Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 134(11): 1465-1482, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38655691

RESUMEN

BACKGROUND: Preclinical studies have shown the therapeutic potential of VEGF-B (vascular endothelial growth factor B) in revascularization of the ischemic myocardium, but the associated cardiac hypertrophy and adverse side effects remain a concern. To understand the importance of endothelial proliferation and migration for the beneficial versus adverse effects of VEGF-B in the heart, we explored the cardiac effects of autocrine versus paracrine VEGF-B expression in transgenic and gene-transduced mice. METHODS: We used single-cell RNA sequencing to compare cardiac endothelial gene expression in VEGF-B transgenic mouse models. Lineage tracing was used to identify the origin of a VEGF-B-induced novel endothelial cell population and adeno-associated virus-mediated gene delivery to compare the effects of VEGF-B isoforms. Cardiac function was investigated using echocardiography, magnetic resonance imaging, and micro-computed tomography. RESULTS: Unlike in physiological cardiac hypertrophy driven by a cardiomyocyte-specific VEGF-B transgene (myosin heavy chain alpha-VEGF-B), autocrine VEGF-B expression in cardiac endothelium (aP2 [adipocyte protein 2]-VEGF-B) was associated with septal defects and failure to increase perfused subendocardial capillaries postnatally. Paracrine VEGF-B led to robust proliferation and myocardial migration of a novel cardiac endothelial cell lineage (VEGF-B-induced endothelial cells) of endocardial origin, whereas autocrine VEGF-B increased proliferation of VEGF-B-induced endothelial cells but failed to promote their migration and efficient contribution to myocardial capillaries. The surviving aP2-VEGF-B offspring showed an altered ratio of secreted VEGF-B isoforms and developed massive pathological cardiac hypertrophy with a distinct cardiac vessel pattern. In the normal heart, we found a small VEGF-B-induced endothelial cell population that was only minimally expanded during myocardial infarction but not during physiological cardiac hypertrophy associated with mouse pregnancy. CONCLUSIONS: Paracrine and autocrine secretions of VEGF-B induce expansion of a specific endocardium-derived endothelial cell population with distinct angiogenic markers. However, autocrine VEGF-B signaling fails to promote VEGF-B-induced endothelial cell migration and contribution to myocardial capillaries, predisposing to septal defects and inducing a mismatch between angiogenesis and myocardial growth, which results in pathological cardiac hypertrophy.


Asunto(s)
Cardiomegalia , Linaje de la Célula , Endocardio , Células Endoteliales , Ratones Transgénicos , Factor B de Crecimiento Endotelial Vascular , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Factor B de Crecimiento Endotelial Vascular/metabolismo , Factor B de Crecimiento Endotelial Vascular/genética , Ratones , Endocardio/metabolismo , Endocardio/patología , Comunicación Paracrina , Proliferación Celular , Comunicación Autocrina , Ratones Endogámicos C57BL , Femenino , Masculino , Movimiento Celular
2.
Circulation ; 143(1): 65-77, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33203221

RESUMEN

BACKGROUND: Recent discoveries have indicated that, in the developing heart, sinus venosus and endocardium provide major sources of endothelium for coronary vessel growth that supports the expanding myocardium. Here we set out to study the origin of the coronary vessels that develop in response to vascular endothelial growth factor B (VEGF-B) in the heart and the effect of VEGF-B on recovery from myocardial infarction. METHODS: We used mice and rats expressing a VEGF-B transgene, VEGF-B-gene-deleted mice and rats, apelin-CreERT, and natriuretic peptide receptor 3-CreERT recombinase-mediated genetic cell lineage tracing and viral vector-mediated VEGF-B gene transfer in adult mice. Left anterior descending coronary vessel ligation was performed, and 5-ethynyl-2'-deoxyuridine-mediated proliferating cell cycle labeling; flow cytometry; histological, immunohistochemical, and biochemical methods; single-cell RNA sequencing and subsequent bioinformatic analysis; microcomputed tomography; and fluorescent- and tracer-mediated vascular perfusion imaging analyses were used to study the development and function of the VEGF-B-induced vessels in the heart. RESULTS: We show that cardiomyocyte overexpression of VEGF-B in mice and rats during development promotes the growth of novel vessels that originate directly from the cardiac ventricles and maintain connection with the coronary vessels in subendocardial myocardium. In adult mice, endothelial proliferation induced by VEGF-B gene transfer was located predominantly in the subendocardial coronary vessels. Furthermore, VEGF-B gene transduction before or concomitantly with ligation of the left anterior descending coronary artery promoted endocardium-derived vessel development into the myocardium and improved cardiac tissue remodeling and cardiac function. CONCLUSIONS: The myocardial VEGF-B transgene promotes the formation of endocardium-derived coronary vessels during development, endothelial proliferation in subendocardial myocardium in adult mice, and structural and functional rescue of cardiac tissue after myocardial infarction. VEGF-B could provide a new therapeutic strategy for cardiac neovascularization after coronary occlusion to rescue the most vulnerable myocardial tissue.


Asunto(s)
Vasos Coronarios/metabolismo , Endocardio/metabolismo , Infarto del Miocardio/metabolismo , Regeneración/fisiología , Factor B de Crecimiento Endotelial Vascular/biosíntesis , Animales , Transdiferenciación Celular/fisiología , Células Cultivadas , Vasos Coronarios/patología , Endocardio/patología , Ratones , Ratones Transgénicos , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Transgénicas , Factor B de Crecimiento Endotelial Vascular/deficiencia , Factor B de Crecimiento Endotelial Vascular/genética
3.
Int J Mol Sci ; 23(9)2022 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-35563050

RESUMEN

The left atrial appendage (LAA) of the adult heart has been shown to contain cardiac and myeloid progenitor cells. The resident myeloid progenitor population expresses an array of pro-regenerative paracrine factors. Cardiac constructs have been shown to inhibit deleterious remodeling of the heart using physical support. Due to these aspects, LAA holds promise as a regenerative transplant. LAAs from adult mT/mG mice were transplanted to the recipient 129X1-SvJ mice simultaneously as myocardial infarction (MI) was performed. A decellularized LAA patch was implanted in the control group. Two weeks after MI, the LAA patch had integrated to the ventricular wall, and migrated cells were seen in the MI area. The cells had two main phenotypes: small F4/80+ cells and large troponin C+ cells. After follow-up at 8 weeks, the LAA patch remained viable, and the functional status of the heart improved. Cardiac echo demonstrated that, after 6 weeks, the mice in the LAA-patch-treated group showed an increasing and statistically significant improvement in cardiac performance when compared to the MI and MI + decellularized patch controls. Physical patch-support (LAA and decellularized LAA patch) had an equal effect on the inhibition of deleterious remodeling, but only the LAA patch inhibited the hypertrophic response. Our study demonstrates that the LAA transplantation has the potential for use as a treatment for myocardial infarction. This method can putatively combine cell therapy (regenerative effect) and physical support (inhibition of deleterious remodeling).


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Infarto del Miocardio , Animales , Ecocardiografía , Ventrículos Cardíacos , Ratones , Infarto del Miocardio/terapia
4.
Proc Natl Acad Sci U S A ; 115(28): E6467-E6476, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29941602

RESUMEN

Loss of endothelial integrity promotes capillary leakage in numerous diseases, including sepsis, but there are no effective therapies for preserving endothelial barrier function. Angiopoietin-2 (ANGPT2) is a context-dependent regulator of vascular leakage that signals via both endothelial TEK receptor tyrosine kinase (TIE2) and integrins. Here, we show that antibodies against ß1-integrin decrease LPS-induced vascular leakage in murine endotoxemia, as either a preventative or an intervention therapy. ß1-integrin inhibiting antibodies bound to the vascular endothelium in vivo improved the integrity of endothelial cell-cell junctions and protected mice from endotoxemia-associated cardiac failure, without affecting endothelial inflammation, serum proinflammatory cytokine levels, or TIE receptor signaling. Moreover, conditional deletion of a single allele of endothelial ß1-integrin protected mice from LPS-induced vascular leakage. In endothelial monolayers, the inflammatory agents thrombin, lipopolysaccharide (LPS), and IL-1ß decreased junctional vascular endothelial (VE)-cadherin and induced actin stress fibers via ß1- and α5-integrins and ANGPT2. Additionally, ß1-integrin inhibiting antibodies prevented inflammation-induced endothelial cell contractility and monolayer permeability. Mechanistically, the inflammatory agents stimulated ANGPT2-dependent translocation of α5ß1-integrin into tensin-1-positive fibrillar adhesions, which destabilized the endothelial monolayer. Thus, ß1-integrin promotes endothelial barrier disruption during inflammation, and targeting ß1-integrin signaling could serve as a novel means of blocking pathological vascular leak.


Asunto(s)
Células Endoteliales/metabolismo , Endotoxemia/metabolismo , Integrina beta1/metabolismo , Uniones Intercelulares/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Endotoxemia/inducido químicamente , Endotoxemia/genética , Endotoxemia/patología , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrina beta1/genética , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
5.
Int J Mol Sci ; 22(12)2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-34205699

RESUMEN

Epitranscriptomic modifications in RNA can dramatically alter the way our genetic code is deciphered. Cells utilize these modifications not only to maintain physiological processes, but also to respond to extracellular cues and various stressors. Most often, adenosine residues in RNA are targeted, and result in modifications including methylation and deamination. Such modified residues as N-6-methyl-adenosine (m6A) and inosine, respectively, have been associated with cardiovascular diseases, and contribute to disease pathologies. The Ischemic Heart Disease Epitranscriptomics and Biomarkers (IHD-EPITRAN) study aims to provide a more comprehensive understanding to their nature and role in cardiovascular pathology. The study hypothesis is that pathological features of IHD are mirrored in the blood epitranscriptome. The IHD-EPITRAN study focuses on m6A and A-to-I modifications of RNA. Patients are recruited from four cohorts: (I) patients with IHD and myocardial infarction undergoing urgent revascularization; (II) patients with stable IHD undergoing coronary artery bypass grafting; (III) controls without coronary obstructions undergoing valve replacement due to aortic stenosis and (IV) controls with healthy coronaries verified by computed tomography. The abundance and distribution of m6A and A-to-I modifications in blood RNA are charted by quantitative and qualitative methods. Selected other modified nucleosides as well as IHD candidate protein and metabolic biomarkers are measured for reference. The results of the IHD-EPITRAN study can be expected to enable identification of epitranscriptomic IHD biomarker candidates and potential drug targets.


Asunto(s)
Epigénesis Genética , Epigenómica/métodos , Isquemia Miocárdica/metabolismo , ARN/metabolismo , Transcriptoma , Biomarcadores , Estudios de Casos y Controles , Humanos , Proyectos de Investigación
6.
Proc Natl Acad Sci U S A ; 113(46): 13144-13149, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799559

RESUMEN

Congestive heart failure is one of the leading causes of disability in long-term survivors of cancer. The anthracycline antibiotic doxorubicin (DOX) is used to treat a variety of cancers, but its utility is limited by its cumulative cardiotoxicity. As advances in cancer treatment have decreased cancer mortality, DOX-induced cardiomyopathy has become an increasing problem. However, the current means to alleviate the cardiotoxicity of DOX are limited. We considered that vascular endothelial growth factor-B (VEGF-B), which promotes coronary arteriogenesis, physiological cardiac hypertrophy, and ischemia resistance, could be an interesting candidate for prevention of DOX-induced cardiotoxicity and congestive heart failure. To study this, we administered an adeno-associated viral vector expressing VEGF-B or control vector to normal and tumor-bearing mice 1 wk before DOX treatment, using doses mimicking the concentrations used in the clinics. VEGF-B treatment completely inhibited the DOX-induced cardiac atrophy and whole-body wasting. VEGF-B also prevented capillary rarefaction in the heart and improved endothelial function in DOX-treated mice. VEGF-B also protected cultured endothelial cells from apoptosis and restored their tube formation. VEGF-B increased left ventricular volume without compromising cardiac function, reduced the expression of genes associated with pathological remodeling, and improved cardiac mitochondrial respiration. Importantly, VEGF-B did not affect serum or tissue concentrations of DOX or augment tumor growth. By inhibiting DOX-induced endothelial damage, VEGF-B could provide a novel therapeutic possibility for the prevention of chemotherapy-associated cardiotoxicity in cancer patients.


Asunto(s)
Cardiotoxicidad/terapia , Terapia Genética , Factor B de Crecimiento Endotelial Vascular/genética , Tejido Adiposo Blanco/metabolismo , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/sangre , Antibióticos Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Cardiotoxicidad/patología , Cardiotoxicidad/fisiopatología , Línea Celular Tumoral , Daño del ADN , Doxorrubicina/efectos adversos , Doxorrubicina/sangre , Doxorrubicina/farmacocinética , Células Endoteliales/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Factor B de Crecimiento Endotelial Vascular/sangre , Factor B de Crecimiento Endotelial Vascular/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(42): 13063-8, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26430242

RESUMEN

Cardiac hypertrophy accompanies many forms of heart disease, including ischemic disease, hypertension, heart failure, and valvular disease, and it is a strong predictor of increased cardiovascular morbidity and mortality. Deletion of bone marrow kinase in chromosome X (Bmx), an arterial nonreceptor tyrosine kinase, has been shown to inhibit cardiac hypertrophy in mice. This finding raised the possibility of therapeutic use of Bmx tyrosine kinase inhibitors, which we have addressed here by analyzing cardiac hypertrophy in gene-targeted mice deficient in Bmx tyrosine kinase activity. We found that angiotensin II (Ang II)-induced cardiac hypertrophy is significantly reduced in mice deficient in Bmx and in mice with inactivated Bmx tyrosine kinase compared with WT mice. Genome-wide transcriptomic profiling showed that Bmx inactivation suppresses myocardial expression of genes related to Ang II-induced inflammatory and extracellular matrix responses whereas expression of RNAs encoding mitochondrial proteins after Ang II administration was maintained in Bmx-inactivated hearts. Very little or no Bmx mRNA was expressed in human cardiomyocytes whereas human cardiac endothelial cells expressed abundant amounts. Ang II stimulation of endothelial cells increased Bmx phosphorylation, and Bmx gene silencing inhibited downstream STAT3 signaling, which has been implicated in cardiac hypertrophy. Furthermore, activation of the mechanistic target of rapamycin complex 1 pathway by Ang II treatment was decreased in the Bmx-deficient hearts. Our results demonstrate that inhibition of the cross-talk between endothelial cells and cardiomyocytes by Bmx inactivation suppresses Ang II-induced signals for cardiac hypertrophy. These results suggest that the endothelial Bmx tyrosine kinase could provide a target to attenuate the development of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/enzimología , Endotelio Vascular/enzimología , Proteínas Tirosina Quinasas/metabolismo , Angiotensina II/farmacología , Animales , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/enzimología , Transducción de Señal
8.
Clin Sci (Lond) ; 128(11): 735-47, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25534858

RESUMEN

Hypertension and persistent activation of the renin-angiotensin system (RAS) are predisposing factors for the development of acute kidney injury (AKI). Although bone-marrow-derived stromal cells (BMSCs) have shown therapeutic promise in treatment of AKI, the impact of pathological RAS on BMSC functionality has remained unresolved. RAS and its local components in the bone marrow are involved in several key steps of cell maturation processes. This may also render the BMSC population vulnerable to alterations even in the early phases of RAS pathology. We isolated transgenic BMSCs (TG-BMSCs) from young end-organ-disease-free rats with increased RAS activation [human angiotensinogen/renin double transgenic rats (dTGRs)] that eventually develop hypertension and die of end-organ damage and kidney failure at 8 weeks of age. Control cells (SD-BMSCs) were isolated from wild-type Sprague-Dawley rats. Cell phenotype, mitochondrial reactive oxygen species (ROS) production and respiration were assessed, and gene expression profiling was carried out using microarrays. Cells' therapeutic efficacy was evaluated in a rat model of acute ischaemia/reperfusion-induced AKI. Serum urea and creatinine were measured at 24 h and 48 h. Acute tubular damage was scored and immunohistochemistry was used for evaluation for markers of inflammation [monocyte chemoattractant protein (MCP-1), ED-1], and kidney injury [kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL)]. TG-BMSCs showed distinct mitochondrial morphology, decreased cell respiration and increased production of ROS. Gene expression profiling revealed a pronounced pro-inflammatory phenotype. In contrast with the therapeutic effect of SD-BMSCs, administration of TG-BMSCs in the AKI model resulted in exacerbation of kidney injury and high mortality. Our results demonstrate that early persistent RAS activation can dramatically compromise therapeutic potential of BMSCs by causing a shift into a pro-inflammatory phenotype with mitochondrial dysfunction.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Sistema Renina-Angiotensina/fisiología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/genética , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Perfilación de la Expresión Génica , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Inmunohistoquímica , Lipocalina 2 , Lipocalinas/genética , Lipocalinas/metabolismo , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Consumo de Oxígeno , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas Sprague-Dawley , Ratas Transgénicas , Especies Reactivas de Oxígeno/metabolismo , Insuficiencia Renal/genética , Insuficiencia Renal/fisiopatología , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/genética
9.
Acta Neuropathol Commun ; 12(1): 10, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38229173

RESUMEN

Mesencephalic astrocyte-derived neurotrophic factor (MANF) has cytoprotective effects on various injuries, including cerebral ischemia, and it can promote recovery even when delivered intracranially several days after ischemic stroke. In the uninjured rodent brain, MANF protein is expressed almost exclusively in neurons, but post-ischemic MANF expression has not been characterized. We aimed to investigate how endogenous cerebral MANF protein expression evolves in infarcted human brains and rodent ischemic stroke models. During infarct progression, the cerebral MANF expression pattern both in human and rat brains shifted drastically from neurons to expression in inflammatory cells. Intense MANF immunoreactivity took place in phagocytic microglia/macrophages in the ischemic territory, peaking at two weeks post-stroke in human and one-week post-stroke in rat ischemic cortex. Using double immunofluorescence and mice lacking MANF gene and protein from neuronal stem cells, neurons, astrocytes, and oligodendrocytes, we verified that MANF expression was induced in microglia/macrophage cells in the ischemic hemisphere. Embarking on the drastic expression transition towards inflammatory cells and the impact of blood-borne inflammation in stroke, we hypothesized that exogenously delivered MANF protein can modulate tissue recovery processes. In an attempt to enhance recovery, we designed a set of proof-of-concept studies using systemic delivery of recombinant MANF in a rat model of cortical ischemic stroke. Intranasal recombinant MANF treatment decreased infarct volume and reduced the severity of neurological deficits. Intravenous recombinant MANF treatment decreased the levels of pro-inflammatory cytokines and increased the levels of anti-inflammatory cytokine IL-10 in the infarcted cortex one-day post-stroke. In conclusion, MANF protein expression is induced in activated microglia/macrophage cells in infarcted human and rodent brains, and this could implicate MANF's involvement in the regulation of post-stroke inflammation in patients and experimental animals. Moreover, systemic delivery of recombinant MANF shows promising immunomodulatory effects and therapeutic potential in experimental ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Ratas , Ratones , Animales , Accidente Cerebrovascular Isquémico/metabolismo , Ratas Sprague-Dawley , Encéfalo/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/uso terapéutico , Accidente Cerebrovascular/metabolismo , Infarto Cerebral/metabolismo , Inflamación/metabolismo
11.
Circulation ; 122(17): 1725-33, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20937974

RESUMEN

BACKGROUND: Vascular endothelial growth factor-B (VEGF-B) binds to VEGF receptor-1 and neuropilin-1 and is abundantly expressed in the heart, skeletal muscle, and brown fat. The biological function of VEGF-B is incompletely understood. METHODS AND RESULTS: Unlike placenta growth factor, which binds to the same receptors, adeno-associated viral delivery of VEGF-B to mouse skeletal or heart muscle induced very little angiogenesis, vascular permeability, or inflammation. As previously reported for the VEGF-B(167) isoform, transgenic mice and rats expressing both isoforms of VEGF-B in the myocardium developed cardiac hypertrophy yet maintained systolic function. Deletion of the VEGF receptor-1 tyrosine kinase domain or the arterial endothelial Bmx tyrosine kinase inhibited hypertrophy, whereas loss of VEGF-B interaction with neuropilin-1 had no effect. Surprisingly, in rats, the heart-specific VEGF-B transgene induced impressive growth of the epicardial coronary vessels and their branches, with large arteries also seen deep inside the subendocardial myocardium. However, VEGF-B, unlike other VEGF family members, did not induce significant capillary angiogenesis, increased permeability, or inflammatory cell recruitment. CONCLUSIONS: VEGF-B appears to be a coronary growth factor in rats but not in mice. The signals for the VEGF-B-induced cardiac hypertrophy are mediated at least in part via the endothelium. Because cardiomyocyte damage in myocardial ischemia begins in the subendocardial myocardium, the VEGF-B-induced increased arterial supply to this area could have therapeutic potential in ischemic heart disease.


Asunto(s)
Permeabilidad Capilar/fisiología , Vasos Coronarios/crecimiento & desarrollo , Inflamación/fisiopatología , Neovascularización Fisiológica/fisiología , Factor B de Crecimiento Endotelial Vascular/fisiología , Adenoviridae/genética , Animales , Cardiomegalia/fisiopatología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Modelos Animales , Músculo Esquelético/irrigación sanguínea , Miocardio , Neuropilina-1/fisiología , Ratas , Ratas Transgénicas , Ratas Wistar , Factor B de Crecimiento Endotelial Vascular/genética
12.
Microb Pathog ; 51(3): 217-24, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21421042

RESUMEN

BACKGROUND: Pathogens such as Aggregatibacter actinomycetemcomitans (Aa) and Chlamydia pneumoniae (Cpn) associate with an increased risk for cardiovascular diseases by inducing inflammation. We hypothesized that the pathogens affect the vascular wall by disturbing cholesterol homeostasis and endothelial function. METHODS: Aa- and Cpn-infections were induced in apoE-deficient mice by intravenous and intranasal applications, respectively. Cholesterol efflux from mouse peritoneal macrophages to apo(lipoprotein)A-I was assessed. The efflux capacity of mouse sera as acceptors of cholesterol from RAW264.7-macrophages was determined. Additionally, endothelial function was studied by following the relaxation capacity of rat mesenteric arteries after incubation in the conditioned culture media of the peritoneal macrophages isolated from the mice. RESULTS: Infection increased serum phospholipid transfer protein (PLTP) and lipopolysaccharide (LPS) activity, as well as serum amyloid A (SAA) and TNF-α concentrations. Peritoneal macrophages of mice with Aa-infection showed increased cholesterol uptake and reduced cholesterol efflux. Sera of Cpn and Cpn + Aa-infected mice had reduced cholesterol efflux capacity from RAW264.7-macrophages. Conditioned macrophage medium from mice with chronic C. pneumoniae infection induced endothelial dysfunction. Additionally, concentrations of serum adhesion molecules, intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM) in Cpn-groups and E-selectin in Cpn + Aa-group, were elevated. The serum markers of endothelial function correlated positively with SAA. CONCLUSIONS: Aa- and Cpn-infections may generate proatherogenic changes in the vascular wall by affecting the macrophage cholesterol homeostasis and endothelial function.


Asunto(s)
Apolipoproteínas E/deficiencia , Chlamydophila pneumoniae/patogenicidad , Colesterol/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Pasteurellaceae/patogenicidad , Animales , Infecciones por Chlamydophila/microbiología , Infecciones por Chlamydophila/patología , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Homeostasis , Lipopolisacáridos/sangre , Masculino , Ratones , Ratones Noqueados , Infecciones por Pasteurellaceae/microbiología , Infecciones por Pasteurellaceae/patología , Proteínas de Transferencia de Fosfolípidos/sangre , Ratas , Suero/química , Proteína Amiloide A Sérica/análisis , Factor de Necrosis Tumoral alfa/sangre
13.
Elife ; 102021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33661096

RESUMEN

Aging, obesity, hypertension, and physical inactivity are major risk factors for endothelial dysfunction and cardiovascular disease (CVD). We applied fluorescence-activated cell sorting (FACS), RNA sequencing, and bioinformatic methods to investigate the common effects of CVD risk factors in mouse cardiac endothelial cells (ECs). Aging, obesity, and pressure overload all upregulated pathways related to TGF-ß signaling and mesenchymal gene expression, inflammation, vascular permeability, oxidative stress, collagen synthesis, and cellular senescence, whereas exercise training attenuated most of the same pathways. We identified collagen chaperone Serpinh1 (also called as Hsp47) to be significantly increased by aging and obesity and repressed by exercise training. Mechanistic studies demonstrated that increased SERPINH1 in human ECs induced mesenchymal properties, while its silencing inhibited collagen deposition. Our data demonstrate that CVD risk factors significantly remodel the transcriptomic landscape of cardiac ECs inducing inflammatory, senescence, and mesenchymal features. SERPINH1 was identified as a potential therapeutic target in ECs.


Cardiovascular diseases are the number one cause of death in the western world. Endothelial cells that line the blood vessels of the heart play a central role in the development of these diseases. In addition to helping transport blood, these cells support the normal running of the heart, and help it to grow and regenerate. Over time as the body ages and experiences stress, endothelial cells start to deteriorate. This can cause the cells to undergo senescence and stop dividing, and lay down scar-like tissue via a process called fibrosis. As a result, the blood vessels start to stiffen and become less susceptible to repair. Ageing, obesity, high blood pressure, and inactivity all increase the risk of developing cardiovascular diseases, whereas regular exercise has a protective effect. But it was unclear how these different factors affect endothelial cells. To investigate this, Hemanthakumar et al. compared the gene activity of different sets of mice: old vs young, obese vs lean, heart problems vs healthy, and fit vs sedentary. All these risk factors ­ age, weight, inactivity and heart defects ­ caused the mice's endothelial cells to activate mechanisms that lead to stress, senescence and fibrosis. Whereas exercise training had the opposite effect, and turned off the same genes and pathways. All of the at-risk groups also had high levels of a gene called SerpinH1, which helps produce tissue fiber and collagen. Experiments increasing the levels of SerpinH1 in human endothelial cells grown in the laboratory recreated the effects seen in mice, and switched on markers of stress, senescence and fibrosis. According to the World Health Organization, cardiovascular disease now accounts for 10% of the disease burden worldwide. Revealing the affects it has on gene activity could help identify new targets for drug development, such as SerpinH1. Understanding the molecular effects of exercise on blood vessels could also aid in the design of treatments that mimic exercise. This could help people who are unable to follow training programs to reduce their risk of cardiovascular disease.


Asunto(s)
Senescencia Celular , Células Endoteliales/fisiología , Proteínas del Choque Térmico HSP47/genética , Corazón/fisiopatología , Mesodermo/fisiología , Animales , Enfermedades Cardiovasculares/fisiopatología , Modelos Animales de Enfermedad , Femenino , Proteínas del Choque Térmico HSP47/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Riesgo
14.
Front Cell Dev Biol ; 9: 715093, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422835

RESUMEN

Cell-cell interactions are crucial for organ development and function. In the heart, endothelial cells engage in bidirectional communication with cardiomyocytes regulating cardiac development and growth. We aimed to elucidate the organotypic development of cardiac endothelial cells and cardiomyocyte and endothelial cell crosstalk using human induced pluripotent stem cells (hiPSC). Single-cell RNA sequencing was performed with hiPSC-derived cardiomyocytes (hiPS-CMs) and endothelial cells (hiPS-ECs) in mono- and co-culture. The presence of hiPS-CMs led to increased expression of transcripts related to vascular development and maturation, cardiac development, as well as cardiac endothelial cell and endocardium-specific genes in hiPS-ECs. Interestingly, co-culture induced the expression of cardiomyocyte myofibrillar genes and MYL7 and MYL4 protein expression was detected in hiPS-ECs. Major regulators of BMP- and Notch-signaling pathways were induced in both cell types in co-culture. These results reflect the findings from animal studies and extend them to human endothelial cells, demonstrating the importance of EC-CM interactions during development.

15.
Cardiovasc Diabetol ; 9: 5, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20105289

RESUMEN

BACKGROUND: Diabetes is associated with changes in myocardial stress-response pathways and is recognized as an independent risk factor for cardiac remodeling. Using spontaneously diabetic Goto Kakizaki rats as a model of type 2 DM we investigated whether post-translational modifications in the Akt - FOXO3a pathway, Sirt1 - p53 pathway and the mitogen activated protein kinase p38 regulator are involved in post-infarct cardiac remodeling METHODS: Experimental myocardial infarction (MI) was induced by left anterior descending coronary artery ligation in spontaneously diabetic Goto-Kakizaki rats and non-diabetic Wistar controls. Cardiac function was studied by echocardiography. Myocardial hypertrophy, cardiomyocyte apoptosis and cardiac fibrosis were determined histologically 12 weeks post MI or Sham operation. Western blotting was used to study Caspase-3, Bax, Sirt1, acetylation of p53 and phosphorylation of p38, Akt and FOXO3a. Electrophoretic mobility shift assay was used to assess FOXO3a activity and its nuclear localization. RESULTS: Post-infarct heart failure in diabetic GK rats was associated with pronounced cardiomyocyte hypertrophy, increased interstitial fibrosis and sustained cardiomyocyte apoptosis as compared with their non-diabetic Wistar controls. In the GK rat myocardium, Akt- and FOXO3a-phosphorylation was decreased and nuclear localization of FOXO3a was increased concomitantly with increased PTEN protein expression. Furthermore, increased Sirt1 protein expression was associated with decreased p53 acetylation, and phosphorylation of p38 was increased in diabetic rats with MI. CONCLUSIONS: Post-infarct heart failure in diabetic GK rats was associated with more pronounced cardiac hypertrophy, interstitial fibrosis and sustained cardiomyocyte apoptosis as compared to their non-diabetic controls. The present study suggests important roles for Akt-FOXO3a, Sirt1 - p53 and p38 MAPK in the regulation of post-infarct cardiac remodeling in type 2 diabetes.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Factores de Transcripción Forkhead/metabolismo , Infarto del Miocardio/patología , Sirtuina 1/metabolismo , Animales , Colágeno/metabolismo , Complicaciones de la Diabetes/diagnóstico por imagen , Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/fisiopatología , Diástole/fisiología , Ecocardiografía , Proteína Forkhead Box O3 , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Procesamiento Proteico-Postraduccional , Ratas , Ratas Endogámicas , Ratas Wistar , Sirtuina 1/genética , Volumen Sistólico , Sístole/fisiología
16.
Circ Res ; 103(9): 1018-26, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18757827

RESUMEN

Vascular endothelial growth factor (VEGF)-B is poorly angiogenic but prominently expressed in metabolically highly active tissues, including the heart. We produced mice expressing a cardiac-specific VEGF-B transgene via the alpha-myosin heavy chain promoter. Surprisingly, the hearts of the VEGF-B transgenic mice showed concentric cardiac hypertrophy without significant changes in heart function. The cardiac hypertrophy was attributable to an increased size of the cardiomyocytes. Blood capillary size was increased, whereas the number of blood vessels per cell nucleus remained unchanged. Despite the cardiac hypertrophy, the transgenic mice had lower heart rate and blood pressure than their littermates, and they responded similarly to angiotensin II-induced hypertension, confirming that the hypertrophy does not compromise heart function. Interestingly, the isolated transgenic hearts had less cardiomyocyte damage after ischemia. Significantly increased ceramide and decreased triglyceride levels were found in the transgenic hearts. This was associated with structural changes and eventual lysis of mitochondria, resulting in accumulation of intracellular vacuoles in cardiomyocytes and increased death of the transgenic mice, apparently because of mitochondrial lipotoxicity in the heart. These results suggest that VEGF-B regulates lipid metabolism, an unexpected function for an angiogenic growth factor.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomiopatías/metabolismo , Metabolismo de los Lípidos , Miocardio/metabolismo , Factor B de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda , Angiotensina II , Animales , Presión Sanguínea , Capilares/metabolismo , Capilares/patología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Tamaño de la Célula , Ceramidas/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Humanos , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/fisiopatología , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , Neovascularización Fisiológica , Oxidación-Reducción , Regiones Promotoras Genéticas , Piel/irrigación sanguínea , Piel/metabolismo , Factores de Tiempo , Triglicéridos/metabolismo , Regulación hacia Arriba , Factor B de Crecimiento Endotelial Vascular/genética , Miosinas Ventriculares/genética
17.
Circ J ; 74(7): 1479-87, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20526041

RESUMEN

BACKGROUND: Biological substances derived from perivascular fat modulate vascular tone, thus alterations in periadventitial adipose tissue (PVAT) may aggravate endothelial dysfunction in obesity. METHODS AND RESULTS: Male C57Bl/6 mice were fed either a high-fat diet or standard laboratory chow for 8 months. Vascular responses were studied in organ bath chambers from abdominal aortic ring preparations in the absence or presence of PVAT. The amount of PVAT as well as the cross-sectional area of adipocytes were increased in obese mice. In the presence of PVAT, obese aortas displayed impaired endothelium-dependent vasodilation whereas endothelium-independent vasodilatation was unaltered. Endothelium-dependent vasodilatation was restored after removal of PVAT and after reducing superoxide and hydrogen peroxide formation in the vascular wall by Tiron or polyethylene-glycol-catalase, respectively. PVAT from obese mice showed increased formation of hydrogen peroxide and superoxide. The PVAT-derived oxidative stress was abolished by pretreatment with the reduced nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase inhibitor, apocynin. The anti-contractile function of PVAT found in lean mice was completely abolished in obese mice, but partially restored after pretreatment with Tiron. The mRNA expressions of monocyte chemotactic protein-1, leptin and NADPH oxidase were markedly higher in the PVAT of obese than lean mice. CONCLUSIONS: PVAT promotes endothelial dysfunction in diet-induced obese C57Bl/6 mice via mechanisms that are linked to increased NADPH oxidase-derived oxidative stress and increased production of pro-inflammatory cytokines.


Asunto(s)
Tejido Adiposo/fisiopatología , Endotelio Vascular/fisiopatología , Obesidad/fisiopatología , Estrés Oxidativo , Adipocitos/patología , Animales , Aorta/patología , Citocinas/biosíntesis , Dieta/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas , Obesidad/metabolismo , Vasodilatación
18.
Heart Vessels ; 25(3): 254-62, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20512454

RESUMEN

Obesity is frequently associated with endothelial dysfunction. We hypothesized that high-fat feeding dysregulates the balance between endothelial derived nitric oxide and superoxide formation. Furthermore, we examined whether caloric restriction could reverse the detrimental vascular effects related to obesity. Male C57Bl/6 mice were fed with normal-fat diet (fat 17%) or high-fat diet (fat 60%) for 150 days. After establishment of obesity at day 100, a subgroup of obese mice were put on caloric restriction (CR) (70% of ad libitum energy intake) for an additional 50 days. At day 100, aortic rings from obese mice receiving high-fat diet showed impaired endothelium-dependent vasodilation in response to acetylcholine (ACh). Caloric restriction reversed high-fat diet-induced endothelial dysfunction. At day 150, impaired vasodilatory responses to ACh in obese mice without caloric restriction were markedly improved by preincubation with the tetrahydrobiopterin (BH(4)) precursor sepiapterin and L-arginine, a substrate for endothelial nitric oxide synthase (eNOS). Additionally, inhibition of vascular arginase by L-norvaline partially, and superoxide scavenging by Tiron completely, restored endothelial cell function. Obese mice showed increased vascular superoxide production, which was diminished by endothelial denudation, pretreated of the vascular rings with apocynin (an inhibitor of reduced nicotinamide adenine dinucleotide phosphate [NADPH] oxidase), oxypurinol (an inhibitor of xanthine oxidase), N(G)-nitro-L-arginine methyl ester (LNAME; an inhibitor of eNOS), or by adding the BH(4) precursor sepiapterin. Caloric restriction markedly attenuated vascular superoxide production. In obese mice on CR, endothelial denudation increased superoxide formation whereas vascular superoxide production was unaffected by L-NAME. Western blot analysis revealed decreased phosphorylated eNOS (Ser1177)-to-total eNOS expression ratio in obese mice as compared to lean controls, whereas the phospho-eNOS/NOS ratio in obese mice on CR did not differ from the lean controls. In conclusion, the present study suggests that caloric restriction reverses obesity induced endothelial dysfunction and vascular oxidative stress, and underscores the importance of uncoupled eNOS in the pathogenesis.


Asunto(s)
Restricción Calórica , Enfermedades Cardiovasculares/dietoterapia , Endotelio Vascular/fisiopatología , Obesidad/dietoterapia , Superóxidos/metabolismo , Vasodilatación , Animales , Arginasa/antagonistas & inhibidores , Arginasa/metabolismo , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Peso Corporal , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Grasas de la Dieta , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Depuradores de Radicales Libres/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/fisiopatología , Fosforilación , Factores de Tiempo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/metabolismo
19.
Blood Press ; 19(3): 196-205, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20429690

RESUMEN

There is compelling evidence to indicate an important role for increased local renin-angiotensin system activity in the pathogenesis of cardiac hypertrophy and heart failure. Resveratrol is a natural polyphenol that activates SIRT1, a novel cardioprotective and longevity factor having NAD(+)-dependent histone deacetylase activity. We tested the hypothesis whether resveratrol could prevent from angiotensin II (Ang II)-induced cardiovascular damage. Four-week-old double transgenic rats harboring human renin and human angiotensinogen genes (dTGR) were treated for 4 weeks either with SIRT1 activator resveratrol or SIRT1 inhibitor nicotinamide. Untreated dTGR and their normotensive Sprague-Dawley control rats (SD) received vehicle. Untreated dTGR developed severe hypertension as well as cardiac hypertrophy, and showed pronounced cardiovascular mortality compared with normotensive SD rats. Resveratrol slightly but significantly decreased blood pressure, ameliorated cardiac hypertrophy and prevented completely Ang II-induced mortality, whereas nicotinamide increased blood pressure without significantly influencing cardiac hypertrophy or survival. Resveratrol decreased cardiac ANP mRNA expression and induced cardiac mRNA expressions of mitochondrial biogenesis markers peroxisome proliferator-activated receptor-gamma coactivator (PGC-1alpha), mitochondrial transcription factor (Tfam), nuclear respiratory factor 1 (NRF-1) and cytochrome c oxidase subunit 4 (cox4). Resveratrol dose-dependently increased SIRT1 activity in vitro. Our findings suggest that the beneficial effects of SIRT1 activator resveratrol on Ang II-induced cardiac remodeling are mediated by blood pressure-dependent pathways and are linked to increased mitochondrial biogenesis.


Asunto(s)
Angiotensina II/farmacología , Angiotensinógeno/genética , Renina/genética , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensinógeno/metabolismo , Angiotensinógeno/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Genes/efectos de los fármacos , Corazón/fisiopatología , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/patología , Masculino , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Renina/sangre , Renina/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/genética , Resveratrol , Estilbenos
20.
J Am Soc Nephrol ; 20(2): 353-62, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18842989

RESUMEN

Although metabolic derangement plays a central role in diabetic nephropathy, a better understanding of secondary mediators of injury may lead to new therapeutic strategies. Expression of macrophage migration inhibitory factor (MIF) is increased in experimental diabetic nephropathy, and increased tubulointerstitial mRNA expression of its receptor, CD74, has been observed in human diabetic nephropathy. Whether CD74 transduces MIF signals in podocytes, however, is unknown. Here, we found glomerular and tubulointerstitial CD74 mRNA expression to be increased in Pima Indians with type 2 diabetes and diabetic nephropathy. Immunohistochemistry confirmed the increased glomerular and tubular expression of CD74 in clinical and experimental diabetic nephropathy and localized glomerular CD74 to podocytes. In cultured human podocytes, CD74 was expressed at the cell surface, was upregulated by high concentrations of glucose and TNF-alpha, and was activated by MIF, leading to phosphorylation of extracellular signal-regulated kinase 1/2 and p38. High glucose also induced CD74 expression in a human proximal tubule cell line (HK2). In addition, MIF induced the expression of the inflammatory mediators TRAIL and monocyte chemoattractant protein 1 in podocytes and HK2 cells in a p38-dependent manner. These data suggest that CD74 acts as a receptor for MIF in podocytes and may play a role in the pathogenesis of diabetic nephropathy.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/fisiología , Nefropatías Diabéticas/metabolismo , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/fisiología , Podocitos/metabolismo , Antígenos de Diferenciación de Linfocitos B/biosíntesis , Antígenos de Diferenciación de Linfocitos B/metabolismo , Biopsia , Quimiocina CCL2/metabolismo , Antígenos de Histocompatibilidad Clase II/biosíntesis , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunohistoquímica/métodos , Inflamación , Túbulos Renales/metabolismo , Sistema de Señalización de MAP Quinasas , ARN Interferente Pequeño/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/química , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA