Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Gynecol Endocrinol ; 37(3): 269-272, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33480311

RESUMEN

OBJECTIVE: Endothelial dysfunction and denudation are considered a first step in atherosclerosis. Endothelial proliferation is key for cellular repair. The effect of bazedoxifene on the vascular endothelium has not been explored. We investigated the effect of bazedoxifene on endothelial cell proliferation. METHODS: Primary cultures from human umbilical artery endothelial cells were used in dose-response experiments (0.1, 1.0, and 10.0 EC50 dose) with bazedoxifene, estradiol, raloxifene and a combination of bazedoxifene and estradiol. Proliferation was assessed with the XTT colorimetric cell-proliferation assay. The possible participation of cyclins A, B, D1 and p27Kip1 was analyzed by the measurement of their expression at both the protein and the gene levels. RESULTS: A significant increase of similar size for cell proliferation was obtained with bazedoxifene, estradiol and raloxifene, but no significant change was observed for the association of bazedoxifene and estradiol. The impact was detected at the first 0.1 EC50 dose and was not dose-dependent. Estradiol achieved a significant increase in the protein expression of cyclin A and p27Kip1, but no change was detected for the other compounds at either the gene or protein level. CONCLUSION: Bazedoxifene demonstrated a proliferative effect of similar size to estradiol in cultured human umbilical artery endothelial cells. The molecular mechanisms need further investigation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Indoles/farmacología , Proliferación Celular/genética , Células Cultivadas , Ciclina A/genética , Ciclina A/metabolismo , Ciclina B/genética , Ciclina B/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endoteliales/fisiología , Endotelio Vascular/citología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Recién Nacido , Embarazo , Arterias Umbilicales/citología
2.
Am J Physiol Endocrinol Metab ; 320(3): E453-E466, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427050

RESUMEN

Progesterone acts directly on vascular smooth muscle cells (VSMCs) through activation of membrane progesterone receptor α (mPRα)-dependent signaling to rapidly decrease cytosolic Ca2+ concentrations and induce muscle relaxation. However, it is not known whether this progesterone action involves uptake of Ca2+ by the sarco/endoplasmic reticulum (SR) and increased sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. The present results show that treatment of cultured human VSMCs with progesterone and the selective mPR agonist Org OD-02-0 (OD 02-0) but not with the nuclear PR agonist R5020 increased SERCA protein expression, which was blocked by knockdown of mPRα with siRNA. Moreover, treatments with progesterone and OD 02-0, but not with R5020, increased phospholamban (PLB) phosphorylation, which would result in disinhibition of SERCA function. Progesterone and OD 02-0 significantly increased Ca2+ levels in the SR and caused VSMC relaxation. These effects were blocked by pretreatment with cyclopiazonic acid (CPA), a SERCA inhibitor, and by knockdown of SERCA2 with siRNA, suggesting that SERCA2 plays a critical role in progesterone induction of VSMC relaxation. Treatment with inhibitors of inhibitory G proteins (Gi, NF023), MAP kinase (AZD 6244), Akt/Pi3k (wortmannin), and a Rho activator (calpeptin) blocked the progesterone- and OD 02-0-induced increase in Ca2+ levels in the SR and SERCA expressions. These results suggest that the rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.NEW & NOTEWORTHY The rapid effects of progesterone on cytosolic Ca2+ levels and relaxation of VSMCs through mPRα involve regulation of the functions of SERCA2 and PLB through Gi, MAP kinase, and Akt signaling pathways and downregulation of RhoA activity.


Asunto(s)
Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Progesterona/farmacología , Receptores de Progesterona/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Relajación Muscular/genética , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Arterias Umbilicales/citología , Arterias Umbilicales/efectos de los fármacos , Arterias Umbilicales/metabolismo
3.
Exp Biol Med (Maywood) ; 246(1): 84-96, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33167688

RESUMEN

Acute myeloid leukemia (AML) is a malignant clonal disease derived from hematopoietic stem/progenitor cell. Leukemia blasts cause extensive hypoxia of bone marrow (BM), which lead to disorder and remodeling of BM niche, thereby becoming "leukemic niche" to support the development and drug-resistance of AML as well as the maintenance of normal hematopoietic stem cells. In this study, the biological characteristics (such as self-renewal, apoptosis, migration, autocrine) and function (vascularization) of mesenchymal stem cells (MSCs) and human umbilical artery endothelial cells (HUAECs) that make up BM arteriolar niche in simulated hypoxia AML context were investigated. It was found that moderate hypoxia enhanced the viability of the arteriolar niche cells, but severe hypoxia of AML BM resulted in the damage of arteriolar niche cells and the disorder of vascular cytokines C-X-C motif chemokine ligand 6 (CXCL6). The dynamic changes of CXCL6 in the system as well as its anti-apoptotic and promoting angiogenic effects suggested that CXCL6 played an important role in the remodeling of BM arteriolar niche in AML. Taking advantage of CXCL6 can save the damaged MSCs and HUAECs, which is the hope of rescuing arteriolar niche. It is suggested that CXCL6 may be an assistant strategy for microenvironment targeted therapy of AML.


