Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.056
Filtrar
1.
J Biol Chem ; 299(2): 102804, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36529290

RESUMEN

Fibroblast growth factor (FGF) is a multifunctional protein that exhibits a wide range of biological effects. Most commonly, it acts as a mitogen, but it also has regulatory, morphological, and endocrine effects. The four receptor subtypes of FGF are activated by more than 20 different FGF ligands. FGF2, one of the FGF ligands, is an essential factor for cell culture in stem cells for regenerative medicine; however, recombinant FGF2 is extremely unstable. Here, we successfully generated homobivalent agonistic single-domain antibodies (variable domain of heavy chain of heavy chain antibodies referred to as VHHs) that bind to domain III and induce activation of the FGF receptor 1 and thus transduce intracellular signaling. This agonistic VHH has similar biological activity (EC50) as the natural FGF2 ligand. Furthermore, we determined that the agonistic VHH could support the proliferation of human-induced pluripotent stem cells (PSCs) and human mesenchymal stem cells, which are PSCs for regenerative medicine. In addition, the agonistic VHH could maintain the ability of mesenchymal stem cells to differentiate into adipocytes or osteocytes, indicating that it could maintain the properties of PSCs. These results suggest that the VHH agonist may function as an FGF2 mimetic in cell preparation of stem cells for regenerative medicine with better cost effectiveness.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Dominios Proteicos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Anticuerpos de Dominio Único , Humanos , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ligandos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Osteocitos/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/agonistas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Medicina Regenerativa , Transducción de Señal/efectos de los fármacos , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología
2.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884557

RESUMEN

Octacalcium phosphate (OCP, Ca8H2(PO4)6·5H2O) is known to be a possible precursor of biological hydroxyapatite formation of organic bone tissue. OCP has higher biocompatibility and osseointegration rate compared to other calcium phosphates. In this work, the synthesis of low-temperature calcium phosphate compounds and substituted forms of those at physiological temperatures is shown. Strontium is used to improve bioactive properties of the material. Strontium was inserted into the OCP structure by ionic substitution in solutions. The processes of phase formation of low-temperature OCP with theoretical substitution of strontium for calcium up to 50 at.% in conditions close to physiological, i.e., temperature 35-37 °C and normal pressure, were described. The effect of strontium substitution range on changes in the crystal lattice of materials, the microstructural features, surface morphology and biological properties in vitro has been established. The results of the study indicate the effectiveness of using strontium in OCP for improving biocompatibility of OCP based composite materials intended for bone repair.


Asunto(s)
Materiales Biocompatibles/farmacología , Regeneración Ósea , Huesos/citología , Fosfatos de Calcio/síntesis química , Fosfatos de Calcio/farmacología , Mesodermo/citología , Animales , Materiales Biocompatibles/síntesis química , Huesos/efectos de los fármacos , Durapatita/química , Técnicas In Vitro , Mesodermo/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Especies Reactivas de Oxígeno/metabolismo , Estroncio/química , Ingeniería de Tejidos
3.
Nutrients ; 13(7)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209455

RESUMEN

Glucose-based solutions remain the most used osmotic agents in peritoneal dialysis (PD), but unavoidably they contribute to the loss of peritoneal filtration capacity. Here, we evaluated at a molecular level the effects of XyloCore, a new PD solution with a low glucose content, in mesothelial and endothelial cells. Cell viability, integrity of mesothelial and endothelial cell membrane, activation of mesothelial and endothelial to mesenchymal transition programs, inflammation, and angiogenesis were evaluated by several techniques. Results showed that XyloCore preserves mesothelial and endothelial cell viability and membrane integrity. Moreover XyloCore, unlike glucose-based solutions, does not exert pro-fibrotic, -inflammatory, and -angiogenic effects. Overall, the in vitro evidence suggests that XyloCore could represent a potential biocompatible solution promising better outcomes in clinical practice.


Asunto(s)
Soluciones para Diálisis/farmacología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Glucosa/farmacología , Inflamación/patología , Mesodermo/metabolismo , Neovascularización Fisiológica , Diálisis Peritoneal , Biomarcadores/metabolismo , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Impedancia Eléctrica , Células Epiteliales/efectos de los fármacos , Epitelio/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Mesodermo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Permeabilidad , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Cell Death Dis ; 12(7): 677, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226515

RESUMEN

Muscular dystrophies are debilitating neuromuscular disorders for which no cure exists. As this disorder affects both cardiac and skeletal muscle, patients would benefit from a cellular therapy that can simultaneously regenerate both tissues. The current protocol to derive bipotent mesodermal progenitors which can differentiate into cardiac and skeletal muscle relies on the spontaneous formation of embryoid bodies, thereby hampering further clinical translation. Additionally, as skeletal muscle is the largest organ in the human body, a high myogenic potential is necessary for successful regeneration. Here, we have optimized a protocol to generate chemically defined human induced pluripotent stem cell-derived mesodermal progenitors (cdMiPs). We demonstrate that these cells contribute to myotube formation and differentiate into cardiomyocytes, both in vitro and in vivo. Furthermore, the addition of valproic acid, a clinically approved small molecule, increases the potential of the cdMiPs to contribute to myotube formation that can be prevented by NOTCH signaling inhibitors. Moreover, valproic acid pre-treated cdMiPs injected in dystrophic muscles increase physical strength and ameliorate the functional performances of transplanted mice. Taken together, these results constitute a novel approach to generate mesodermal progenitors with enhanced myogenic potential using clinically approved reagents.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mesodermo/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Receptores Notch/metabolismo , Ácido Valproico/farmacología , Animales , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/trasplante , Ratones , Ratones Noqueados , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/trasplante , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofias Musculares/cirugía , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Fenotipo , Ratas , Transducción de Señal
5.
ChemMedChem ; 16(21): 3300-3305, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34309224

RESUMEN

Human pluripotent stem cells (hPSCs) hold great promise for applications in cell therapy and drug screening in the cardiovascular field. Bone morphogenetic protein 4 (BMP4) is key for early cardiac mesoderm induction in hPSC and subsequent cardiomyocyte derivation. Small-molecular BMP4 mimetics may help to standardize cardiomyocyte derivation from hPSCs. Based on observations that chalcones can stimulate BMP4 signaling pathways, we hypothesized their utility in cardiac mesoderm induction. To test this, we set up a two-tiered screening strategy, (1) for directed differentiation of hPSCs with commercially available chalcones (4'-hydroxychalcone [4'HC] and Isoliquiritigen) and 24 newly synthesized chalcone derivatives, and (2) a functional screen to assess the propensity of the obtained cardiomyocytes to self-organize into contractile engineered human myocardium (EHM). We identified 4'HC, 4-fluoro-4'-methoxychalcone, and 4-fluoro-4'-hydroxychalcone as similarly effective in cardiac mesoderm induction, but only 4'HC as an effective replacement for BMP4 in the derivation of contractile EHM-forming cardiomyocytes.


Asunto(s)
Chalconas/farmacología , Mesodermo/efectos de los fármacos , Miocardio/citología , Células Madre Pluripotentes/efectos de los fármacos , Ingeniería de Tejidos , Chalconas/química , Relación Dosis-Respuesta a Droga , Humanos , Mesodermo/metabolismo , Estructura Molecular , Células Madre Pluripotentes/metabolismo , Relación Estructura-Actividad
6.
Chem Biol Interact ; 345: 109562, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34153226

RESUMEN

Advanced glycation end products (AGEs) are associated with the pathogenesis of diabetic vascular complications. Induction of the endothelial-to-mesenchymal transition (EndMT) is associated with the pathogenesis of fibrotic diseases. The roles of AGEs in islet EndMT induction and diabetes-related islet microvasculopathy and fibrosis remain unclear. This study investigated the pathological roles of AGEs in islet EndMT induction and fibrosis in vitro and in vivo. Non-cytotoxic concentrations of AGEs upregulated the protein expression of fibronectin, vimentin, and α-smooth muscle actin (α-SMA) (mesenchymal/myofibroblast markers) and downregulated the protein expression of vascular endothelial (VE)-cadherin and cluster of differentiation (CD) 31 (endothelial cell markers) in cultured mouse pancreatic islet endothelial cells, which was prevented by the AGE cross-link breaker alagebrium chloride. In streptozotocin-induced diabetic mice, the average islet area and islet immunoreactivities for insulin and CD31 were decreased and the islet immunoreactivities for AGEs and α-SMA and fibrosis were increased, which were prevented by the AGE inhibitor aminoguanidine. Immunofluorescence double staining showed that α-SMA-positive staining co-localized with CD31-positive staining in the diabetic islets, which was effectively prevented by aminoguanidine. These results demonstrate that AGEs can induce EndMT in islet endothelial cells and islet fibrosis in diabetic mice, suggesting that AGE-induced EndMT may contribute to islet fibrosis in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Productos Finales de Glicación Avanzada/farmacología , Islotes Pancreáticos/patología , Mesodermo/patología , Animales , Diferenciación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Insulina/metabolismo , Mesodermo/efectos de los fármacos , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
7.
Biomed Res Int ; 2021: 6657206, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33860049

