Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(48): e2308342120, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-37983492

RESUMEN

COVID-19 pneumonia causes acute lung injury and acute respiratory distress syndrome (ALI/ARDS) characterized by early pulmonary endothelial and epithelial injuries with altered pulmonary diffusing capacity and obstructive or restrictive physiology. Growth hormone-releasing hormone receptor (GHRH-R) is expressed in the lung and heart. GHRH-R antagonist, MIA-602, has been reported to modulate immune responses to bleomycin lung injury and inflammation in granulomatous sarcoidosis. We hypothesized that MIA-602 would attenuate rVSV-SARS-CoV-2-induced pulmonary dysfunction and heart injury in a BSL-2 mouse model. Male and female K18-hACE2tg mice were inoculated with SARS-CoV-2/USA-WA1/2020, BSL-2-compliant recombinant VSV-eGFP-SARS-CoV-2-Spike (rVSV-SARS-CoV-2), or PBS, and lung viral load, weight loss, histopathology, and gene expression were compared. K18-hACE2tg mice infected with rVSV-SARS-CoV-2 were treated daily with subcutaneous MIA-602 or vehicle and conscious, unrestrained plethysmography performed on days 0, 3, and 5 (n = 7 to 8). Five days after infection mice were killed, and blood and tissues collected for histopathology and protein/gene expression. Both native SARS-CoV-2 and rVSV-SARS-CoV-2 presented similar patterns of weight loss, infectivity (~60%), and histopathologic changes. Daily treatment with MIA-602 conferred weight recovery, reduced lung perivascular inflammation/pneumonia, and decreased lung/heart ICAM-1 expression compared to vehicle. MIA-602 rescued altered respiratory rate, increased expiratory parameters (Te, PEF, EEP), and normalized airflow parameters (Penh and Rpef) compared to vehicle, consistent with decreased airway inflammation. RNASeq followed by protein analysis revealed heightened levels of inflammation and end-stage necroptosis markers, including ZBP1 and pMLKL induced by rVSV-SARS-CoV-2, that were normalized by MIA-602 treatment, consistent with an anti-inflammatory and pro-survival mechanism of action in this preclinical model of COVID-19 pneumonia.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Ratones , Masculino , Femenino , Animales , SARS-CoV-2 , COVID-19/patología , Pulmón/patología , Inflamación/patología , Síndrome de Dificultad Respiratoria/patología , Pérdida de Peso , Ratones Transgénicos , Modelos Animales de Enfermedad
2.
FASEB J ; 36(1): e22106, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34918375

RESUMEN

Choroidal neovascularization (CNV), a leading cause of blindness in the elderly, is routinely treated with vascular endothelial growth factor (VEGF) inhibitors that have limited efficacy and potentially adverse side effects. An unmet clinical need is to develop novel therapies against other angiogenic factors for alternative or combination treatment to improve efficacy and safety. We recently described secretogranin III (Scg3) as a disease-selective angiogenic factor, causally linked to diabetic retinopathy and acting independently of the VEGF pathway. An important question is whether such a disease-selective Scg3 pathway contributes to other states of pathological angiogenesis beyond diabetic retinopathy. By applying a novel in vivo endothelial ligand binding assay, we found that the binding of Scg3 to CNV vessels in live mice was markedly increased over background binding to healthy choriocapillaris and blocked by an Scg3-neutralizing antibody, whereas VEGF showed no such differential binding. Intravitreal injection of anti-Scg3 humanized antibody Fab (hFab) inhibited Matrigel-induced CNV with similar efficacy to the anti-VEGF drug aflibercept. Importantly, a combination of anti-Scg3 hFab and aflibercept synergistically alleviated CNV. Homozygous deletion of the Scg3 gene markedly reduced CNV severity and abolished the therapeutic activity of anti-Scg3 hFab, but not aflibercept, suggesting a role for Scg3 in VEGF-independent CNV pathogenesis and therapy. Our work demonstrates the stringent disease selectivity of Scg3 binding and positions anti-Scg3 hFab as a next-generation disease-targeted anti-angiogenic therapy for CNV.


Asunto(s)
Neovascularización Coroidal/metabolismo , Cromograninas/metabolismo , Transducción de Señal , Animales , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/genética , Cromograninas/genética , Femenino , Fragmentos Fab de Inmunoglobulinas/farmacología , Masculino , Ratones , Ratones Noqueados , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Cell Mol Life Sci ; 79(1): 63, 2022 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-35006382

RESUMEN

Conventional angiogenic factors, such as vascular endothelial growth factor (VEGF), regulate both pathological and physiological angiogenesis indiscriminately, and their inhibitors may elicit adverse side effects. Secretogranin III (Scg3) was recently reported to be a diabetes-restricted VEGF-independent angiogenic factor, but the disease selectivity of Scg3 in retinopathy of prematurity (ROP), a retinal disease in preterm infants with concurrent pathological and physiological angiogenesis, was not defined. Here, using oxygen-induced retinopathy (OIR) mice, a surrogate model of ROP, we quantified an exclusive binding of Scg3 to diseased versus healthy developing neovessels that contrasted sharply with the ubiquitous binding of VEGF. Functional immunohistochemistry visualized Scg3 binding exclusively to disease-related disorganized retinal neovessels and neovascular tufts, whereas VEGF bound to both disorganized and well-organized neovessels. Homozygous deletion of the Scg3 gene showed undetectable effects on physiological retinal neovascularization but markedly reduced the severity of OIR-induced pathological angiogenesis. Furthermore, anti-Scg3 humanized antibody Fab (hFab) inhibited pathological angiogenesis with similar efficacy to anti-VEGF aflibercept. Aflibercept dose-dependently blocked physiological angiogenesis in neonatal retinas, whereas anti-Scg3 hFab was without adverse effects at any dose and supported a therapeutic window at least 10X wider than that of aflibercept. Therefore, Scg3 stringently regulates pathological but not physiological angiogenesis, and anti-Scg3 hFab satisfies essential criteria for development as a safe and effective disease-targeted anti-angiogenic therapy for ROP.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Cromograninas/inmunología , Cromograninas/metabolismo , Neovascularización Patológica/genética , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/patología , Animales , Capilares/metabolismo , Cromograninas/antagonistas & inhibidores , Cromograninas/genética , Modelos Animales de Enfermedad , Fragmentos Fab de Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/efectos adversos , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/farmacología , Neovascularización Retiniana/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
4.
BMC Nephrol ; 24(1): 300, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37828432

RESUMEN

BACKGROUND: Alport syndrome (AS) is caused by mutations in type IV collagen genes that typically target and compromise the integrity of basement membranes in kidney, ocular, and sensorineural cochlear tissues. Type IV and V collagens are also integral components of arterial walls, and whereas collagenopathies including AS are implicated in aortic disease, the incidence of aortic aneurysm in AS is unknown probably because of underreporting. Consequently, AS is not presently considered an independent risk factor for aortic aneurysm and more detailed case studies including histological evidence of basement membrane abnormalities are needed to determine such a possible linkage. CASE PRESENTATION: Here, we present unique histopathological findings of an ascending aortic aneurysm collected at the time of surgery from an AS patient wherein hypertension was the only other known risk factor. CONCLUSIONS: The studies reveal classical histological features of aortic aneurysm, including atheroma, lymphocytic infiltration, elastin disruption, and myxoid degeneration with probable AS association.


Asunto(s)
Aneurisma de la Aorta Ascendente , Aneurisma de la Aorta , Nefritis Hereditaria , Humanos , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/genética , Nefritis Hereditaria/patología , Riñón/patología , Colágeno Tipo IV/genética , Aneurisma de la Aorta/diagnóstico por imagen , Aneurisma de la Aorta/genética
5.
Int J Mol Sci ; 24(13)2023 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-37445707

RESUMEN

Diabetic retinopathy (DR), a leading cause of vision loss in working-age adults, induces mosaic patterns of vasculopathy that may be associated with spatial heterogeneity of intraretinal endothelial cells. We recently reported that secretogranin III (Scg3), a neuron-derived angiogenic and vascular leakage factor, selectively binds retinal vessels of diabetic but not healthy mice. Here, we investigated endothelial heterogeneity of three retinal vascular plexuses in DR pathogenesis and the therapeutic implications. Our unique in vivo ligand binding assay detected a 22.7-fold increase in Scg3 binding to retinal vessels of diabetic mice relative to healthy mice. Functional immunohistochemistry revealed that Scg3 predominantly binds to the DR-stressed CD31- deep retinal vascular plexus but not to the relatively healthy CD31+ superficial and intermediate plexuses within the same diabetic retina. In contrast, VEGF bound to healthy and diabetic retinal vessels indiscriminately with low activity. FITC-dextran assays indicated that selectively increased retinal vascular leakage coincides with Scg3 binding in diabetic mice that was independent of VEGF, whereas VEGF-induced leakage did not distinguish between diabetic and healthy mice. Dose-response curves showed that the anti-Scg3 humanized antibody (hAb) and anti-VEGF aflibercept alleviated DR leakage with equivalent efficacies, and that the combination acted synergistically. These findings suggest: (i) the deep plexus is highly sensitive to DR; (ii) Scg3 binding to the DR deep plexus coincides with the loss of CD31 and compromised endothelial junctions; (iii) anti-Scg3 hAb alleviates vascular leakage by selectively targeting the DR-stressed deep plexus within the same diabetic retina; (iv) combined anti-Scg3 and anti-VEGF treatments synergistically ameliorate DR through distinct mechanisms.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Animales , Ratones , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/etiología , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Diabetes Mellitus Experimental/patología , Retina/metabolismo , Vasos Retinianos/metabolismo
6.
Cell Mol Life Sci ; 78(16): 5977-5985, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34230991

RESUMEN

Diabetic retinopathy (DR) is the leading cause of vision loss in working adults in developed countries. The disease traditionally classified as a microvascular complication of diabetes is now widely recognized as a neurovascular disorder resulting from disruption of the retinal neurovascular unit (NVU). The NVU comprising retinal neurons, glia and vascular cells coordinately regulates blood flow, vascular density and permeability to maintain homeostasis. Disturbance of the NVU during DR can lead to vision-threatening clinical manifestations. A limited number of signaling pathways have been identified for intercellular communication within the NVU, including vascular endothelial growth factor (VEGF), the master switch for angiogenesis. VEGF inhibitors are now widely used to treat DR, but their limited efficacy implies that other signaling molecules are involved in the pathogenesis of DR. By applying a novel screening technology called comparative ligandomics, we recently discovered secretogranin III (Scg3) as a unique DR-selective angiogenic and vascular leakage factor with therapeutic potential for DR. This review proposes neuron-derived Scg3 as the first diabetes-selective neurovascular regulator and discusses important features of Scg3 inhibition for next-generation disease-targeted anti-angiogenic therapies of DR.


Asunto(s)
Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Animales , Cromograninas/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Humanos , Neuronas/metabolismo , Neuronas/patología , Retina/metabolismo , Retina/patología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Int J Mol Sci ; 23(12)2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35743114

RESUMEN

Alport syndrome (AS) is a hereditary renal disorder with no etiological therapy. In the preclinical Col4a3-/- model of AS, disease progression and severity vary depending on mouse strain. The sodium-glucose cotransporter 2 (SGLT2) is emerging as an attractive therapeutic target in cardiac/renal pathologies, but its application to AS remains untested. This study investigates cardiorespiratory function and SGLT2 renal expression in Col4a3-/- mice from three different genetic backgrounds, 129x1/SvJ, C57Bl/6 and Balb/C. male Col4a3-/- 129x1/SvJ mice displayed alterations consistent with heart failure with preserved ejection fraction (HFpEF). Female, but not male, C57Bl/6 and Balb/C Col4a3-/- mice exhibited mild changes in systolic and diastolic function of the heart by echocardiography. Male C57Bl/6 Col4a3-/- mice presented systolic dysfunction by invasive hemodynamic analysis. All strains except Balb/C males demonstrated alterations in respiratory function. SGLT2 expression was significantly increased in AS compared to WT mice from all strains. However, cardiorespiratory abnormalities and SGLT2 over-expression were significantly less in AS Balb/C mice compared to the other two strains. Systolic blood pressure was significantly elevated only in mutant 129x1/SvJ mice. The results provide further evidence for strain-dependent cardiorespiratory and hypertensive phenotype variations in mouse AS models, corroborated by renal SGLT2 expression, and support ongoing initiatives to develop SGLT2 inhibitors for the treatment of AS.


Asunto(s)
Autoantígenos/metabolismo , Colágeno Tipo IV/metabolismo , Insuficiencia Cardíaca , Nefritis Hereditaria , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nefritis Hereditaria/genética , Fenotipo , Transportador 2 de Sodio-Glucosa/genética , Volumen Sistólico
8.
Am J Physiol Heart Circ Physiol ; 320(5): H1862-H1872, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33769915

RESUMEN

There are currently no Food and Drug Administration-approved treatments for heart failure with preserved ejection fraction (HFpEF). Here we compared the effects of exercise with and without α/ß-adrenergic blockade with carvedilol in Col4a3-/- Alport mice, a model of the phenogroup 3 subclass of HFpEF with underlying renal dysfunction. Alport mice were assigned to the following groups: no treatment control (n = 29), carvedilol (n = 11), voluntary exercise (n = 9), and combination carvedilol and exercise (n = 8). Cardiac function was assessed by echocardiography after 4-wk treatments. Running activity of Alport mice was similar to wild types at 1 mo of age but markedly reduced at 2 mo (1.3 ± 0.40 vs. 4.5 ± 1.02 km/day, P < 0.05). There was a nonsignificant trend for increased running activity at 2 mo by carvedilol in the combination treatment group. Combination treatments conferred increased body weight of Col4a3-/- mice (22.0 ± 1.18 vs. 17.8 ± 0.29 g in untreated mice, P < 0.01), suggesting improved physiology, and heart rates declined by similar increments in all carvedilol-treatment groups. The combination treatment improved systolic parameters; stroke volume (30.5 ± 1.99 vs. 17.8 ± 0.77 µL, P < 0.0001) as well as ejection fraction and global longitudinal strain compared with controls. Myocardial performance index was normalized by all interventions (P < 0.0001). Elevated osteopontin plasma levels in control Alport mice were significantly lowered only by combination treatment, and renal function of the Alport group assessed by urine albumin creatinine ratio was significantly improved by all treatments. The results support synergistic roles for exercise and carvedilol to augment cardiac systolic function of Alport mice with moderately improved renal functions but no change in diastole.NEW & NOTEWORTHY In an Alport mouse model of heart failure with preserved ejection fraction (HFpEF), exercise and carvedilol synergistically improved systolic function without affecting diastole. Carvedilol alone or in combination with exercise also improved kidney function. Molecular analyses indicate that the observed improvements in cardiorenal functions were mediated at least in part by effects on serum osteopontin and related inflammatory cytokine cascades. The work presents new potential therapeutic targets and approaches for HFpEF.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Carvedilol/farmacología , Colágeno Tipo IV/deficiencia , Terapia por Ejercicio , Insuficiencia Cardíaca/terapia , Nefritis Hereditaria/terapia , Osteopontina/sangre , Disfunción Ventricular Izquierda/terapia , Función Ventricular Izquierda/efectos de los fármacos , Animales , Autoantígenos/genética , Biomarcadores/sangre , Colágeno Tipo IV/genética , Terapia Combinada , Diástole , Modelos Animales de Enfermedad , Regulación hacia Abajo , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Ratones de la Cepa 129 , Ratones Noqueados , Nefritis Hereditaria/sangre , Nefritis Hereditaria/genética , Nefritis Hereditaria/fisiopatología , Recuperación de la Función , Sístole , Disfunción Ventricular Izquierda/sangre , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología
9.
Am J Physiol Regul Integr Comp Physiol ; 320(5): R575-R587, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33565369

RESUMEN

Approximately 14% of the general population suffer from chronic kidney disease that can lead to acute kidney injury (AKI), a condition with up to 50% mortality for which there is no effective treatment. Hypertension, diabetes, and cardiovascular disease are the main comorbidities, and more than 660,000 Americans have kidney failure. ß2-Adrenergic receptors (ß2ARs) have been extensively studied in association with lung and cardiovascular disease, but with limited scope in kidney and renal diseases. ß2ARs are expressed in multiple parts of the kidney including proximal and distal convoluted tubules, glomeruli, and podocytes. Classical and noncanonical ß2AR signaling pathways interface with other intracellular mechanisms in the kidney to regulate important cellular functions including renal blood flow, electrolyte balance and salt handling, and tubular function that in turn exert control over critical physiology and pathology such as blood pressure and inflammatory responses. Nephroprotection through activation of ß2ARs has surfaced as a promising field of investigation; however, there is limited data on the pharmacology and potential side effects of renal ß2AR modulation. Here, we provide updates on some of the major areas of preclinical kidney research involving ß2AR signaling that have advanced to describe molecular pathways and identify potential drug targets some of which are currently under clinical development for the treatment of kidney-related diseases.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Riñón/efectos de los fármacos , Receptores Adrenérgicos beta 2/efectos de los fármacos , Equilibrio Hidroelectrolítico/efectos de los fármacos , Animales , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal
10.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062733

RESUMEN

Retinopathy of prematurity (ROP) is an ocular vascular disease affecting premature infants, characterized by pathological retinal neovascularization (RNV), dilated and tortuous retinal blood vessels, and retinal or vitreous hemorrhages that may lead to retinal detachment, vision impairment and blindness. Compared with other neovascular diseases, ROP is unique because of ongoing and concurrent physiological and pathological angiogenesis in the developing retina. While the disease is currently treated by laser or cryotherapy, anti-vascular endothelial growth factor (VEGF) agents have been extensively investigated but are not approved in the U.S. because of safety concerns that they negatively interfere with physiological angiogenesis of the developing retina. An ideal therapeutic strategy would selectively inhibit pathological but not physiological angiogenesis. Our group recently described a novel strategy that selectively and safely alleviates pathological RNV in animal models of ROP by targeting secretogranin III (Scg3), a disease-restricted angiogenic factor. The preclinical profile of anti-Scg3 therapy presents a high potential for next-generation disease-targeted anti-angiogenic therapy for the ROP indication. This review focuses on retinal vessel development in neonates, the pathogenesis of ROP and its underlying molecular mechanisms, including different animal models, and provides a summary of current and emerging therapies.


Asunto(s)
Cromograninas/genética , Neovascularización Patológica/tratamiento farmacológico , Oxígeno/uso terapéutico , Retinopatía de la Prematuridad/tratamiento farmacológico , Animales , Animales Recién Nacidos , Cromograninas/antagonistas & inhibidores , Humanos , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Retina/efectos de los fármacos , Retina/crecimiento & desarrollo , Retina/patología , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/patología , Factor A de Crecimiento Endotelial Vascular/genética
11.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008775

RESUMEN

The retinopathy of prematurity (ROP), a neovascular retinal disorder presenting in premature infants, is the leading causes of blindness in children. Currently, there is no approved drug therapy for ROP in the U.S., highlighting the urgent unmet clinical need for a novel therapeutic to treat the disease. Secretogranin III (Scg3) was recently identified as a disease-selective angiogenic factor, and Scg3-neutralizing monoclonal antibodies were reported to alleviate pathological retinal neovascularization in mouse models. In this study, we characterized the efficacy and safety of a full-length humanized anti-Scg3 antibody (hAb) to ameliorate retinal pathology in oxygen-induced retinopathy (OIR) mice, a surrogate model of ROP, by implementing histological and functional analyses. Our results demonstrate that the anti-Scg3 hAb outperforms the vascular endothelial growth factor inhibitor aflibercept in terms of efficacy and safety to treat OIR mice. Our findings support the development of anti-Scg3 hAb for clinical application.


Asunto(s)
Anticuerpos/uso terapéutico , Cromograninas/inmunología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/inmunología , Animales , Animales Recién Nacidos , Anticuerpos/administración & dosificación , Humanos , Inyecciones Intravítreas , Ratones Endogámicos C57BL , Oxígeno , Enfermedades de la Retina/fisiopatología , Neovascularización Retiniana/patología , Resultado del Tratamiento , Visión Ocular
12.
Circ Res ; 122(10): 1395-1408, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29618597

RESUMEN

RATIONALE: Vascular calcification (VC) is a marker of the severity of atherosclerotic disease. Hormones play important roles in regulating calcification; estrogen and parathyroid hormones exert opposing effects, the former alleviating VC and the latter exacerbating it. To date no treatment strategies have been developed to regulate clinical VC. OBJECTIVE: The objective of this study was to investigate the effect of growth hormone-releasing hormone (GHRH) and its agonist (GHRH-A) on the blocking of VC in a mouse model. METHODS AND RESULTS: Young adult osteoprotegerin-deficient mice were given daily subcutaneous injections of GHRH-A (MR409) for 4 weeks. Significant reductions in calcification of the aortas of MR409-treated mice were paralleled by markedly lower alkaline phosphatase activity and a dramatic reduction in the expression of transcription factors, including the osteogenic marker gene Runx2 and its downstream factors, osteonectin and osteocalcin. The mechanism of action of GHRH-A was dissected in smooth muscle cells isolated from human and mouse aortas. Calcification of smooth muscle cells induced by osteogenic medium was inhibited in the presence of GHRH or MR409, as evidenced by reduced alkaline phosphatase activity and Runx2 expression. Inhibition of calcification by MR409 was partially reversed by MIA602, a GHRH antagonist, or a GHRH receptor-selective small interfering RNA. Treatment with MR409 induced elevated cytosolic cAMP and its target, protein kinase A which in turn blocked nicotinamide adenine dinucleotide phosphate oxidase activity and reduced production of reactive oxygen species, thus blocking the phosphorylation of nuclear factor κB (p65), a key intermediate in the ligand of receptor activator for nuclear factor-κ B-Runx2/alkaline phosphatase osteogenesis program. A protein kinase A-selective small interfering RNA or the chemical inhibitor H89 abolished these beneficial effects of MR409. CONCLUSIONS: GHRH-A controls osteogenesis in smooth muscle cells by targeting cross talk between protein kinase A and nuclear factor κB (p65) and through the suppression of reactive oxygen species production that induces the Runx2 gene and alkaline phosphatase. Inflammation-mediated osteogenesis is thereby blocked. GHRH-A may represent a new pharmacological strategy to regulate VC.


Asunto(s)
Fragmentos de Péptidos/uso terapéutico , Calcificación Vascular/prevención & control , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Animales , Aorta/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Medios de Cultivo/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hormona Liberadora de Hormona del Crecimiento , Trasplante de Corazón , Humanos , Isoquinolinas/farmacología , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Osteoprotegerina/deficiencia , Fragmentos de Péptidos/farmacología , ARN Interferente Pequeño/genética , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/antagonistas & inhibidores , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Sulfonamidas/farmacología , Factor de Transcripción ReIA/metabolismo , Calcificación Vascular/fisiopatología
13.
Am J Physiol Heart Circ Physiol ; 317(4): H867-H876, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31441677

RESUMEN

Atherosclerosis is the most common underlying cause of cardiovascular morbidity and mortality worldwide. c-Kit (CD117) is a member of the receptor tyrosine kinase family, which regulates differentiation, proliferation, and survival of multiple cell types. Recent studies have shown that c-Kit and its ligand stem cell factor (SCF) are present in arterial endothelial cells and smooth muscle cells (SMCs). The role of c-Kit in cardiovascular disease remains unclear. The aim of the current study is to determine the role of c-Kit in atherogenesis. For this purpose, atherosclerotic plaques were quantified in c-Kit-deficient mice (KitMut) after they were fed a high-fat diet (HFD) for 16 wk. KitMut mice demonstrated substantially greater atherosclerosis compared with control (KitWT) littermates (P < 0.01). Transplantation of c-Kit-positive bone marrow cells into KitMut mice failed to rescue the atherogenic phenotype, an indication that increased atherosclerosis was associated with reduced arterial c-Kit. To investigate the mechanism, SMC organization and morphology were analyzed in the aorta by histopathology and electron microscopy. SMCs were more abundant, disorganized, and vacuolated in aortas of c-Kit mutant mice compared with controls (P < 0.05). Markers of the "contractile" SMC phenotype (calponin, SM22α) were downregulated with pharmacological and genetic c-Kit inhibition (P < 0.05). The absence of c-Kit increased lipid accumulation and significantly reduced the expression of the ATP-binding cassette transporter G1 (ABCG1) necessary for lipid efflux in SMCs. Reconstitution of c-Kit in cultured KitMut SMCs resulted in increased spindle-shaped morphology, reduced proliferation, and elevated levels of contractile markers, all indicators of their restored contractile phenotype (P < 0.05).NEW & NOTEWORTHY This study describes the novel vasculoprotective role of c-Kit against atherosclerosis and its function in the preservation of the SMC contractile phenotype.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Hiperlipidemias/complicaciones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aorta/metabolismo , Aorta/ultraestructura , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hiperlipidemias/metabolismo , Ratones Noqueados para ApoE , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/ultraestructura , Mutación , Miocitos del Músculo Liso/ultraestructura , Fenotipo , Placa Aterosclerótica , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal , Calponinas
14.
Cell Mol Life Sci ; 75(4): 635-647, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28856381

RESUMEN

Secretogranin III (Scg3) is a member of the granin protein family that regulates the biogenesis of secretory granules. Scg3 was recently discovered as an angiogenic factor, expanding its functional role to extrinsic regulation. Unlike many other known angiogenic factors, the pro-angiogenic actions of Scg3 are restricted to pathological conditions. Among thousands of quantified endothelial ligands, Scg3 has the highest binding activity ratio to diabetic vs. healthy mouse retinas and lowest background binding to normal vessels. In contrast, vascular endothelial growth factor binds to and stimulates angiogenesis of both diabetic and control vasculature. Consistent with its role in pathological angiogenesis, Scg3-neutralizing antibodies alleviate retinal vascular leakage in mouse models of diabetic retinopathy and retinal neovascularization in oxygen-induced retinopathy mice. This review summarizes our current knowledge of Scg3 as a regulatory protein of secretory granules, highlights its new role as a highly disease-selective angiogenic factor, and envisions Scg3 inhibitors as "selective angiogenesis blockers" for targeted therapy.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Cromograninas/fisiología , Retinopatía Diabética , Neovascularización Patológica/genética , Animales , Cromograninas/genética , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Humanos , Ratones , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Vesículas Secretoras/patología
15.
Ann Vasc Surg ; 48: 222-232, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29197603

RESUMEN

BACKGROUND: Lack of a reliable hind limb gangrene animal model limits preclinical studies of gangrene, a severe form of critical limb ischemia. We develop a novel mouse hind limb gangrene model to facilitate translational studies. METHODS: BALB/c, FVB, and C57BL/6 mice underwent femoral artery ligation (FAL) with or without administration of NG-nitro-L-arginine methyl ester (L-NAME), an endothelial nitric oxide synthase inhibitor. Gangrene was assessed using standardized ischemia scores ranging from 0 (no gangrene) to 12 (forefoot gangrene). Laser Doppler imaging (LDI) and DiI perfusion quantified hind limb reperfusion postoperatively. RESULTS: BALB/c develops gangrene with FAL-only (n = 11/11, 100% gangrene incidence), showing mean limb ischemia score of 12 on postoperative days (PODs) 7 and 14 with LDI ranging from 0.08 to 0.12 on respective PODs. Most FVB did not develop gangrene with FAL-only (n = 3/9, 33% gangrene incidence) but with FAL and L-NAME (n = 9/9, 100% gangrene incidence). Mean limb ischemia scores for FVB undergoing FAL with L-NAME were significantly higher than for FVB receiving FAL-only. LDI score and capillary density by POD 28 were significantly lower in FVB undergoing FAL with L-NAME. C57BL/6 did not develop gangrene with FAL-only or FAL and L-NAME. CONCLUSIONS: Reproducible murine gangrene models may elucidate molecular mechanisms for gangrene development, facilitating therapeutic intervention.


Asunto(s)
Arteria Femoral/cirugía , Isquemia/etiología , Músculo Esquelético/irrigación sanguínea , NG-Nitroarginina Metil Éster , Enfermedad Arterial Periférica/etiología , Animales , Velocidad del Flujo Sanguíneo , Modelos Animales de Enfermedad , Gangrena , Miembro Posterior , Isquemia/enzimología , Isquemia/patología , Isquemia/fisiopatología , Ligadura , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Enfermedad Arterial Periférica/enzimología , Enfermedad Arterial Periférica/patología , Enfermedad Arterial Periférica/fisiopatología , Flujo Sanguíneo Regional , Especificidad de la Especie , Factores de Tiempo
16.
Angiogenesis ; 20(4): 479-492, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28447229

RESUMEN

Angiogenic factors play an important role in the pathogenesis of diabetic retinopathy (DR), neovascular age-related macular degeneration (nAMD) and retinopathy of prematurity (ROP). Pleiotrophin, a well-known angiogenic factor, was recently reported to be upregulated in the vitreous fluid of patients with proliferative DR (PDR). However, its pathogenic role and therapeutic potential in ocular vascular diseases have not been defined in vivo. Here using corneal pocket assays, we demonstrated that pleiotrophin induced angiogenesis in vivo. To investigate the pathological role of pleiotrophin we used neutralizing antibody to block its function in multiple in vivo models of ocular vascular diseases. In a mouse model of DR, intravitreal injection of pleiotrophin-neutralizing antibody alleviated diabetic retinal vascular leakage. In a mouse model of oxygen-induced retinopathy (OIR), which is a surrogate model of ROP and PDR, we demonstrated that intravitreal injection of anti-pleiotrophin antibody prevented OIR-induced pathological retinal neovascularization and aberrant vessel tufts. Finally, pleiotrophin-neutralizing antibody ameliorated laser-induced choroidal neovascularization, a mouse model of nAMD, suggesting that pleiotrophin is involved in choroidal vascular disease. These findings suggest that pleiotrophin plays an important role in the pathogenesis of DR with retinal vascular leakage, ROP with retinal neovascularization and nAMD with choroidal neovascularization. The results also support pleiotrophin as a promising target for anti-angiogenic therapy.


Asunto(s)
Proteínas Portadoras/uso terapéutico , Citocinas/uso terapéutico , Neovascularización Retiniana/tratamiento farmacológico , Animales , Proteínas Portadoras/farmacología , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/patología , Citocinas/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Retina/efectos de los fármacos , Retina/patología , Neovascularización Retiniana/patología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología
17.
Stem Cells ; 34(7): 1846-58, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27145179

RESUMEN

Our previous study showed that the therapeutic effects of mesenchymal stem cells (MSCs) transplantation were improved by enhancing migration. MicroRNA-211 (miR-211) can modulate the migratory properties of some cell types by mechanisms that are not fully understood. This study was designed to investigate a possible role for miR-211 in MSC migration, and whether genetic manipulation of miR-211 in MSCs could be used to enhance its beneficial effects of cell transplantation. Transwell assays confirmed that MSCs migration of was significantly impaired by miR-211 knockdown but enhanced by miR-211 overexpression. MiR-211 overexpressing MSCs also exhibited significantly increased cell engraftment in the peri-infarct areas of female rat hearts 2 days after intravenous transplantation of male MSCs as shown by GFP tracking and SYR gene quantification. This conferred a significant decrease in infarct size and improved cardiac performance. By using a loss or gain of gene function approach, we demonstrated that miR-211 targeted STAT5A to modulate MSCs migration, possibly by interacting with MAPK signaling. Furthermore, the beneficial effects of miR-211 overexpression in MSCs were abolished by simultaneous overexpression of STAT5A whereas the negative effects of miR-211 silencing on MSC migration were rescued by simultaneous downregulation of STAT5A. Finally, using ChIP-PCR and luciferase assays, we provide novel evidence that STAT3 can directly bind to promoter elements that activate miR-211 expression. STAT3/miR-211/STAT5A signaling plays a key role in MSCs migration. Intravenous infusion of genetically modified miR-211 overexpressing MSCs conveys enhanced protection from adverse post-MI remodeling compared with unmodified MSCs. Stem Cells 2016;34:1846-1858.


Asunto(s)
Movimiento Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , MicroARNs/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Administración Intravenosa , Animales , Secuencia de Bases , Hipoxia de la Célula/genética , Senescencia Celular/genética , Regulación hacia Abajo/genética , Femenino , Humanos , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Ratas Sprague-Dawley
18.
Arterioscler Thromb Vasc Biol ; 36(4): 663-672, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26868211

RESUMEN

OBJECTIVE: The efficiency of cell therapy is limited by poor cell survival and engraftment. Here, we studied the effect of the growth hormone-releasing hormone agonist, JI-34, on mesenchymal stem cell (MSC) survival and angiogenic therapy in a mouse model of critical limb ischemia. APPROACH AND RESULTS: Mouse bone marrow-derived MSCs were incubated with or without 10(-8) mol/L JI-34 for 24 hours. MSCs were then exposed to hypoxia and serum deprivation to detect the effect of preconditioning on cell apoptosis, migration, and tube formation. For in vivo tests, critical limb ischemia was induced by femoral artery ligation. After surgery, mice received 50 µL phosphate-buffered saline or with 1×10(6) MSCs or with 1×10(6) JI-34-reconditioned MSCs. Treatment of MSCs with JI-34 improved MSC viability and mobility and markedly enhanced their capability to promote endothelial tube formation in vitro. These effects were paralleled by an increased phosphorylation and nuclear translocation of signal transducer and activator of transcription 3. In vivo, JI-34 pretreatment enhanced the engraftment of MSCs into ischemic hindlimb muscles and augmented reperfusion and limb salvage compared with untreated MSCs. Significantly more vasculature and proliferating CD31(+) and CD34(+) cells were detected in ischemic muscles that received MSCs treated with JI-34. CONCLUSIONS: Our studies demonstrate a novel role for JI-34 to markedly improve therapeutic angiogenesis in hindlimb ischemia by increasing the viability and mobility of MSCs. These findings support additional studies to explore the full potential of growth hormone-releasing hormone agonists to augment cell therapy in the management of ischemia.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/análogos & derivados , Hormona Liberadora de Hormona del Crecimiento/agonistas , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Fragmentos de Péptidos/farmacología , Transporte Activo de Núcleo Celular , Animales , Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Hormona Liberadora de Hormona del Crecimiento/farmacología , Miembro Posterior , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/agonistas , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
19.
Stem Cells ; 33(6): 1850-1862, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25754303

RESUMEN

Heparanase plays important roles in tumor angiogenesis. Our previous study demonstrated that hypoxic preconditioning (HPC) enhanced the angiogenic and therapeutic effects of mesenchymal stem cells (MSCs), effects that were paralleled by enhanced heparanase expression. This study was designed to elucidate the role of heparanase in the improved therapeutic properties of HPC-MSCs and to explore underlying mechanisms using an ischemic rat hind limb model. MSCs transfected with heparanase (MSC(hpa) ) or empty vector (MSC(null) ) were delivered by intramuscular injections to ischemic hind limbs. Hind limbs that received MSC(hpa) recovered blood flow more rapidly at 7 days and acquired higher capillary density at 14 days compared with MSC(null) . Conditioned medium from MSC(hpa) increased endothelial cell migration and promoted greater tube formation relative to that from the MSC(null) groups. Vascular endothelial growth factor receptor 2 (VEGFR2, Flk-1) and its downstream signaling pathway (p38MAPK/HSP27) were significantly increased in human umbilical vein endothelial cells (HUVECs) after treatment with MSC(hpa) conditioned medium. Each of these responses was decreased by cocultured with MSC(hpa-KD) conditioned medium. MSC(hpa) conditioned medium activated hypoxia-inducible factor-2α (HIF-2α) and increased in parallel the transcript level of Flk-1 as determined by chromatin immunoprecipitation-PCR and luciferase assays. Analyses of integrin expression revealed an important role for integrin ß1 in the regulation of HIF-2α. All angiogenic effects of MSC(hpa) conditioned medium were abolished by knockdown of integrin ß1, HIF-2α, and Flk-1 in HUVECs with selective shRNAs. These findings identify heparanse as a key regulator of angiogenesis by MSCs. We propose a novel pathway wherein heparanse sequentially activates integrin ß1, HIF-2α, Flk-1, and p38MAPK/HSP27 with corresponding enhancement of angiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular/fisiología , Glucuronidasa/metabolismo , Integrina beta1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Patológica/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Neovascularización Fisiológica , Ratas Sprague-Dawley , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Stem Cells ; 32(10): 2702-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24989835

RESUMEN

Hypoxia preconditioning enhances the therapeutic effect of mesenchymal stem cells (MSCs). However, the mechanism underlying hypoxia-induced augmentation of the protective effect of MSCs on myocardial infarction (MI) is poorly understood. We show that hypoxia-enhanced survival, mobility, and protection of cocultured cardiomyocytes were paralleled by increased expression of leptin and cell surface receptor CXCR4. The enhanced activities were abolished by either knockdown of leptin with a selective shRNA or by genetic deficiency of leptin or its receptor in MSCs derived, respectively, from ob/ob or db/db mice. To characterize the role of leptin in the regulation of MSC functions by hypoxia and its possible contribution to enhanced therapeutic efficacy, cell therapy using MSCs derived from wild-type, ob/ob, or db/db mice was implemented in mouse models of acute MI. Augmented protection by hypoxia pretreatment was only seen with MSCs from wild-type mice. Parameters that were differentially affected by hypoxia pretreatment included MSC engraftment, c-Kit(+) cell recruitment to the infarct, vascular density, infarct size, and long-term contractile function. These data show that leptin signaling is an early and essential step for the enhanced survival, chemotaxis, and therapeutic properties of MSCs conferred by preculture under hypoxia. Leptin may play a physiological role in priming MSCs resident in the bone marrow endosteum for optimal response to systemic signaling molecules and subsequent tissue repair.


Asunto(s)
Hipoxia/patología , Precondicionamiento Isquémico Miocárdico , Leptina/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Transducción de Señal , Animales , Apoptosis , Supervivencia Celular , Citoprotección , Pruebas de Función Cardíaca , Humanos , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Comunicación Paracrina , Receptores CXCR4/metabolismo , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA