as matrix metalloproteinases (MMPs) and endogenous tissue inhibitors of metalloproteinase (TIMPs), has been shown to contribute to this process. In the present study, a full-length TIMP-3 recombinant protein (rTIMP-3) was encapsulated in a specifically formulated hyaluronic acid (HA)-based hydrogel that contained MMP-cleavable peptide cross-links, which influenced the rate of rTIMP-3 release from the HA gel. The effects of localized delivery of this MMP-sensitive HA gel (HAMMPS) alone and containing rTIMP-3 (HAMMPS/rTIMP-3) were examined in terms of the natural history of post-MI remodeling. Pigs were randomized to one of the following three different groups: MI and saline injection (MI/saline group, 100-µl injection at nine injection sites, n = 7), MI and HAMMPS injection (MI/HAMMPS group; 100-µl injection at nine injection sites, n = 7), and MI and HAMMPS/rTIMP-3 injection (MI/HAMMPS/rTIMP-3 group; 20-µg/100-µl injection at nine injection sites, n = 7). Left ventricular (LV) echocardiography was serially performed up to 28 days post-MI. LV dilation, as measured by end-diastolic volume, and the degree of MI wall thinning were reduced by ~50% in the HAMMPS/rTIMP-3 group ( P < 0.05). Furthermore, indexes of heart failure progression post-MI, such as LV filling pressures and left atrial size, were also attenuated to the greatest degree in the HAMMPS/rTIMP-3 group. At 28 days post-MI, HAMMPS/rTIMP-3 caused a relative reduction in the transcriptional profile for myofibroblasts as well as profibrotic pathways, which was confirmed by subsequent histochemistry. In conclusion, these findings suggest that localized delivery of a MMP-sensitive biomaterial that releases a recombinant TIMP holds promise as a means to interrupt adverse post-MI remodeling. NEW & NOTEWORTHY The present study targeted a myocardial matrix proteolytic system, matrix metalloproteinases (MMPs), through the use of a recombinant tissue inhibitor of MMPs incorporated into a MMP-sensitive hydrogel, which was regionally injected using a large animal model of myocardial infarction. Left ventricular geometry and function and indexes of myocardial remodeling were improved with this approach and support the advancement of localized therapeutic strategies that specifically target the myocardial matrix.


Asunto(s)
Fármacos Cardiovasculares/administración & dosificación , Sulfato de Dextran/química , Portadores de Fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ácido Hialurónico/química , Infarto del Miocardio/tratamiento farmacológico , Inhibidor Tisular de Metaloproteinasa-3/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Fármacos Cardiovasculares/química , Preparaciones de Acción Retardada , Sulfato de Dextran/análogos & derivados , Modelos Animales de Enfermedad , Composición de Medicamentos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Perfilación de la Expresión Génica/métodos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Ácido Hialurónico/análogos & derivados , Hidrogeles , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proteínas Recombinantes/administración & dosificación , Inhibidor Tisular de Metaloproteinasa-3/química , Transcripción Genética/efectos de los fármacos , Transcriptoma
11.
Am J Physiol Heart Circ Physiol ; 313(4): H690-H699, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28754718

RESUMEN

Ischemia-reperfusion (IR) and myocardial infarction (MI) cause adverse left ventricular (LV) remodeling and heart failure and are facilitated by an imbalance in matrix metalloproteinase (MMP) activation and the endogenous tissue inhibitors of metalloproteinase (TIMPs). We have identified that myocardial injections of recombinant TIMP-3 (rTIMP-3; human full length) can interrupt post-MI remodeling. However, whether and to what degree intracoronary delivery of rTIMP-3 post-IR is feasible and effective remained to be established. Pigs (25 kg) underwent coronary catheterization and balloon occlusion of the left anterior descending coronary artery (LAD) for 90 min whereby at the final 4 min, rTIMP-3 (30 mg, n = 9) or saline was infused in the distal LAD. LV echocardiography was performed at 3-28 days post-IR, and LV ejection fraction (EF) and LV end-diastolic volume were measured. LV EF fell and LV end-diastolic volume increased from baseline (pre-IR) values (66 ± 1% and 40 ± 1 ml, respectively, means ± standard deviation) in both groups; however, the extent of LV dilation was reduced in the rTIMP-3 group by 40% at 28 days post-IR (P < 0.05) and the fall in LV EF was attenuated. Despite equivalent plasma troponin levels (14 ± 3 ng/ml), computed MI size at 28 days was reduced by over 45% in the rTIMP-3 group (P < 0.05), indicating that rTIMP-3 treatment abrogated MI expansion post-IR. Plasma NH2-terminal pro-brain natriuretic peptide levels, an index of heart failure progression, were reduced by 25% in the rTIMP-3 group compared with MI saline values (P < 0.05). Although the imbalance between MMPs and TIMPs has been recognized as a contributory factor for post-MI remodeling, therapeutic strategies targeting this imbalance have not been forthcoming. This study is the first to demonstrate that a relevant delivery approach (intracoronary) using rTIMP can alter the course of post-MI remodeling.NEW & NOTEWORTHY Myocardial ischemia and reperfusion injury remain significant causes of morbidity and mortality whereby alterations in the balance between matrix metalloproteinase and tissue inhibitor of metalloproteinase have been identified as contributory biological mechanisms. This novel translational study advances the concept of targeted delivery of recombinant proteins to modify adverse myocardial remodeling in ischemia-reperfusion injury.


Asunto(s)
Infarto del Miocardio , Daño por Reperfusión , Inhibidor Tisular de Metaloproteinasa-3/farmacología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Vasos Coronarios , Ecocardiografía , Infusiones Intraarteriales , Péptido Natriurético Encefálico/sangre , Péptido Natriurético Encefálico/efectos de los fármacos , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Volumen Sistólico/efectos de los fármacos , Porcinos , Troponina/sangre , Troponina/efectos de los fármacos
12.
Cardiovasc Drugs Ther ; 31(4): 445-458, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28735360

RESUMEN

PURPOSE: The need for novel approaches to cardiovascular drug development served as the impetus to convene an open meeting of experts from the pharmaceutical industry and academia to assess the challenges and develop solutions for drug discovery in cardiovascular disease. METHODS: The Novel Cardiovascular Therapeutics Summit first reviewed recent examples of ongoing or recently completed programs translating basic science observations to targeted drug development, highlighting successes (protein convertase sutilisin/kexin type 9 [PCSK9] and neprilysin inhibition) and targets still under evaluation (cholesteryl ester transfer protein [CETP] inhibition), with the hope of gleaning key lessons to successful drug development in the current era. Participants then reviewed the use of innovative approaches being explored to facilitate rapid and more cost-efficient evaluations of drug candidates in a short timeframe. RESULTS: We summarize observations gleaned from this summit and offer insight into future cardiovascular drug development. CONCLUSIONS: The rapid development in genetic and high-throughput drug evaluation technologies, coupled with new approaches to rapidly evaluate potential cardiovascular therapies with in vitro techniques, offer opportunities to identify new drug targets for cardiovascular disease, study new therapies with better efficiency and higher throughput in the preclinical setting, and more rapidly bring the most promising therapies to human testing. However, there must be a critical interface between industry and academia to guide the future of cardiovascular drug development. The shared interest among academic institutions and pharmaceutical companies in developing promising therapies to address unmet clinical needs for patients with cardiovascular disease underlies and guides innovation and discovery platforms that are significantly altering the landscape of cardiovascular drug development.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Diseño de Fármacos , Animales , Fármacos Cardiovasculares/farmacología , Enfermedades Cardiovasculares/fisiopatología , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica , Humanos
13.
J Biol Chem ; 292(31): 12787-12800, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28637870

RESUMEN

Stress-induced p38 mitogen-activated protein kinase (MAPK) activity is implicated in pathological remodeling in the heart. For example, constitutive p38 MAPK activation in cardiomyocytes induces pathological features, including myocyte hypertrophy, apoptosis, contractile dysfunction, and fetal gene expression. However, the physiological function of cardiomyocyte p38 MAPK activity in beneficial compensatory vascular remodeling is unclear. This report investigated the functional role and the underlying mechanisms of cardiomyocyte p38 MAPK activity in cardiac remodeling induced by chronic stress. Using both in vitro and in vivo model systems, we found that p38 MAPK activity is required for hypoxia-induced pro-angiogenic activity from cardiomyocytes and that p38 MAPK activation in cardiomyocyte is sufficient to promote paracrine signaling-mediated, pro-angiogenic activity. We further demonstrate that VEGF is a paracrine factor responsible for the p38 MAPK-mediated pro-angiogenic activity from cardiomyocytes and that p38 MAPK pathway activation is sufficient for inducing VEGF secretion from cardiomyocytes in an Sp1-dependent manner. More significantly, cardiomyocyte-specific inactivation of p38α in mouse heart impaired compensatory angiogenesis after pressure overload and promoted early onset of heart failure. In summary, p38αMAPK has a critical role in the cross-talk between cardiomyocytes and vasculature by regulating stress-induced VEGF expression and secretion in cardiomyocytes. We conclude that as part of a stress-induced signaling pathway, p38 MAPK activity significantly contributes to both pathological and compensatory remodeling in the heart.


Asunto(s)
Endotelio Vascular/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Isquemia Miocárdica/metabolismo , Revascularización Miocárdica , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Hipoxia de la Célula , Células Cultivadas , Cruzamientos Genéticos , Endotelio Vascular/citología , Endotelio Vascular/patología , Activación Enzimática , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Isquemia Miocárdica/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Interferencia de ARN , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Sus scrofa , Factor A de Crecimiento Endotelial Vascular/agonistas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/agonistas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
Sci Transl Med ; 5(187): 187ra69, 2013 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-23720580

RESUMEN

Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Vasos Coronarios/efectos de los fármacos , Corazón/efectos de los fármacos , Indoles/farmacología , Microvasos/efectos de los fármacos , Pericitos/efectos de los fármacos , Pirroles/farmacología , Animales , Vasos Coronarios/citología , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Sunitinib
15.
Hum Mol Genet ; 22(15): 3123-37, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23591991

RESUMEN

Mutations in ACTA2, encoding the smooth muscle cell (SMC)-specific isoform of α-actin (α-SMA), cause thoracic aortic aneurysms and dissections and occlusive vascular diseases, including early onset coronary artery disease and stroke. We have shown that occlusive arterial lesions in patients with heterozygous ACTA2 missense mutations show increased numbers of medial or neointimal SMCs. The contribution of SMC hyperplasia to these vascular diseases and the pathways responsible for linking disruption of α-SMA filaments to hyperplasia are unknown. Here, we show that the loss of Acta2 in mice recapitulates the SMC hyperplasia observed in ACTA2 mutant SMCs and determine the cellular pathways responsible for SMC hyperplasia. Acta2(-/-) mice showed increased neointimal formation following vascular injury in vivo, and SMCs explanted from these mice demonstrated increased proliferation and migration. Loss of α-SMA induced hyperplasia through focal adhesion (FA) rearrangement, FA kinase activation, re-localization of p53 from the nucleus to the cytoplasm and increased expression and ligand-independent activation of platelet-derived growth factor receptor beta (Pdgfr-ß). Disruption of α-SMA in wild-type SMCs also induced similar cellular changes. Imatinib mesylate inhibited Pdgfr-ß activation and Acta2(-/-) SMC proliferation in vitro and neointimal formation with vascular injury in vivo. Loss of α-SMA leads to SMC hyperplasia in vivo and in vitro through a mechanism involving FAK, p53 and Pdgfr-ß, supporting the hypothesis that SMC hyperplasia contributes to occlusive lesions in patients with ACTA2 missense mutations.


Asunto(s)
Actinas/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Actinas/genética , Animales , Movimiento Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Activación Enzimática , Hiperplasia , Ratones , Ratones Noqueados , Modelos Biológicos , Fenotipo , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo
16.
Stem Cells Dev ; 22(1): 148-57, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22734943

RESUMEN

Over the past 10 years, a great deal has been learned about the fundamental biology and therapeutic application of bone marrow-derived human mesenchymal stem cells (MSCs). Intravenous administration of these cells is the preferred route for therapeutic delivery of MSCs. Vascular endothelial cells (ECs) are the first cell type that MSCs encounter following IV administration. However, little is known about the biological consequences of interactions between MSCs and ECs, and if any therapeutic benefit results from this interaction. We show that MSCs exert potent stabilizing effects on ECs using an in vitro coculture system. Such effects include decreased EC proliferation and the reduction of EC vascular network formation in matrigel. Interestingly, these effects appear to require EC-MSC contact and result in enhanced colocalization of VE-Cadherin and ß-catenin at the cell membrane. Disruption of the VE-Cadherin/ß-catenin interaction abrogates the observed effects. As a functional in vivo correlate, we show that intravenously administered MSCs strongly inhibit angiogenesis in a matrigel plug assay. Taken together, these results identify a novel mechanism of action of MSCs that involves a contact-dependent EC interaction. These findings are relevant to intravenous use of MSCs and provide insight into further optimizing therapeutic strategies involving MSCs.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , beta Catenina/metabolismo , Uniones Adherentes/metabolismo , Animales , Comunicación Celular , Técnicas de Cocultivo , Colágeno , Combinación de Medicamentos , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Laminina , Masculino , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Proteoglicanos , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo
17.
Sci Transl Med ; 4(161): 161ra150, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175708

RESUMEN

Mesenchymal stem cells (MSCs) may be useful for treating a variety of disease states associated with vascular instability including traumatic brain injury (TBI). A soluble factor, tissue inhibitor of matrix metalloproteinase-3 (TIMP3), produced by MSCs is shown to recapitulate the beneficial effects of MSCs on endothelial function and to ameliorate the effects of a compromised blood-brain barrier (BBB) due to TBI. Intravenous administration of recombinant TIMP3 inhibited BBB permeability caused by TBI, whereas attenuation of TIMP3 expression in intravenously administered MSCs blocked the beneficial effects of the MSCs on BBB permeability and stability. MSCs increased circulating concentrations of soluble TIMP3, which blocked vascular endothelial growth factor-A-induced breakdown of endothelial cell adherens junctions in vitro and in vivo. These findings elucidate a potential molecular mechanism for the beneficial effects of MSCs on the BBB after TBI and demonstrate a role for TIMP3 in the regulation of BBB integrity.


Asunto(s)
Barrera Hematoencefálica/patología , Lesiones Encefálicas/patología , Lesiones Encefálicas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Administración Intravenosa , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/metabolismo , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Infiltración Neutrófila/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/farmacología , Bazo/efectos de los fármacos , Bazo/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/sangre
18.
Nat Rev Clin Oncol ; 9(8): 471-8, 2012 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-22751283

RESUMEN

Our understanding of the biology of cancer and the application of this knowledge to cancer treatment has greatly outpaced what we know of the biology underlying the symptoms and toxic effects that therapies produce. These adverse effects of therapy cause substantial discomfort and distress to patients and their families, limit treatment tolerability and can persist indefinitely in post-treatment survivorship. Despite these concerns, little research effort is targeted at documenting the nature of these effects. Similarly, limited efforts are being made in the drug-development arena to identify or develop treatments that might prevent or reduce toxicities. A panel of clinicians and researchers as well as representatives from advocacy groups, federal agencies and the pharmaceutical industry was convened to identify gaps in cancer treatment toxicity research and to provide direction for future action. With an emphasis on coordinating multidisciplinary efforts, this panel has presented a strategy to increase funding for the field and develop a coherent research agenda.


Asunto(s)
Antineoplásicos/efectos adversos , Neoplasias/terapia , Planificación de Atención al Paciente , Complicaciones Posoperatorias , Traumatismos por Radiación/prevención & control , Humanos
19.
Tex Heart Inst J ; 39(6): 890-3, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23304048

RESUMEN

A 41-year-old woman with metastatic melanoma was admitted to the hospital because of syncopal episodes, which had developed after the administration of an experimental chemotherapy agent that targeted Notch signaling, as part of a phase I clinical trial. Cardiac monitoring revealed recurrent episodes of polymorphic ventricular tachycardia correlating with the patient's syncope. Investigations into the cause of the arrhythmia led to the discovery of metastatic lesions within the left ventricular myocardium. In presenting this case of polymorphic ventricular tachycardia as the antemortem clinical manifestation of metastatic melanoma involving the heart, we discuss the importance of recognizing that cardiac metastases can manifest themselves as arrhythmias in patients with malignant melanoma who are undergoing active anticancer treatment.


Asunto(s)
Electrocardiografía , Neoplasias Cardíacas/complicaciones , Melanoma/complicaciones , Neoplasias Cutáneas/complicaciones , Taquicardia Ventricular/etiología , Adulto , Diagnóstico Diferencial , Femenino , Neoplasias Cardíacas/secundario , Ventrículos Cardíacos , Humanos , Imagen por Resonancia Cinemagnética , Melanoma/secundario , Neoplasias Cutáneas/patología , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/fisiopatología
20.
Tex Heart Inst J ; 38(3): 253-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21720463

RESUMEN

The convergence of novel molecular techniques for the study of cardiovascular disease­together with the development of an armamentarium of molecular-targeted anticancer therapies­is fueling the interest of molecular cardiologists in probing the mechanisms of cancer-therapy­induced cardiac toxicity. Such studies have the potential to develop strategies that will prevent cardiac toxicity from becoming a barrier to effective anticancer therapy. They might also provide novel insights into the pathogenesis of non-cancer-therapy­induced human heart disease.


Asunto(s)
Antineoplásicos/efectos adversos , Cardiopatías/inducido químicamente , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Corazón/fisiopatología , Cardiopatías/patología , Cardiopatías/fisiopatología , Cardiopatías/terapia , Humanos , Miocitos Cardíacos/patología , Pronóstico , Medición de Riesgo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...