RESUMEN

The study is aimed at investigating the effects of Ginkgo biloba extract EGB761 on renal tubular damage and endoplasmic reticulum stress (ERS) in diabetic kidney disease (DKD). A total of 50 C57BL/6 N mice were randomly divided into the normal group, DKD group, DKD+EGB761 group (36 mg/kg), and DKD+4-phenylbutyrate (4-PBA) group (1 g/kg). The DKD model was replicated by high-fat diet combined with intraperitoneal injection of streptozotocin (STZ). Renal tubular epithelial cells (HK-2) were divided into the control group, high-glucose group (30 mmol/L), EGB761 group (40 mg/L, 20 mg/L, 10 mg/L), TM group, and TM+4-PBA group. After 8 weeks of administration, expressions of serum creatinine (Scr), blood urea nitrogen (BUN), 24 h urinary protein (24 h Pro), fasting blood glucose (FBG), ß 2-microglobulin (ß 2-MG), and retinol binding protein 4 (RBP4) of mice were tested. The pathological changes of renal tissue were observed. The expressions of extracellular matrix (ECM) accumulation and epithelial-mesenchymal transition (EMT) markers α-smooth muscle actin (α-SMA), E-cadherin, fibronectin, and collagen IV, as well as the ERS markers GRP78 and ATF6, were tested by Western blot, qPCR, immunohistochemistry, or immunofluorescence. EGB761 could decrease the Scr, BUN, 24 h Pro, and FBG levels in the DKD group, alleviate renal pathological injury, decrease urine ß 2-MG, RBP4 levels, and decrease the expression of α-SMA, collagen IV, fibronectin, and GRP78, as well as ATF6, while increase the expression of E-cadherin. These findings demonstrate that EGB761 can improve renal function, reduce tubular injury, and ameliorate ECM accumulation and EMT in DKD kidney tubules, and the mechanism may be related to the inhibition of ERS.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Estrés del Retículo Endoplásmico , Matriz Extracelular/metabolismo , Mesodermo/patología , Extractos Vegetales/uso terapéutico , Animales , Línea Celular , Línea Celular Transformada , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Ginkgo biloba , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/lesiones , Túbulos Renales/fisiopatología , Túbulos Renales/ultraestructura , Masculino , Mesodermo/efectos de los fármacos , Ratones Endogámicos C57BL , Extractos Vegetales/farmacología , Proteínas Plasmáticas de Unión al Retinol/orina , Microglobulina beta-2/orina
8.
Oxid Med Cell Longev ; 2021: 8869085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33574986

RESUMEN

Endothelial-to-mesenchymal transition (EndMT), which is involved in the development of various cardiovascular diseases, is induced by dyslipidemia or obesity. In dyslipidemia, the increased levels of oxidized low-density lipoproteins (oxLDL) upregulated the lectin-type oxidized LDL receptor 1 (Lox-1), which then upregulated the down signaling pathways of PKC-α/MMPs/TGF-ß/SMAD2 or 3 and increased the EndMT. In this study, we investigated the effect of pyrogallol-phloroglucinol-6,6-bieckol (PPB), which is a compound of Ecklonia cava (E. cava), on decreased blood pressure (BP) by attenuating the EndMT in a high-fat diet- (HFD-) fed animal model. We also investigated PPB's attenuation effect on EndMT in oxLDL-treated mouse endothelial cells as an in vitro model. The results indicated that, in the aorta or endothelial cells of mice, the HFD or oxLDL treatment significantly increased the expression of Lox-1/PKC-α/MMP9/TGF-ß/SMAD2/SMAD3. The PPB treatment significantly decreased its expression. In contrast, the HFD or oxLDL treatment significantly decreased the expression of the EC markers (PECAM-1 and vWF) while the PPB treatment significantly increased them. Moreover, the HFD or oxLDL treatment significantly increased the expression of the mesenchymal cell markers (α-SMA and vimentin) while PPB treatment significantly decreased them. PPB decreased the intima-media thickness and extracellular matrix amount of the aorta and attenuated the BP, which was increased by the HFD. In conclusion, PPB attenuated the upregulation of Lox-1/PKC-α/MMP9/TGF-ß/SMAD2 and 3 and restored the EndMT in HFD-fed animals. Moreover, PPB showed a restoring effect on HFD-induced hypertension.


Asunto(s)
Aorta/patología , Benzofuranos/uso terapéutico , Dieta Alta en Grasa , Endotelio Vascular/patología , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Mesodermo/patología , Taninos/uso terapéutico , Animales , Aorta/efectos de los fármacos , Aorta/fisiopatología , Benzofuranos/administración & dosificación , Benzofuranos/farmacología , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Grosor Intima-Media Carotídeo , Dislipidemias/complicaciones , Dislipidemias/fisiopatología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Hipertensión/complicaciones , Hipertensión/fisiopatología , Lipoproteínas LDL , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Mesodermo/efectos de los fármacos , Mesodermo/fisiopatología , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Proteína Quinasa C-alfa/metabolismo , Receptores Depuradores de Clase E/metabolismo , Proteínas Smad/metabolismo , Taninos/administración & dosificación , Taninos/farmacología , Factor de Crecimiento Transformador beta/metabolismo
9.
Cell Mol Life Sci ; 78(5): 2199-2212, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32910224

RESUMEN

It is critical to specify a signal that directly drives the transition that occurs between cell states. However, such inferences are often confounded by indirect intercellular communications or secondary transcriptomic changes due to primary transcription factors. Although FGF is known for its importance during mesoderm-to-endothelium differentiation, its specific role and signaling mechanisms are still unclear due to the confounding factors referenced above. Here, we attempted to minimize the secondary artifacts by manipulating FGF and its downstream mediators with a short incubation time before sampling and protein-synthesis blockage in a low-density angioblastic/endothelial differentiation system. In less than 8 h, FGF started the conversion of KDRlow/PDGFRAlow nascent mesoderm into KDRhigh/PDGFRAlow angioblasts, and the priming by FGF was necessary to endow endothelial formation 72 h later. Further, the angioblastic conversion was mediated by the FGFR1/BRAF/MEK/ERK pathway in mesodermal cells. Finally, two transcription factors, ETV2 and LMO2, were the early direct functional responders downstream of the FGF pathway, and ETV2 alone was enough to complement the absence of FGF. FGF's selective role in mediating the first-step, angioblastic conversion from mesoderm-to-endothelium thus allows for refined control over acquiring and manipulating angioblasts. The noise-minimized differentiation/analysis platform presented here is well-suited for studies on the signaling switches of other mesodermal-lineage fates as well.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas con Dominio LIM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción/metabolismo , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Nat Commun ; 11(1): 5640, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159078

RESUMEN

Lymphangioleiomyomatosis (LAM) is a rare fatal cystic lung disease due to bi-allelic inactivating mutations in tuberous sclerosis complex (TSC1/TSC2) genes coding for suppressors of the mechanistic target of rapamycin complex 1 (mTORC1). The origin of LAM cells is still unknown. Here, we profile a LAM lung compared to an age- and sex-matched healthy control lung as a hypothesis-generating approach to identify cell subtypes that are specific to LAM. Our single-cell RNA sequencing (scRNA-seq) analysis reveals novel mesenchymal and transitional alveolar epithelial states unique to LAM lung. This analysis identifies a mesenchymal cell hub coordinating the LAM disease phenotype. Mesenchymal-restricted deletion of Tsc2 in the mouse lung produces a mTORC1-driven pulmonary phenotype, with a progressive disruption of alveolar structure, a decline in pulmonary function, increase of rapamycin-sensitive expression of WNT ligands, and profound female-specific changes in mesenchymal and epithelial lung cell gene expression. Genetic inactivation of WNT signaling reverses age-dependent changes of mTORC1-driven lung phenotype, but WNT activation alone in lung mesenchyme is not sufficient for the development of mouse LAM-like phenotype. The alterations in gene expression are driven by distinctive crosstalk between mesenchymal and epithelial subsets of cells observed in mesenchymal Tsc2-deficient lungs. This study identifies sex- and age-specific gene changes in the mTORC1-activated lung mesenchyme and establishes the importance of the WNT signaling pathway in the mTORC1-driven lung phenotype.


Asunto(s)
Pulmón/metabolismo , Linfangioleiomiomatosis/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mesodermo/metabolismo , Factores de Edad , Anciano , Animales , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/fisiopatología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Mesodermo/efectos de los fármacos , Ratones , Factores Sexuales , Sirolimus/administración & dosificación , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Vía de Señalización Wnt
11.
J Mol Cell Cardiol ; 148: 50-62, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32889002

RESUMEN

Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by pulmonary arterial vasoconstriction and remodeling. Src family tyrosine kinases, including Fyn, play critical roles in vascular remodeling via the inhibition of STAT3 signaling. EPA is known to inhibit Fyn kinase activity. This study investigated the therapeutic potential and underlying mechanisms of EPA and its metabolite, resolvin E1 (RvE1), to treat PAH using monocrotaline-induced PAH model rats (MCT-PAH), human pulmonary artery endothelial cells (HPAECs), and human pulmonary artery smooth muscle cells (HPASMCs). Administration of EPA 1 and 2 weeks after MCT injection both ameliorated right ventricular hypertrophy, remodeling and dysfunction, and medial wall thickening of the pulmonary arteries and prolonged survival in MCT-PAH rats. EPA attenuated the enhanced contractile response to 5-hydroxytryptamine in isolated pulmonary arteries of MCT-PAH rats. Mechanistically, the treatment with EPA and RvE1 or the introduction of dominant-negative Fyn prevented TGF-ß2-induced endothelial-to-mesenchymal transition and IL-6-induced phosphorylation of STAT3 in cultured HPAECs. EPA and RvE1 suppressed Src family kinases' activity as evaluated by their phosphorylation status in cultured HPAECs and HPASMCs. EPA and RvE1 suppressed vasocontraction of rat and human PA. Furthermore, EPA and RvE1 inhibited the enhanced proliferation and activity of Src family kinases in HPASMCs derived from patients with idiopathic PAH. EPA ameliorated PAH's pathophysiology by mitigating vascular remodeling and vasoconstriction, probably inhibiting Src family kinases, especially Fyn. Thus, EPA is considered a potent therapeutic agent for the treatment of PAH.


Asunto(s)
Ácido Eicosapentaenoico/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/enzimología , Proteínas Proto-Oncogénicas c-fyn/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Humanos , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/fisiopatología , Interleucina-6/farmacología , Masculino , Mesodermo/efectos de los fármacos , Mesodermo/patología , Mesodermo/fisiopatología , Monocrotalina , Contracción Miocárdica/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Análisis de Supervivencia , Factor de Crecimiento Transformador beta2/farmacología , Vasodilatación/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Familia-src Quinasas/metabolismo
12.
Nat Commun ; 11(1): 3978, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32770044

RESUMEN

Methionine restriction, a dietary regimen that protects against metabolic diseases and aging, represses cancer growth and improves cancer therapy. However, the response of different cancer cells to this nutritional manipulation is highly variable, and the molecular determinants of this heterogeneity remain poorly understood. Here we report that hepatocyte nuclear factor 4α (HNF4α) dictates the sensitivity of liver cancer to methionine restriction. We show that hepatic sulfur amino acid (SAA) metabolism is under transcriptional control of HNF4α. Knocking down HNF4α or SAA enzymes in HNF4α-positive epithelial liver cancer lines impairs SAA metabolism, increases resistance to methionine restriction or sorafenib, promotes epithelial-mesenchymal transition, and induces cell migration. Conversely, genetic or metabolic restoration of the transsulfuration pathway in SAA metabolism significantly alleviates the outcomes induced by HNF4α deficiency in liver cancer cells. Our study identifies HNF4α as a regulator of hepatic SAA metabolism that regulates the sensitivity of liver cancer to methionine restriction.


Asunto(s)
Factor Nuclear 4 del Hepatocito/metabolismo , Neoplasias Hepáticas/metabolismo , Metionina/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Cisteína/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Nuclear 4 del Hepatocito/genética , Hígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Mesodermo/efectos de los fármacos , Mesodermo/patología , Redes y Vías Metabólicas/efectos de los fármacos , Metaboloma/efectos de los fármacos , Ratones , Sorafenib/farmacología , Transcripción Genética/efectos de los fármacos
13.
Endocrinol Metab (Seoul) ; 35(2): 384-395, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32615723

RESUMEN

BACKGROUND: Endothelial-to-mesenchymal transition (EndMT) contributes to inflammatory conditions inducing conversion of endothelial cells (ECs) into activated fibroblasts, promoting fibrotic diseases. Pro-inflammatory cytokine is the most potent inducer of EndMT. We investigated inhibition of interleukin-1ß (IL-1ß)-induced EndMT by gemigliptin, a dipeptidyl peptidase-IV inhibitor. METHODS: We exposed human umbilical vein endothelial cells (HUVECs) to 10 ng/mL IL-1ß/20 µM gemigliptin and analyzed the expression of endothelial, smooth muscle, mesenchymal, and osteoblastic markers, bone morphogenetic protein (BMP), Smad, and non-Smad signaling pathway proteins. RESULTS: Morphological changes showed gemigliptin blocked IL-1ß-induced EndMT, upregulated EC markers, and downregulated smooth muscle and mesenchymal markers. IL-1ß activation of HUVECs is initiated by the BMP/Smad and non-smad BMP signaling pathways. Gemigliptin inhibited IL-1ß induction of BMP2 and 7, activin receptor type IA, BMP receptor type IA, and BMP receptor type II. Reversal of IL-1ß-mediated inhibition of BMP-induced Smad1/5/8, Smad2, and Smad3 phosphorylation by gemigliptin suggests involvement of the Smad pathway in gemigliptin action. In the non-Smad BMP pathway, gemigliptin treatment significantly increased the deactivation of extracellular regulated protein kinase (ERK), p38, and JNK by IL-1ß. Gemigliptin treatment suppressed BMP-2-induced expression of key osteoblastic markers including osterix, runt-related transcription factor 2, and hepcidin during IL-1ß-induced EndMT. CONCLUSION: We demonstrated a novel protective mechanism of gemigliptin against fibrosis by suppressing IL-1ß-induced EndMT.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Transición Epitelial-Mesenquimal , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Interleucina-1beta/farmacología , Mesodermo/efectos de los fármacos , Piperidonas/farmacología , Pirimidinas/farmacología , Proteínas Morfogenéticas Óseas/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Transducción de Señal
14.
Cell Death Dis ; 11(7): 549, 2020 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-32683412

RESUMEN

Endothelial-mesenchymal transition (EndMT) is considered one of the processes underlying tissue fibrosis by contributing to the pool of myofibroblasts. In the present study, we investigated the epigenetic mechanism whereby angiotensin II (Ang II) regulates EndMT to promote cardiac fibrosis focusing on the role of chromatin remodeling protein BRG1. BRG1 knockdown or inhibition attenuated Ang II-induced EndMT, as evidenced by down-regulation of CDH5, an endothelial marker, and up-regulation of COL1A2, a mesenchymal marker, in cultured vascular endothelial cells. On the one hand, BRG1 interacted with and was recruited by Sp1 to the SNAI2 (encoding SLUG) promoter to activate SNAI2 transcription in response to Ang II stimulation. Once activated, SLUG bound to the CDH5 promoter to repress CDH5 transcription. On the other hand, BRG1 interacted with and was recruited by SRF to the COL1A2 promoter to activate COL1A2 transcription. Mechanistically, BRG1 evicted histones from the target promoters to facilitate the bindings of Sp1 and SRF. Finally, endothelial conditional BRG1 knockout mice (CKO) exhibited a reduction in cardiac fibrosis, compared to the wild type (WT) littermates, in response to chronic Ang II infusion. In conclusion, our data demonstrate that BRG1 is a key transcriptional coordinator programming Ang II-induced EndMT to contribute to cardiac fibrosis.


Asunto(s)
Angiotensina II/farmacología , Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Células Endoteliales/metabolismo , Mesodermo/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , ADN Helicasas/deficiencia , Células Endoteliales/efectos de los fármacos , Fibrosis , Histonas/metabolismo , Humanos , Mesodermo/efectos de los fármacos , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Proteínas Nucleares/deficiencia , Unión Proteica/efectos de los fármacos , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/deficiencia , Transcripción Genética/efectos de los fármacos
15.
Eur J Nutr ; 59(2): 517-527, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30725211

RESUMEN

PURPOSE: Endothelial-to-mesenchymal transition (EndMT) plays an important role in pathogenesis of a number of inflammatory diseases. Hydroxytyrosol (HT) and, particularly, its major plasma metabolite HT-3O sulfate (HT-3Os) are known olive oil antioxidant and anti-inflammatory polyphenols which exert benefits against vascular diseases by improving endothelial function. However, to date the HT-3Os role in EndMT is not well known. METHODS: To investigate the HT-3Os effects on EndMT in the inflamed endothelium, we used an in vitro model of endothelial dysfunction, challenging endothelial cells (EC), human umbilical EC (HUVEC) and human retinal EC (HREC) with Interleukin-1ß (IL-1ß), an inflammatory agent. HREC were used as a specific model to investigate HT-3Os effects on vascular retinal diseases. RESULTS: We found that IL-1ß treatment-induced EndMT phenotype in both cell models, also changing cell morphology. HT-3Os protected EC against IL-1ß effects, recovering cell morphology and phenotype. Mechanistically, HT-3Os targeting fibroblast growth factor receptor 1 FGFR1 expression and let-7 miRNA, controlled transforming growth factor beta (TGF-ß) signalling in EC, downregulating transcription factors expression (SNAI1 and ZEB2) and gene expression of late EndMT markers (FN1, VIM, NOTCH3, CNN1, MMP2 and MMP9). CONCLUSION: These results demonstrate that HT-3Os blunts pathological EndMT in inflamed EC, maintaining high let-7 miRNA expression and preventing activation of TGF-ß signalling.


Asunto(s)
Endotelio/efectos de los fármacos , Endotelio/fisiopatología , Inflamación/fisiopatología , Mesodermo/efectos de los fármacos , Mesodermo/fisiopatología , Alcohol Feniletílico/análogos & derivados , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Células Cultivadas , Técnicas In Vitro , Alcohol Feniletílico/farmacología , Sulfatos
16.
Biochem Biophys Res Commun ; 522(4): 838-844, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-31801667

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. The authors found that the experiment to show that HG decreased CD31 (Figure 1 E) and increased Vimentin (Figure 1 F) mRNA level could not be repeated. Moreover, they found that HG had no effect on Ch25h protein level and cannot repeat the experiments that HG decreased Ch25h mRNA level (Figure 1 G). In summary, they cannot conclude that HG suppressed EndoMT through KLF4/Ch25h, which made great weakening to the protection of KLF4/Ch25h axis in blood glucose dysfunction-induced endothelial dysfunction and diabetes. The authors wish to apologize for any inconvenience they may have caused.


Asunto(s)
Endotelio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Mesodermo/metabolismo , Metformina/farmacología , Esteroide Hidroxilasas/metabolismo , Animales , Endotelio/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Glucosa/toxicidad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Factor 4 Similar a Kruppel , Receptores X del Hígado/metabolismo , Masculino , Mesodermo/efectos de los fármacos , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
17.
J Biosci Bioeng ; 129(3): 371-378, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31615734

RESUMEN

We optimized the conditions for the differentiation of human induced pluripotent stem cells (hiPSCs) into mesoderm lineage-committed cells by supplementing the cultures with CHIR, a selective GSK-3 inhibitor, during embryoid body (EB) formation. In vitro treatment with 4 µM CHIR during the late 2 days of a 4-day suspension culture period was most effective at promoting mesodermal differentiation. The resulting EBs showed a significant increase in the expression levels of mesoderm-associated genes (WNT3A, T, DKK1, GATA4, FOXC1, and MESP1) and a maintenance of OCT3/4 and NANOG expressions. Upon subsequent differentiation into a cardiac cell lineage, these EBs were shown to generate contractile cardiomyocytes. When shortening the CHIR treatment period to 1 day, the resulting EBs showed reduced expression of mesoderm-associated genes in comparison to the 2-day CHIR treatment. In particular, the expression level of FOXC1 in the 1-day CHIR-treated EBs was much lower than that of the 2-day CHIR-treated EBs. When the treatment period with CHIR was extended to 4 days, the resulting EBs presented significantly reduced expression of WNT3A, OCT3/4, and NANOG upon CHIR concentrations above 4 µM. Similarly, when CHIR treatment was conducted after the formation of EBs, the effectiveness of the GSK-3 inhibitor was reduced compared to a treatment performed during EB formation. Our results indicate that spatiotemporal constraints associated with EB formation, i.e., three-dimensional structuration and cell development in EBs, should be taken into account when designing EB formation-based differentiation protocol involving CHIR treatment.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cuerpos Embrioides/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mesodermo/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Animales , Línea Celular , Linaje de la Célula/efectos de los fármacos , Cuerpos Embrioides/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Mesodermo/citología , Miocitos Cardíacos/citología
18.
Clin Exp Dent Res ; 5(5): 541-550, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31687189

RESUMEN

Obejective: To investigate the effect of increasing Strontium (Sr) concentrations on the growth and osteogenic behavior of human bone marrow stromal cells (BMSCs) from mesenchymal (i.e., fibula) and ectomesenchymal (i.e., mandible) embryonic origins. Materials and methods: Fibula and mandible BMSCs were cultured in media without (Ctrl) or with Sr in four diverse concentrations: Sr1, 11.3 × 10-3 mg/L, human seric physiological level; Sr2, 13 mg/L, human seric level after strontium ranelate treatment; Sr3, 130 mg/L, and Sr4, 360 mg/L. Proliferation rate (1, 3, and 7 days), osteogenic behavior (alkaline phosphatase [ALP] activity, 7 and 14 days; expression of osteogenic genes (ALP, osteopontin, and osteocalcin at 7, 14, and 21 days), and formation of mineralized nodules (14 and 21 days) of the BMSCs were assessed. Data was compared group- and period-wise using analysis of variance tests. Results: Fibula and mandible BMSCs cultured with Sr4 showed increased proliferation rate, and osteocalcin and osteopontin gene expression together with more evident formation of mineralized nodules, compared all other Sr concentrations. For both cell populations, Sr4 led to lower ALP activity, and ALP gene expression, compared with the other Sr concentrations. Conclusion: BMSCs from mesenchymal (i.e., fibula) and ectomesenchymal (i.e., mandible) embryonic origins showed increased cellular proliferation and osteogenic behavior when cultured with Sr4, in vitro.


Asunto(s)
Calcificación Fisiológica , Células Madre Mesenquimatosas/citología , Mesodermo/citología , Osteogénesis , Estroncio/farmacología , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Persona de Mediana Edad , Osteocalcina/genética , Osteocalcina/metabolismo , Osteopontina/genética , Osteopontina/metabolismo
19.
PLoS Biol ; 17(10): e3000498, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31613879

RESUMEN

During gastrulation, the pluripotent epiblast self-organizes into the 3 germ layers-endoderm, mesoderm and ectoderm, which eventually form the entire embryo. Decades of research in the mouse embryo have revealed that a signaling cascade involving the Bone Morphogenic Protein (BMP), WNT, and NODAL pathways is necessary for gastrulation. In vivo, WNT and NODAL ligands are expressed near the site of gastrulation in the posterior of the embryo, and knockout of these ligands leads to a failure to gastrulate. These data have led to the prevailing view that a signaling gradient in WNT and NODAL underlies patterning during gastrulation; however, the activities of these pathways in space and time have never been directly observed. In this study, we quantify BMP, WNT, and NODAL signaling dynamics in an in vitro model of human gastrulation. Our data suggest that BMP signaling initiates waves of WNT and NODAL signaling activity that move toward the colony center at a constant rate. Using a simple mathematical model, we show that this wave-like behavior is inconsistent with a reaction-diffusion-based Turing system, indicating that there is no stable signaling gradient of WNT/NODAL. Instead, the final signaling state is homogeneous, and spatial differences arise only from boundary effects. We further show that the durations of WNT and NODAL signaling control mesoderm differentiation, while the duration of BMP signaling controls differentiation of CDX2-positive extra-embryonic cells. The identity of these extra-embryonic cells has been controversial, and we use RNA sequencing (RNA-seq) to obtain their transcriptomes and show that they closely resemble human trophoblast cells in vivo. The domain of BMP signaling is identical to the domain of differentiation of these trophoblast-like cells; however, neither WNT nor NODAL forms a spatial pattern that maps directly to the mesodermal region, suggesting that mesoderm differentiation is controlled dynamically by the combinatorial effect of multiple signals. We synthesize our data into a mathematical model that accurately recapitulates signaling dynamics and predicts cell fate patterning upon chemical and physical perturbations. Taken together, our study shows that the dynamics of signaling events in the BMP, WNT, and NODAL cascade in the absence of a stable signaling gradient control fate patterning of human gastruloids.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Gastrulación/genética , Mesodermo/metabolismo , Proteína Nodal/genética , Transducción de Señal , Proteínas Wnt/genética , Benzotiazoles/farmacología , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 4/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Gástrula/citología , Gástrula/efectos de los fármacos , Gástrula/metabolismo , Gastrulación/efectos de los fármacos , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Modelos Biológicos , Modelos Estadísticos , Proteína Nodal/deficiencia , Organoides/citología , Organoides/efectos de los fármacos , Organoides/metabolismo , Proteínas Wnt/metabolismo
20.
Tissue Eng Regen Med ; 16(5): 501-512, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31624705

RESUMEN

Background: Chronic kidney disease is a severe threat to human health with no ideal treatment strategy. Mature mammalian kidneys have a fixed number of nephrons, and regeneration is difficult once they are damaged. For this reason, developing an efficient approach to achieve kidney regeneration is necessary. The technology of the combination of decellularized kidney scaffolds with stem cells has emerged as a new strategy; however, in previous studies, the differentiation of stem cells in decellularized scaffolds was insufficient for functional kidney regeneration, and many problems remain. Methods: We used 0.5% sodium dodecyl sulfate (SDS) to produce rat kidney decellularized scaffolds, and induce adipose-derived stem cells (ADSCs) into intermediate mesoderm by adding Wnt agonist CHIR99021 and FGF9 in vitro. The characteristics of decellularized scaffolds and intermediate mesoderm induced from adipose-derived stem cells were identified. The scaffolds were recellularized with ADSCs and intermediate mesoderm cells through the renal artery and ureter. After cocultured for 10 days, cells adhesion and differentiation was evaluated. Results: Intermediate mesoderm cells were successfully induced from ADSCs and identified by immunofluorescence and Western blotting assays (OSR1 + , PAX2 +). Immunofluorescence showed that intermediate mesoderm cells differentiated into tubular-like (E-CAD + , GATA3 +) and podocyte-like (WT1 +) cells with higher differentiation efficiency than ADSCs in the decellularized scaffolds. Comparatively, this phenomenon was not observed in induced intermediate mesoderm cells cultured in vitro. Conclusion: In this study, we demonstrated that intermediate mesoderm cells could be induced from ADSCs and that they could differentiate well after cocultured with decellularized scaffolds.


Asunto(s)
Riñón/citología , Mesodermo/citología , Ingeniería de Tejidos/métodos , Animales , Electroforesis en Gel de Poliacrilamida , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Riñón/efectos de los fármacos , Mesodermo/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...