Asunto(s)
Arteriolas/metabolismo , Quimiocina CXCL6/metabolismo , Leucemia Mieloide Aguda/metabolismo , Nicho de Células Madre , Remodelación Vascular , Apoptosis , Médula Ósea/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Autorrenovación de las Células , Supervivencia Celular , Citocinas/genética , Regulación Leucémica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Células Madre Mesenquimatosas/metabolismo , Neovascularización Patológica/patología , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral , Arterias Umbilicales/citología , Regulación hacia Arriba/genética
4.
Biochem Biophys Res Commun ; 520(2): 304-310, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31601422

RESUMEN

Pericytes are mural cells that cover small blood vessels. While defects in pericyte coverage are known to be involved in various vessel related pathologies, including diabetic retinopathy, the molecular mechanisms underlying pericyte coverage are not fully understood. In this study, we investigated the contribution of the forkhead transcription factor FOXO1 in endothelial cells to pericyte coverage in the developing retina. We observed retinal pericytes in tamoxifen-inducible endothelium-specific Foxo1 deletion mice. Tamoxifen was injected at postnatal day 1-3 and the retinas were harvested at P21. Our results demonstrated that Foxo1 deletion in the endothelium affected arteriole pericyte morphology without altering pericyte number, proliferation, and apoptosis. We hypothesized that abnormal pericyte morphogenesis in the knockout retina was caused by impaired pericyte differentiation. FOXO1 silencing by siRNA in the primary artery endothelium further revealed that THBS1 (thrombospondin 1), which promotes pericyte differentiation via TGFß activation, was reduced in the FOXO1-deficient endothelium. Immunohistochemistry of FOXO1 knockout mice showed reduced numbers of phospho-Smad3+ arteriole pericytes compared with wild-type mice. In addition, endothelium-pericyte co-culture analysis revealed that pericytes cultured with FOXO1-deficient endothelial cells failed to differentiate sufficiently; this failure was partially rescued by the addition of recombinant THBS1 to the supernatant. The findings suggest that endothelial FOXO1 contributes to pericyte differentiation via regulation of THBS1 expression. This study provides new insights into the molecular mechanism of pericyte coverage in the context of endothelium-derived regulation and highlights a new therapeutic target for pericyte-related pathology.


Asunto(s)
Proteína Forkhead Box O1/genética , Pericitos/patología , Retina/citología , Retina/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Diferenciación Celular , Células Cultivadas , Células Endoteliales , Proteína Forkhead Box O1/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/efectos de los fármacos , Tamoxifeno/farmacología , Trombospondinas/metabolismo , Factor de Crecimiento Transformador beta , Arterias Umbilicales/citología
5.
Int. j. morphol ; 37(1): 111-117, 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-990014

RESUMEN

SUMMARY: Arterial obstruction in small diameter (<6 mm) vessels are many times treated with grafts, however autologous aren't always available and synthetic have a high rate of complications. Decellularization of umbilical arteries may provide a solution, but the ideal method is debatable. We compare effectiveness between SDS and Triton X-100. Umbilical cords obtained from full term pregnancies with normal development and no evident complications in the newborn, were micro-dissected within 12 h and stored in phosphate buffered saline without freezing. Arteries were then processed for decellularization using 0.1 % and 1 % SDS, and 1 % Triton X100 protocols. Evaluation of cellular and nuclear material, collagen fibers, elastic fibers, and glycosoaminoglycans of the extracellular matrix (ECM) were evaluated as well as morphometric analysis under histological and immunohistochemical techniques. Triton X-100 was ineffective, preserving nuclear remains identified by immunofluorescence, had the most notable damage to elastic fibers, and decrease in collagen. SDS effectively eliminated the nuclei and had a less decrease in elastic fibers and collagen. Laminin was preserved in all groups. No significant differences were identified in luminal diameters; however the middle layer decreased due to decellularization of muscle cells. In conclusion, 0.1 % SDS decellularization was the most effective in eliminating cells and preserving the main components of the ECM.


RESUMEN: La obstrucción arterial en vasos de pequeño diámetro (<6 mm) se trata muchas veces con injertos, sin embargo, los autólogos no siempre están disponibles y los sintéticos tienen una alta tasa de complicaciones. La descelularización de las arterias umbilicales puede proporcionar una solución, pero el método ideal es discutible. Comparamos la efectividad entre los métodos SDS y Triton X-100. Cordones umbilicales obtenidos a partir de embarazos a término con evolución normal y sin complicaciones evidentes del recién nacido, se microdiseccionaron en 12 horas y se almacenaron en solución salina con fosfato sin congelación. Las arterias se procesaron luego para la descelularización usando los protocolos de SDS al 0,1 % y 1 %, y Triton X-100 al 1 %. Se realizó la evaluación de material celular y nuclear, fibras de colágeno, fibras elásticas y glucosoaminoglicanos de la matriz extracelular (MEC), así como el análisis morfométrico bajo técnicas histológicas e inmunohistoquímicas. Triton X-100 fue ineficaz, conservando los restos nucleares identificados por inmunofluorescencia, tuvo el daño más notable a las fibras elásticas y la disminución del colágeno. SDS efectivamente eliminó los núcleos y tuvo una disminución menor en las fibras elásticas y el colágeno. Laminina fue preservado en todos los grupos. No se identificaron diferencias significativas en los diámetros luminales; sin embargo, la capa media disminuyó debido a la descelularización de las células musculares. la descelularización con SDS al 0,1 % fue la más efectiva para eliminar células y preservar los principales componentes de la MEC.


Asunto(s)
Humanos , Arterias Umbilicales/citología , Arterias Umbilicales/metabolismo , Ingeniería de Tejidos/métodos , Matriz Extracelular/metabolismo , Arterias Umbilicales/trasplante , Cordón Umbilical , Inmunohistoquímica , Separación Celular , Técnica del Anticuerpo Fluorescente , Colágeno , Injerto Vascular
6.
Am J Trop Med Hyg ; 99(6): 1451-1457, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30398136

RESUMEN

Dengue virus (DENV) infection of humans is presently the most important arthropod-borne viral global threat, for which no suitable or reliable animal model exists. Reports addressing the effect of DENV on vascular components other than endothelial cells are lacking. Dengue virus infection of vascular smooth muscle cells, which play a physiological compensatory response to hypotension in arteries and arterioles, has not been characterized, thus precluding our understanding of the role of these vascular components in dengue pathogenesis. Therefore, we studied the permissiveness of primary human umbilical artery smooth muscle cells (HUASMC) to DENV 1-4 infection and compared with the infection in the previously reported primary human umbilical vein endothelial cells (HUVEC) and the classically used, non-transformed, and highly permissive Lilly Laboratories Cell-Monkey Kidney 2 cells. Our results show that HUASMC are susceptible and productive to infection with the four DENV serotypes, although to a lesser extent when compared with the other cell lines. This is the first report of DENV permissiveness in human smooth muscle cells, which might represent an unexplored pathophysiological contributor to the vascular collapse observed in severe human dengue infection.


Asunto(s)
Virus del Dengue/fisiología , Células Epiteliales/virología , Células Endoteliales de la Vena Umbilical Humana/virología , Miocitos del Músculo Liso/virología , Replicación Viral , Animales , Línea Celular , Virus del Dengue/clasificación , Células Epiteliales/citología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Riñón/citología , Riñón/virología , Macaca mulatta , Miocitos del Músculo Liso/citología , Cultivo Primario de Células , Serogrupo , Arterias Umbilicales/citología , Arterias Umbilicales/virología , Carga Viral , Ensayo de Placa Viral
7.
Hum Immunol ; 79(12): 855-860, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30213613

RESUMEN

Worldwide, there is a great need of small diameter vascular grafts that can be used in human disorders such as cardiovascular and peripheral vascular disease. Until now, severe adverse reactions are caused from the use of synthetic or animal derived grafts, while the use of autologous vessels is restricted only in a small number of patients. The limited availability of the vessels might be resolved by the use of HLA-matched vascular grafts utilizing the decellularized human umbilical arteries. In this study, human umbilical arteries were decellularized and then repopulated with Mesenchymal Stem Cells. The HLA-genotype of the repopulated grafts, analyzed by Next Generation Sequencing technology, indicated their successful production. The HLA-matched vascular grafts could be generated efficiently and might be used in personalized medicine.


Asunto(s)
Antígenos HLA/inmunología , Prueba de Histocompatibilidad/métodos , Arterias Umbilicales/trasplante , Injerto Vascular/métodos , Femenino , Genotipo , Antígenos HLA/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Células Madre Mesenquimatosas/citología , Arterias Umbilicales/citología
8.
PLoS One ; 12(11): e0188530, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29166412

RESUMEN

Macrophages are a main component of atherosclerotic plaques. Recent studies suggest that pro-inflammatory M1 macrophages are pro-atherogenic while M2 macrophages promote plaque stability. Moreover, toll-like receptor signalling pathways are implicated in atherosclerotic plaque formation, evolution and regression. We propose microRNAs as key regulators of these processes. In this context, our goal is to promote inflammation resolution using miR-195 to reduce M1-like macrophage polarization and to evaluate the molecular mechanisms underlying such effect, as well as to explore the functional consequences for smooth muscle cell recruitment. Human primary macrophages were differentiated from peripheral blood monocytes and stimulated with LPS or IL-10 to promote M1 or M2c polarization, respectively. miR-195 levels were upregulated in M2c macrophages compared with M1 macrophages. In THP-1 macrophages stimulated with LPS and IFN-γ, results show that TLR2 levels were reduced by miR-195 overexpression compared with scrambled control. In addition, phosphorylated forms of p54 JNK, p46 JNK and p38 MAPK were decreased by miR-195 in macrophages following M1 stimulation. Moreover, miR-195 significantly decreased levels of IL-1ß, IL-6 and TNF-α pro-inflammatory cytokines in the supernatants of M1-stimulated macrophage cultures. At the functional level, results from smooth muscle cell recruitment and migration models showed that miR-195 impairs the capacity of M1 macrophages to promote smooth muscle cells migration. In conclusion, miR-195 is involved in macrophage polarization and inhibits TLR2 inflammatory pathway mediators. Moreover, miR-195 impairs the effect of macrophages on smooth muscle cells recruitment capacity and migration profile. Thus, miR-195 might be used as a new potential tool to promote inflammation resolution in cardiovascular research.


Asunto(s)
Inflamación/genética , Inflamación/patología , Macrófagos/metabolismo , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-10/farmacología , Espacio Intracelular/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Arterias Umbilicales/citología
9.
Reprod Toxicol ; 74: 181-188, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28986273

RESUMEN

Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor is involved in regulation of many essential biological processes including vascular development and angiogenesis. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an AhR ligand, which regulates immune responses and cancer cell growth. However, the roles of the ITE/AhR pathway in mediating placental angiogenesis remains elusive. Here, we determined if ITE affected placental angiogenic responses via AhR in human umbilical vein (HUVECs) and artery endothelial (HUAECs) cells in vitro. We observed that ITE dose- and time-dependently inhibited proliferation and viability of HUAECs and HUVECs, whereas it inhibited migration of HUAECs, but not HUVECs. While AhR siRNA significantly suppressed AhR protein expression in HUVECs and HUAECs, it attenuated the ITE-inhibited angiogenic responses of HUAECs, but not HUVECs. Collectively, ITE suppressed angiogenic responses of HUAECs and HUVECs, dependent and independent of AhR, respectively. These data suggest that ITE may regulate placental angiogenesis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Indoles/toxicidad , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Hidrocarburo de Aril/fisiología , Tiazoles/toxicidad , Arterias Umbilicales/citología , Venas Umbilicales/citología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocromo P-450 CYP1A1/efectos de los fármacos , Citocromo P-450 CYP1B1/genética , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Neuropilina-1/genética , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
10.
Acta Histochem ; 119(6): 638-647, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28803749

RESUMEN

The blood-brain barrier (BBB), which saves the brain from toxic substances, is formed by endothelial cells. It is mainly composed of tight junction (TJ) proteins existing between endothelial cells. Estrogen is an important regulatory hormone of BBB permeability. It protects the BBB before menopause, but may increase BBB permeability with aging. In addition, nitric oxide modulates BBB permeability. Alcohol impairs the integrity of the BBB with oxidants and inflammatory mediators such as iNOS. We investigated the effects of estrogen on BBB integrity in an in vitro BBB model created with ERα-free HUVEC (human umbilical vein endothelial-like cells) to mimics the menopausal period. In vitro BBB model is created with HUVEC/C6 (rat glioma cells) co-culture. The effect of 17ß-estradiol on ethanol-induced BBB disruption and change/or increase of iNOS activity, which modulate BBB integrity, were evaluated. Inducibility and functionality of BBB were investigated using transendothelial electrical resistance (TEER) and the expression of proteins TJ proteins (occludin and claudin-1) and iNOS activity by immunostaining. Our results revealed that 17ß-estradiol treatment before and after ethanol decrease expression of occludin and claudin-1 and value of TEER which are BBB disrupt indicators. In addition, ethanol and 17ß-estradiol separately and pre- and post-ethanol 17ß-estradiol treatment increased iNOS expression. Thus our study suggests caution in the use of 17ß-estradiol after menopause because 17ß-estradiol at this time may both increase the inflammatory process as well as damage the BBB. We think that beneficial effects of 17ß-estradiol may be through ERα but it needs further studies.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Estradiol/farmacología , Animales , Células Cultivadas , Receptor alfa de Estrógeno/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Modelos Biológicos , Ratas , Arterias Umbilicales/citología
11.
J Steroid Biochem Mol Biol ; 174: 9-13, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28536085

RESUMEN

To further investigate the interaction between vitamin D system and estrogen-mimetic compounds in the human vasculature we studied the effect of the "less- calcemic" analog of 1,25(OH)2D3 (1,25D); JK 1624F2-2 (JKF) in the presence of selective estrogen modulator femarelle (F), the phytoestrogen daidzein (D) and estradiol-17b (E2) on 3[H] thymidine incorporation (DNA synthesis) and creatine kinase specific activity (CK) in human umbilical artery vascular smooth muscle cells (VSMC). F, D and E2, stimulated DNA synthesis at low concentrations, and inhibited it at high concentrations. All estrogen-related compounds increased CK dose- dependently. Daily treatment with JKF (1nM for 3days) resulted in decreased DNA synthesis, increased CK and up- regulation of the stimulation of DNA synthesis by low estrogen-related hormones whereas D- and E2- mediated inhibition of cell proliferation was abolished by JKF. In contrast, inhibition of cell proliferation by F could not be blocked by JKF. JKF also up-regulated the stimulatory effects on CK by F, E2 and D. VSMC expressed Estrogen Receptor (ER)a and ERb mRNA at a relative ratio of 2.7:1.0, respectively. JKF pretreatment increased ERa (∼50%) and decreased ERb (∼25%) expression. E2 did not affect ERs whereas both D and F up-regulated ERb (∼100%) and ERa (∼50%). Additionally, JKF increased the intracellular competitive binding of F (from ∼70 to ∼310%), of D (from ∼60 to ∼250%) and of E2 from (from∼70 to ∼320%). F reciprocally modulated the vitamin D system by up-regulating VDR- and 25 hydroxyy vitamin D 1-a hydroxylase (1OHase) mRNA expression (∼120%). F also stimulated 1OHase activity as indicated by an increase in the production of 1, 25D (∼250%). A similar increase was elicited by D (∼90%) but not by E2. In conclusion, F has unique effects on human VSMC in that it can sustain inhibition of cell growth even in the presence of the vitamin D analog JKF. That JKF increases ER expression and F increased the endogenous production of 1, 25D and VDR expression offer new opportunities to modulate VSMC growth. Whether or not these mutual effects of F and JKF can be exploited to promote vascular health, particularly in estrogen-deficient states (e.g., menopause) is under investigation.


Asunto(s)
Calcitriol/análogos & derivados , Moduladores de los Receptores de Estrógeno/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Extractos Vegetales/farmacología , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilasa/metabolismo , Calcitriol/farmacología , Células Cultivadas , Creatina Quinasa/metabolismo , ADN/metabolismo , Interacciones Farmacológicas , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Estrógenos/farmacología , Humanos , Isoflavonas/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/metabolismo , Receptores de Calcitriol/genética , Arterias Umbilicales/citología
12.
Sci Rep ; 7: 46152, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28393890

RESUMEN

Intercellular adhesion plays a major role in tissue development and homeostasis. Yet, technologies to measure mature cell-cell contacts are not available. We introduce a methodology based on fluidic probe force microscopy to assess cell-cell adhesion forces after formation of mature intercellular contacts in cell monolayers. With this method we quantify that L929 fibroblasts exhibit negligible cell-cell adhesion in monolayers whereas human endothelial cells from the umbilical artery (HUAECs) exert strong intercellular adhesion forces per cell. We use a new in vitro model based on the overexpression of Muscle Segment Homeobox 1 (MSX1) to induce Endothelial-to-Mesenchymal Transition (EndMT), a process involved in cardiovascular development and disease. We reveal how intercellular adhesion forces in monolayer decrease significantly at an early stage of EndMT and we show that cells undergo stiffening and flattening at this stage. This new biomechanical insight complements and expands the established standard biomolecular analyses. Our study thus introduces a novel tool for the assessment of mature intercellular adhesion forces in a physiological setting that will be of relevance to biological processes in developmental biology, tissue regeneration and diseases like cancer and fibrosis.


Asunto(s)
Comunicación Celular , Fenómenos Biomecánicos , Adhesión Celular , Forma de la Célula , Citoesqueleto/metabolismo , Células Endoteliales/citología , Células HEK293 , Humanos , Factor de Transcripción MSX1/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
13.
Int J Immunopathol Pharmacol ; 30(3): 227-237, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28134597

RESUMEN

Systemic inflammation is involved in vascular calcification and cardiovascular disease which is the leading cause of mortality in rheumatoid arthritis (RA). A high level of serum interleukin (IL)-6 plays a key role in local and systemic inflammation in RA. However, the underlying mechanisms remain unclear. We established a human umbilical artery smooth muscle cell (HUASMC) culturing method to investigate the possible role of IL-6 on vascular calcification. HUASMCs were obtained from umbilical arteries of healthy neonates. To detect calcification effects, HUASMCs were treated with (experimental group) or without (control group) recombinant human (rh) IL-6. The calcium deposition stain and calcium concentrations were measured, as well as the mRNA and protein levels of the regulating factor of osteogenic differentiation-bone morphogenetic protein (BMP) 2 and those calcifying related molecules including bone-specific alkaline phosphatase (BAP), osteoprotegerin (OPG), and osteopontin (OPN). Our study showed that rhIL-6 induced calcification of HUASMCs in a time- and dose-dependent manner, and upregulated expressions of BMP2, BAP, OPG, and OPN of HUASMCs. We then used the anti-BMP2 siRNA to knockdown the expression of endogenous BMP2 to confirm its role. HUASMCs were transfected with negative siRNA (control group) or the valid anti-BMP2 siRNA (experimental group) before they were treated with rhIL-6. Cells transfected with negative siRNA without IL-6 stimulating served as the blank group. The results showed that anti-BMP2 siRNA markedly decreased expressions of BMP2, BAP, OPG, and OPN, and also partly reduced the calcification of HUASMCs induced by rhIL-6. Collectively, according to our study, rhIL-6 could induce the extracellular calcification and osteogenic differentiation of human artery smooth muscle cells through upregulating endogenous BMP2 in vitro. This may be one of the underlying mechanisms of the overwhelming vascular calcification in RA.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Interleucina-6 , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Proteína Morfogenética Ósea 2/genética , Células Cultivadas , Humanos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Osteopontina/genética , Osteopontina/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes , Arterias Umbilicales/citología , Calcificación Vascular/inducido químicamente
14.
Nature ; 542(7639): 49-54, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024299

RESUMEN

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Linfangiogénesis , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
15.
Tissue Eng Part C Methods ; 23(1): 38-49, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27923320

RESUMEN

The response of blood vessels to physiological and pathological stimuli partly depends on the cross talk between endothelial cells (EC) lining the luminal side and smooth muscle cells (SMC) building the inner part of the vascular wall. Thus, the in vitro analysis of the pathophysiology of blood vessels requires coculture systems of EC and SMC. We have developed and validated a modified three-dimensional sandwich coculture (3D SW-CC) of EC and SMC using open µ-Slides with a thin glass bottom allowing direct imaging. The culture dish comprises an intermediate plate to minimize the meniscus resulting in homogenous cell distribution. Human umbilical artery SMC were sandwiched between coatings of rat tail collagen I. Following SMC quiescence, human umbilical vein EC were seeded on top of SMC and cultivated until confluence. By day 7, EC had formed a confluent monolayer and continuous vascular endothelial (VE)-cadherin-positive cell/cell contacts. Below, spindle-shaped SMC had formed parallel bundles and showed increased calponin expression compared to day 1. EC and SMC were interspaced by a matrix consisting of laminin, collagen IV, and perlecan. Basal messenger RNA (mRNA) expression levels of E-selectin, angiopoietin-1, calponin, and intercellular adhesion molecule 1 (ICAM-1) of the 3D SW-CC was comparable to that of a freshly isolated mouse inferior vena cava. Addition of tumor necrosis factor alpha (TNF α) to the 3D SW-CC induced E-selectin and ICAM-1 mRNA and protein induction, comparable to the EC and SMC monolayers. In contrast, the addition of activated platelets induced a significantly delayed but more pronounced activation in the 3D SW-CC compared to EC and SMC monolayers. Thus, this 3D SW-CC permits analyzing the cross talk between EC and SMC that mediate cellular quiescence as well as the response to complex activation signals.


Asunto(s)
Comunicación Celular , Endotelio Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arterias Umbilicales/metabolismo , Venas Umbilicales/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , División Celular , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/citología , Humanos , Modelos Biológicos , Miocitos del Músculo Liso/citología , Arterias Umbilicales/citología , Venas Umbilicales/citología
16.
Blood ; 127(11): 1426-37, 2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26834239

RESUMEN

The Gata2 transcription factor is a pivotal regulator of hematopoietic cell development and maintenance, highlighted by the fact that Gata2 haploinsufficiency has been identified as the cause of some familial cases of acute myelogenous leukemia/myelodysplastic syndrome and in MonoMac syndrome. Genetic deletion in mice has shown that Gata2 is pivotal to the embryonic generation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). It functions in the embryo during endothelial cell to hematopoietic cell transition to affect hematopoietic cluster, HPC, and HSC formation. Gata2 conditional deletion and overexpression studies show the importance of Gata2 levels in hematopoiesis, during all developmental stages. Although previous studies of cell populations phenotypically enriched in HPCs and HSCs show expression of Gata2, there has been no direct study of Gata2 expressing cells during normal hematopoiesis. In this study, we generate a Gata2Venus reporter mouse model with unperturbed Gata2 expression to examine the hematopoietic function and transcriptome of Gata2 expressing and nonexpressing cells. We show that all the HSCs are Gata2 expressing. However, not all HPCs in the aorta, vitelline and umbilical arteries, and fetal liver require or express Gata2. These Gata2-independent HPCs exhibit a different functional output and genetic program, including Ras and cyclic AMP response element-binding protein pathways and other Gata factors, compared with Gata2-dependent HPCs. Our results, indicating that Gata2 is of major importance in programming toward HSC fate but not in all cells with HPC fate, have implications for current reprogramming strategies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Animales , Aorta/citología , Aorta/embriología , Proteínas Bacterianas/análisis , Proteínas Bacterianas/genética , Linaje de la Célula , Células Cultivadas , Técnicas de Reprogramación Celular , Factor de Transcripción GATA2/deficiencia , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/fisiología , Genes Reporteros , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/clasificación , Células Madre Hematopoyéticas/fisiología , Hígado/citología , Hígado/embriología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transcriptoma , Transgenes , Arterias Umbilicales/citología , Arterias Umbilicales/embriología
17.
Sci Rep ; 6: 18762, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26727026

RESUMEN

To evaluate the clinical potential of high nitrogen nickel-free austenitic stainless steel (HNNF SS), we have compared the cellular and molecular responses of human umbilical artery smooth muscle cells (HUASMCs) to HNNF SS and 316L SS (nickel-containing austenitic 316L stainless steel). CCK-8 analysis and flow cytometric analysis were used to assess the cellular responses (proliferation, apoptosis, and cell cycle), and quantitative real-time PCR (qRT-PCR) was used to analyze the gene expression profiles of HUASMCs exposed to HNNF SS and 316L SS, respectively. CCK-8 analysis demonstrated that HUASMCs cultured on HNNF SS proliferated more slowly than those on 316L SS. Flow cytometric analysis revealed that HNNF SS could activate more cellular apoptosis. The qRT-PCR results showed that the genes regulating cell apoptosis and autophagy were up-regulated on HNNF SS. Thus, HNNF SS could reduce the HUASMC proliferation in comparison to 316L SS. The findings furnish valuable information for developing new biomedical materials for stent implantation.


Asunto(s)
Miocitos del Músculo Liso/fisiología , Níquel , Acero Inoxidable , Stents , Arterias Umbilicales/citología , Apoptosis , Biomarcadores , Adhesión Celular , Técnicas de Cultivo de Célula , Ciclo Celular , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Propiedades de Superficie , Transcriptoma
18.
Br J Pharmacol ; 173(3): 484-96, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26493539

RESUMEN

BACKGROUND AND PURPOSE: In the phase III clinical trial, RELAX-AHF, serelaxin caused rapid and long-lasting haemodynamic changes. However, the cellular mechanisms involved are unclear in humans. EXPERIMENTAL APPROACH: This study examined the effects of serelaxin in co-cultures of human primary endothelial cells (ECs) and smooth muscle cells (SMCs) on cAMP and cGMP signalling. KEY RESULTS: Stimulation of HUVECs or human coronary artery endothelial cells (HCAECs) with serelaxin, concentration-dependently increased cGMP accumulation in co-cultured SMCs to a greater extent than in monocultures of either cell type. This was not observed in human umbilical artery endothelial cells (HUAECs) that do not express the relaxin receptor, RXFP1. Treatment of ECs with l-N(G) -nitro arginine (NOARG; 30 µM, 30 min) inhibited serelaxin-mediated (30 nM) cGMP accumulation in HUVECs, HCAECs and co-cultured SMCs. In HCAECs, but not HUVECs, pre-incubation with indomethacin (30 µM, 30 min) also inhibited cGMP accumulation in SMCs. Pre-incubation of SMCs with the guanylate cyclase inhibitor ODQ (1 µM, 30 min) had no effect on serelaxin-mediated (30 nM) cGMP accumulation in HUVECs and HCAECs but inhibited cGMP accumulation in SMCs. Serelaxin stimulation of HCAECs, but not HUVECs, increased cAMP accumulation concentration-dependently in SMCs. Pre-incubation of HCAECs with indomethacin, but not l-NOARG, abolished cAMP accumulation in co-cultured SMCs, suggesting involvement of prostanoids. CONCLUSIONS AND IMPLICATIONS: In co-cultures, treatment of ECs with serelaxin caused marked cGMP accumulation in SMCs and with HCAEC also cAMP accumulation. Responses involved EC-derived NO and with HCAEC prostanoid production. Thus, serelaxin differentially modulates vascular tone in different vascular beds.


Asunto(s)
Vasos Coronarios/citología , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Relaxina/farmacología , Arterias Umbilicales/citología , Venas Umbilicales/citología , Técnicas de Cocultivo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Humanos , Indometacina/farmacología , Nitroarginina/farmacología , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal
19.
Cell Signal ; 28(3): 109-116, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26689737

RESUMEN

AIMS: The role of different vascular subtypes of phosphodiesterases (PDE) in cGMP compartmentalization was evaluated in human smooth muscle cells. METHODS AND RESULTS: To understand how the cGMP conveys different information we infected smooth muscle cells with adenovirus containing mutants of the rat olfactory cyclic nucleotide-gated (CNG) channel-subunit and we recorded the associated cGMP-gated current (ICNG). The whole cell configuration of patch clamp technique was used to measure the ICNG and also the potassium current (IK) in human umbilical artery smooth muscle cells (HUASMC). ANP (0.1µM) induced a clear activation of basal ICNG, whereas SNP (100 µM) had a slight effect. The nonselective PDE inhibitor (IBMX; 100 µM), the PDE5 inhibitor (T0-156; 1 µM) and the PDE3 inhibitor (cilostamide; 10 µM), all had a tiny effects on the basal ICNG current. Concerning potassium channels, we observed that ANP and testosterone induced activation of IK and this activation is bigger than that elicited by SNP, cilostamide and T0-156. Cilostamide and T0-156 decreased the CNG stimulation induced by ANP and testosterone, suggesting that pGC pool is controlled by PDE3 and 5. Thus, the effects of SNP show the existence of two separated pools, one localized next to the plasma membrane and controlled by the PDE5 and PDE3, and a second pool localized in the cytosol of the cells that is regulated mainly by PDE3. CONCLUSIONS: Our results show the existence of cGMP compartmentalization in human vascular smooth muscle cells and this phenomenon can open new perspectives concerning the examination of PDE families as therapeutic targets.


Asunto(s)
GMP Cíclico/metabolismo , Músculo Liso Vascular/metabolismo , Adenoviridae/genética , Animales , Factor Natriurético Atrial/farmacología , Células Cultivadas , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Vectores Genéticos/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Microscopía Confocal , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Nitroprusiato/farmacología , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/metabolismo , Potasio/metabolismo , Ratas , Testosterona/farmacología , Arterias Umbilicales/citología
20.
Br J Pharmacol ; 172(4): 1005-19, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25297987

RESUMEN

BACKGROUND AND PURPOSE: In a recently conducted phase III clinical trial, RELAX-AHF, serelaxin infusion over 48 h improved short- and long-term clinical outcomes in patients with acute heart failure. In this study we used human primary cells from the umbilical vasculature to better understand the signalling mechanisms activated by serelaxin. EXPERIMENTAL APPROACH: We examined the acute effects of serelaxin on signal transduction mechanisms in primary human umbilical vascular cells and its chronic actions on markers of cardiovascular function and disease. KEY RESULTS: The RXFP1 receptor, the cognate serelaxin receptor, was expressed at the cell surface in HUVECs and human umbilical vein smooth muscle cells (HUVSMCs), human umbilical artery smooth muscle cells (HUASMCs) and human cardiac fibroblasts (HCFs), but not human umbilical artery endothelial cells. In HUVECs and HUVSMCs, serelaxin increased cAMP, cGMP accumulation and pERK1/2, and the concentration-response curves (CRCs) were bell-shaped. Similar bell-shaped CRCs for cGMP and pERK1/2 were observed in HCFs, whereas in HUASMCs, serelaxin increased cAMP, cGMP and pERK1/2 with sigmoidal CRCs. Gαi/o and lipid raft disruption, but not Gαs inhibition, altered the serelaxin CRC for cAMP and cGMP accumulation in HUVSMC but not HUASMC. Longer term serelaxin exposure increased the expression of neuronal NOS, VEGF, ETß receptors and MMPs (gelatinases) in RXFP1 receptor-expressing cells. CONCLUSIONS AND IMPLICATIONS: Serelaxin caused acute and chronic changes in human umbilical vascular cells that were cell background dependent. Bell-shaped CRCs that were observed only in venous cells and fibroblasts involved Gαi/o located within membrane lipid rafts.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Relaxina/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Receptor de Endotelina B/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Arterias Umbilicales/citología , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA