Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38761998

RESUMEN

BACKGROUND: Previous study implied that local M2 polarization of macrophage promoted mucosal edema and exacerbates Th2 type inflammation in chronic rhinosinusitis with nasal polyps (CRSwNP). However, the specific pathogenic role of M2 macrophages and the intrinsic regulators in the development of CRS remains elusive. OBJECTIVE: We thought to investigate the regulatory role of SIRT5 in the polarization of M2 macrophages and its potential contribution to the development of CRSwNP. METHODS: RT-qPCR and Western blot analyses were performed to examine the expression levels of SIRT5 and markers of M2 macrophages in sinonasal mucosa samples obtained from both CRS and control groups. Wild-type and Sirt5 knockout mice were used to establish nasal polyp model with Th2 inflammation and investigate the effects of SIRT5 in macrophages on disease development. Furthermore, in vitro experiments were conducted to elucidate the regulatory role of SIRT5 in polarization of M2 macrophages. RESULTS: Clinical investigations showed that SIRT5 was highly expressed and positively correlated with M2 macrophages markers in eosinophilic polyps. The expression of SIRT5 in M2 macrophages was found to contribute to the development of the disease, which was impaired in Sirt5 deficiency mice. Mechanistically, SIRT5 was shown to enhance the alternative polarization of macrophages through promoting glutaminolysis. CONCLUSIONS: SIRT5 plays a crucial role in promoting the development of CRSwNP by supporting the alternative polarization of macrophage and thus provides a potential target for CRSwNP interventions.

2.
Am J Physiol Cell Physiol ; 318(5): C943-C953, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32267717

RESUMEN

During pregnancy, placental vascular growth, which is essential for supporting the rapidly growing fetus, is associated with marked elevations in blood flow. These vascular changes take place under chronic physiological low O2 (less than 2-8% O2 in human; chronic physiological normoxia, CPN) throughout pregnancy. O2 level below CPN pertinent to the placenta results in placental hypoxia. Such hypoxia can cause severe endothelial dysfunction, which is associated with adverse pregnancy outcomes (e.g., preeclampsia) and high risk of adult-onset cardiovascular diseases in children born to these pregnancy complications. However, our current knowledge about the mechanisms underlying fetoplacental endothelial function is derived primarily from cell models established under atmospheric O2 (~21% O2 at sea level, hyperoxia). Recent evidence has shown that fetoplacental endothelial cells cultured under CPN have distinct gene expression profiles and cellular responses compared with cells cultured under chronic hyperoxia. These data indicate the critical roles of CPN in programming fetal endothelial function and prompt us to re-examine the mechanisms governing fetoplacental endothelial function under CPN. Better understanding these mechanisms will facilitate us to develop preventive and therapeutic strategies for endothelial dysfunction-associated diseases (e.g., preeclampsia). This review will provide a brief summary on the impacts of CPN on endothelial function and its underlying mechanisms with a focus on fetoplacental endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Oxígeno/metabolismo , Placenta/irrigación sanguínea , Preeclampsia/genética , Movimiento Celular/genética , Células Endoteliales/patología , Femenino , Feto , Humanos , Hiperoxia/genética , Hiperoxia/metabolismo , Hiperoxia/patología , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/patología , Placenta/metabolismo , Placenta/fisiopatología , Preeclampsia/fisiopatología , Embarazo
3.
Biochem Biophys Res Commun ; 522(1): 61-67, 2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31740004

RESUMEN

Allergic rhinitis (AR) is a worldwide highly prevalent nasal inflammatory disease with elusive mechanisms about the regulation of innate immune response. The roles and mechanisms of NLRP3, a typical inflammasome, in AR development remain unclear. Here we investigate the roles of NLRP3 inflammasome activation in the development and progression of AR and try to uncover its potential mechanisms underlying. Wildtype and NLRP3 knockout mice were applied to construct the ovalbumin (OVA)-induced AR model. Caspase-1 specific inhibitor Belnacasan and inflammasome activator ATP were used for adjuvant stimulation of AR-model mice respectively. We found that the production of IL-1ß and the activation of inflammasome were increased in both patients and mice with AR. NLRP3 deficiency markedly suppressed AR progression with reduced inflammatory response and epithelium pyroptosis in mice with AR. Furthermore, Caspase-1 inhibitor treatment in vivo ameliorated the development and progression of AR with favorable outcomes. Mechanistically, inflammation augments and nasal mucosa injury during AR were partially due to ASC-specks accumulation and subsequent cell pyroptosis. Our study reveals the previously unknown roles of NLRP3 inflammasome in promoting the development and progression of AR via enhancing inflammatory response and epithelium pyroptosis and thus provides a potential clue for allergic disease interventions.


Asunto(s)
Epitelio/patología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Rinitis Alérgica/patología , Adolescente , Adulto , Anciano , Animales , Línea Celular , Progresión de la Enfermedad , Epitelio/metabolismo , Femenino , Humanos , Inmunoglobulina E/sangre , Inflamación , Interleucina-1beta/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Ovalbúmina/metabolismo , Rinitis Alérgica/metabolismo , Adulto Joven
4.
Hypertension ; 74(1): 154-163, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31154903

RESUMEN

Preeclampsia impairs fetoplacental vascular function and increases risks of adult-onset cardiovascular disorders in children born to preeclamptic mothers, implicating that preeclampsia programs fetal vasculature in utero. However, the underlying mechanisms remain elusive. We hypothesize that preeclampsia alters fetal endothelial gene expression and disturbs cytokines- and growth factors-induced endothelial responses. RNA sequencing analysis was performed on unpassaged human umbilical vein endothelial cells (HUVECs) from normotensive and preeclamptic pregnancies. Functional assays for endothelial monolayer integrity, proliferation, and migration were conducted on passage 1 HUVECs from normotensive and preeclamptic pregnancies. Compared with normotensive cells, 926 and 172 genes were dysregulated in unpassaged female and male HUVECs from preeclamptic pregnancies, respectively. Many of these preeclampsia-dysregulated genes are associated with cardiovascular diseases (eg, heart failure) and endothelial function (eg, cell migration, calcium signaling, and endothelial nitric oxide synthase signaling). TNF (tumor necrosis factor)-α-, TGF (transforming growth factor)-ß1-, FGF (fibroblast growth factor)-2-, and VEGFA (vascular endothelial growth factor A)-regulated gene networks were differentially disrupted in unpassaged female and male HUVECs from preeclamptic pregnancies. Moreover, preeclampsia decreased endothelial monolayer integrity in responses to TNF-α in both female and male HUVECs. Preeclampsia decreased TGF-ß1-strengthened monolayer integrity in female HUVECs, whereas it enhanced FGF-2-strengthened monolayer integrity in male HUVECs. Preeclampsia promoted TNF-α-, TGF-ß1-, and VEGFA-induced cell proliferation in female, but not in male HUVECs. Preeclampsia inhibited TNF-α-induced cell migration in female HUVECs, but had an opposite effect on male HUVECs. In conclusion, preeclampsia differentially dysregulates cardiovascular diseases- and endothelial function-associated genes/pathways in female and male fetal endothelial cells in association with the sexual dimorphisms of preeclampsia-dysregulated fetal endothelial function.


Asunto(s)
Preeclampsia/genética , Caracteres Sexuales , Transcriptoma/genética , Factor A de Crecimiento Endotelial Vascular/genética , Células Cultivadas , Bases de Datos Factuales , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Preeclampsia/fisiopatología , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Sensibilidad y Especificidad , Transducción de Señal/genética , Estadísticas no Paramétricas , Regulación hacia Arriba
5.
Biochem Biophys Res Commun ; 513(3): 635-641, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30981508

RESUMEN

Clinical data show that part of patients with sinonasal diseases suffered from olfactory dysfunction, especially with allergic rhinitis (AR) and chronic rhinosinusitis (CRS). However, the mechanisms responsible for AR-induced olfactory loss are still largely unknown. Because of the difficulty to obtain human olfactory mucosa, an AR-induced olfactory loss animal model needs to be constructed to clarify the mechanism. The AR mouse model was induced by intraperitoneal sensitizing with ovalbumin (OVA) followed by intranasal challenge lasted from 1 to 12 weeks. For groups with recovery, mice were housed for another 4-week long without any treatment after the last intranasal challenge. Olfactory function, olfactory receptor neurons (ORNs) density and leukocytes infiltration were examined at different time points. Olfactory loss occurs immediately after 1-week intranasal challenge and deteriorates almost to anosmia after 8th week, and after that olfactory loss become irreversible. Nasal inflammation induces significant infiltration of leukocytes into olfactory epithelium (OE), which negatively correlated with the density of ORNs and mouse olfaction in a time dependent manner. The neutrophilic subtype dominates in number amongst the total infiltrated leukocytes, indicating its pivotal role in nasal inflammation-induced olfactory dysfunction. In this study, we constructed a persistent AR-induced olfactory loss mouse model, losing the ability to recover from dysfunction if the disease duration more than eight weeks, which implies that timely treatments are necessary. Meanwhile, this mouse model could provide an easy and reliable system to clarify the mechanisms of AR-induced irreversible olfactory dysfunction.


Asunto(s)
Trastornos del Olfato/etiología , Rinitis Alérgica/complicaciones , Animales , Modelos Animales de Enfermedad , Leucocitos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Trastornos del Olfato/fisiopatología , Neuronas Receptoras Olfatorias/patología , Ovalbúmina , Rinitis Alérgica/inducido químicamente , Rinitis Alérgica/fisiopatología , Olfato
6.
J Cell Physiol ; 234(7): 10184-10195, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30387149

RESUMEN

During pregnancy, a tremendous increase in fetoplacental angiogenesis is associated with elevated blood flow. Aberrant fetoplacental vascular function may lead to pregnancy complications including pre-eclampsia. Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA) are crucial regulators of fetoplacental endothelial function. G protein α subunit 14 (GNA14), a member of Gαq/11 subfamily is involved in mediating hypertensive diseases and tumor vascularization. However, little is known about roles of GNA14 in mediating the FGF2- and VEGFA-induced fetoplacental endothelial function. Using human umbilical vein endothelial cells (HUVECs) cultured under physiological chronic low oxygen (3% O2 ) as a cell model, we show that transfecting cells with adenovirus carrying GNA14 complementary DNA (cDNA; Ad-GNA14) increases (p < 0.05) protein expression of GNA14. GNA14 overexpression blocks (p < 0.05) FGF2-stimulated endothelial migration, whereas it enhances (p < 0.05) endothelial monolayer integrity (maximum increase of ~35% over the control at 24 hr) in response to FGF2. In contrast, GNA14 overexpression does not significantly alter VEGFA-stimulated cell migration, VEGFA-weakened cell monolayer integrity, and intracellular Ca++ mobilization in response to adenosine triphosphate (ATP), FGF2, and VEGFA. GNA14 overexpression does not alter either FGF2- or VEGFA-induced phosphorylation of ERK1/2. However, GNA14 overexpression time-dependently elevates (p < 0.05) phosphorylation of phospholipase C-ß3 (PLCß3) at S1105 in response to FGF2, but not VEGFA. These data suggest that GNA14 distinctively mediates fetoplacental endothelial cell migration and permeability in response to FGF2 and VEGFA, possibly in part by altering activation of PLCß3 under physiological chronic low oxygen.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/fisiología , Placenta/irrigación sanguínea , Permeabilidad Capilar/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Humanos , Embarazo
7.
J Physiol ; 596(12): 2333-2344, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29659033

RESUMEN

KEY POINTS: Fetoplacental vascular growth is critical to fetal growth. Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA) are two major regulators of fetoplacental vascular growth. G protein α subunit 11 (GNA11) transmits signals from many external stimuli to the cellular interior and may mediate endothelial function. It is not known whether GNA11 mediates FGF2- and VEGFA-induced endothelial cell responses under physiological chronic low O2 . In the present study, we show that knockdown of GNA11 significantly decreases FGF2- and VEGFA-induced fetoplacental endothelial cell migration but not proliferation and permeability. Such decreases in endothelial migration are associated with increased phosphorylation of phospholipase C-ß3. The results of the present study suggest differential roles of GNA11 with respect to mediating FGF2- and VEGFA-induced fetoplacental endothelial function. ABSTRACT: During pregnancy, fetoplacental angiogenesis is dramatically increased in association with rapidly elevated blood flow. Any disruption of fetoplacental angiogenesis may lead to pregnancy complications such as intrauterine growth restriction. Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor A (VEGFA) are crucial regulators of fetoplacental angiogenesis. G protein α subunits q (GNAq) and 11 (GNA11) are two members of the Gαq/11 subfamily involved in mediating vascular growth and basal blood pressure. However, little is known about the roles of GNA11 alone with respect to mediating the FGF2- and VEGFA-induced fetoplacental endothelial function. Using a cell model of human umbilical cord vein endothelial cells cultured under physiological chronic low O2 (3% O2 ), we showed that GNA11 small interfering RNA (siRNA) dramatically inhibited (P < 0.05) FGF2- and VEGFA-stimulated fetoplacental endothelial migration (by ∼36% and ∼50%, respectively) but not proliferation and permeability. GNA11 siRNA also elevated (P < 0.05) FGF2- and VEGFA-induced phosphorylation of phospholipase C-ß3 (PLCß3) at S537 in a time-dependent fashion but not mitogen-activated protein kinase 3/1 (ERK1/2) and v-akt murine thymoma viral oncogene homologue 1 (AKT1). These data suggest that GNA11 mediates FGF2- and VEGFA-stimulated fetoplacental endothelial cell migration partially via altering the activation of PLCß3.


Asunto(s)
Movimiento Celular , Proliferación Celular , Feto/fisiología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Fosfolipasa C beta/metabolismo , Placenta/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Femenino , Feto/citología , Subunidades alfa de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP/genética , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Placenta/citología , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Cordón Umbilical/citología , Cordón Umbilical/fisiología
8.
J Inequal Appl ; 2018(1): 43, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29497263

RESUMEN

A concept of the sub-band operator of multi-band wavelets is introduced, the theory of d-circular matrices is developed and the upper bound and the lower bound of the norm of the sub-band operator are obtained. Examples are provided to illustrate the results proposed in this paper.

9.
Exp Lung Res ; 43(8): 283-292, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29140133

RESUMEN

AIM: Pulmonary arterial hypertension (PAH), a deadly disorder is associated with excessive growth of human pulmonary artery endothelial (HPAECs) and smooth muscle (HPASMCs) cells. Current therapies primarily aim at promoting vasodilation, which only ameliorates clinical symptoms without a cure. 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE) is an endogenous aryl hydrocarbon receptor (AhR) ligand, and mediates many cellular function including cell growth. However, the roles of ITE in human lung endothelial cells remain elusive. Herein, we tested a hypothesis that ITE inhibits growth of human pulmonary artery endothelial cells via AhR. MATERIALS AND METHODS: Immunohistochemistry was performed to localize AhR expression in human lung tissues. The crystal violet method and MTT assay were used to determine ITE's effects on growth of HPAECs. The AhR activation in HPAECs was confirmed using Western blotting and RT-qPCR. The role of AhR in ITE-affected proliferation of HPAECs was assessed using siRNA knockdown method followed by the crystal violet method. RESULTS: Immunohistochemistry revealed that AhR was present in human lung tissues, primarily in endothelial and smooth muscle cells of pulmonary veins and arteries, as well as in bronchial and alveolar sac epithelia. We also found that ITE dose- and time-dependently inhibited proliferation of HPAECs with a maximum inhibition of 83% at 20 µM after 6 days of treatment. ITE rapidly decreased AhR protein levels, while it increased mRNA levels of cytochrome P450 (CYP), family 1, member A1 (CYP1A1) and B1 (CYP1B1), indicating activation of the AhR/CYP1A1 and AhR/CYP1B1 pathways in HPAECs. The AhR siRNA significantly suppressed AhR protein expression, whereas it did not significantly alter ITE-inhibited growth of HPAECs. CONCLUSIONS: ITE suppresses growth of HPAECs independent of AhR, suggesting that ITE may play an important role in preventing excessive growth of lung endothelial cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Indoles/farmacología , Arteria Pulmonar/citología , Tiazoles/farmacología , Células Cultivadas , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Células Endoteliales/citología , Humanos , Receptores de Hidrocarburo de Aril/análisis , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Tiempo
10.
J Inequal Appl ; 2017(1): 222, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28979084

RESUMEN

A class of 4-band symmetric biorthogonal wavelet bases has been constructed, in which any wavelet system the high-pass filters can be determined by exchanging position and changing the sign of the two low-pass filters. Thus, the least restrictive conditions are needed for forming a wavelet so that the free degrees can be reversed for application requirement. Some concrete examples with high vanishing moments are also given, the properties of the transformation matrix are studied and the optimal model is constructed. These wavelets can process the boundary conveniently, and they lead to highly efficient computations in applications.

11.
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
12.
Mol Med Rep ; 16(6): 9095-9101, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29039558

RESUMEN

Drug resistance is an obstacle to effective treatment of ovarian cancer. There have been substantial evidences supporting the association between diabetes and the sensitivity to chemotherapy. Insulin (INS) is believed to be the strongest, most lasting hypoglycemic drug. Therefore, the present study aimed to elucidate whether insulin could facilitate the anti­proliferative activities of cisplatin (cis­diamminedichloroplatinum, DDP) in the A2780 ovarian cancer cell line. The inhibitory effects of DPP with/without INS on the growth of A2780 cells was measured by MTT assay. The cell cycle stages and levels of apoptosis were determined by flow cytometry. The amounts of signaling elements involved in the regulation of were examined using western blotting and reverse transcription­quantitative polymerase chain reaction analysis. The results indicated that INS pre­treatment enhanced the inhibitory effect of DDP on the proliferation of A2780 cells, and facilitated the apoptosis induced by DDP. INS­DDP treatment led to a marked decrease in the percentage of G0/G1 phase cells, but a corresponding increase in the proportion of S phase cells. Furthermore, A2780 cells pretreated with INS followed by DDP upregulated the protein expression level of phosphorylated c­Jun N­terminal kinase (JNK), which resulted in a substantial increase in the expression levels of p53 mRNA and protein, compared with DDP administration alone. In conclusion, the combination of INS and DDP facilitated the apoptosis of A2780 cells, which may be associated with the activation of the JNK signaling pathway and consequently the involvement of p53 at both mRNA and protein expression levels. These results may be useful in furthering our understanding of the mechanisms involved in the chemotherapeutic treatment of ovarian cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Insulina/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/patología , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
13.
J Clin Endocrinol Metab ; 102(9): 3470-3479, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911139

RESUMEN

Context: Preeclampsia is a leading cause of fetal and maternal morbidity and mortality during pregnancy. Although the etiology of preeclampsia is unknown, preeclampsia offspring have increased risks of developing cardiovascular disorders in adulthood, implicating that preeclampsia programs fetal vasculature in utero. Objective: We hypothesize that preeclampsia alters expression profiles of endothelial microRNAs (miRNAs) in fetal endothelial cells and disturbs the vascular endothelial growth factor A (VEGFA)- and fibroblast growth factor 2 (FGF2)-induced endothelial function. Design and Setting: Unpassaged (P0) human umbilical vein endothelial cells (HUVECs) were isolated immediately after cesarean-section delivery from normotensive (NT) and preeclamptic (PE) pregnancies. Differentially expressed miRNAs between P0-HUVECs from NT and PE pregnancies were identified using a miRNA polymerase chain reaction (PCR) array and confirmed using reverse transcription quantitative PCR. To determine the function of these differentially expressed miRNAs, miRNAs of interest were knocked down in NT-HUVECs following by cell functional assays. Results: Sixteen miRNAs, including miR-29a/c-3p, were downregulated in P0-HUVECs from the PE group compared with the NT group. Bioinformatics analysis predicted the PI3K-v-akt murine thymoma viral oncogene homolog 1 (AKT) signaling pathway was dysregulated in P0-HUVECs from the PE group, which was associated with the miR-29a/c-3p downregulation. We further demonstrated that miR-29a/c-3p knockdown inhibited the VEGFA- and FGF2-induced endothelial migration as well as FGF2-induced AKT1 phosphorylation in HUVECs. However, miR-29a/c-3p knockdown did not alter the extracellular signal-regulated kinase 1/2 phosphorylation, cell proliferation, and endothelial monolayer integrity in response to VEGFA and FGF2 in HUVECs. Conclusions: Preeclampsia-downregulated miR-29a/c-3p may impair fetal endothelial function by disturbing the FGF2-activated PI3K-AKT signaling pathway, hence inhibiting endothelial cell migration.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Preeclampsia/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética , Adulto , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo , Endotelio Vascular/fisiología , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Preeclampsia/fisiopatología , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sensibilidad y Especificidad , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Toxicology ; 336: 70-8, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26275813

RESUMEN

Placental angiogenesis is dramatically increased during pregnancy in association with the elevated placental blood flows to support the rapidly growing fetus. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental toxicant and a ligand of aryl hydrocarbon receptor (AhR). Herein, we investigated the effects of TCDD on proliferation, migration, and viability of fetoplacental endothelial cells in response to a complete growth medium which contained serum and growth supplement using human umbilical cord vein (HUVECs) and artery (HUAECs) cells as models. We found that TCDD dose- and time-dependently inhibited (p < 0.05) proliferation of HUVECs and HUAECs. Treatment with TCDD at 10 nM for 6 days inhibited (p < 0.05) migration (by ∼ 30%) of HUAECs, but not HUVECs. TCDD at 10nM also decreased (p < 0.05) viability of HUVECs and HUAECs. Interestingly, specific AhR siRNA blocked (p < 0.05) the TCDD-inhibited cellular responses in HUAECs, but not HUVECs. Nonetheless, TCDD at 10nM neither affected the cell cycle progression, nor did it induce cell apoptosis in HUVECs and HUAECs. In addition, TCDD at 10 nM also did not alter activation of ERK1/2 and AKT1 in HUVECs and HUAECs. Collectively, TCDD suppresses proliferation and/or migration (two key steps of angiogenesis) of HUVECs and HUAECs independent and dependent of AhR, respectively. These data suggest that TCDD inhibited growth of HUVECs and HUAECs via decreasing cell viability. Thus, TCDD may inhibit fetoplacental angiogenesis, leading to negative pregnancy outcomes.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Placenta/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Arterias Umbilicales/efectos de los fármacos , Venas Umbilicales/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Placenta/irrigación sanguínea , Embarazo , Arterias Umbilicales/citología , Venas Umbilicales/citología
15.
Med Hypotheses ; 84(5): 494-7, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25697115

RESUMEN

Preterm birth (PTB) is defined as birth before 37 weeks of gestation and is a leading cause of neonatal mortality and morbidity. To date, the etiology of spontaneous PTB (sPTB) remains unclear; however, intrauterine bacterial infection-induced inflammation is considered to be one of the major triggers. Aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor. Upon activation, AhR signaling mediates many biological processes. AhR is abundantly expressed in human placentas, primarily in trophoblasts, and several fetal organs and tissues. The activation of AhR signaling can modulate inflammatory responses via promoting production of pro-inflammatory cytokines by the placenta and fetal membranes. These cytokines could enhance expression and/or activity of cyclooxygenase-2 (COX2) in human trophoblasts and amniotic epithelia, which in turn stimulate synthesis and release of prostaglandins (PGs; e.g., PGE2 and PGF2α). Given the discovery of a number of natural and endogenous AhR ligands in human, we hypothesize that in a subset of patients with high AhR expression in placentas and fetal membranes, repeated exposure to these AhR ligands hyperactivates AhR, inducing hyperactivation of the cytokines/COX2/PGs pathway, resulting in myometrial contractions, ultimately leading to sPTB. We further hypothesize that hyperactivation of this AhR pathway can induce sPTB either directly or in synergy with the bacterial infection. Proof of this hypothesis may provide a novel mechanism underlying sPTB.


Asunto(s)
Modelos Biológicos , Placenta/metabolismo , Nacimiento Prematuro/etiología , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/fisiología , Ciclooxigenasa 2/metabolismo , Femenino , Humanos , Embarazo , Prostaglandinas/metabolismo
16.
J Histochem Cytochem ; 63(1): 22-31, 2015 01.
Artículo en Inglés | MEDLINE | ID: mdl-25362142

RESUMEN

Preeclampsia is characterized by maternal endothelial dysfunction (e.g., increased maternal vascular permeability caused by the disassembly of endothelial junction proteins). However, it is unclear if preeclampsia is associated with impaired vascular growth and expression of endothelial junction proteins in human placentas. Herein, we examined vascular growth in placentas from women with normal term (NT) and preeclamptic (PE) pregnancies using two endothelial junction proteins as endothelial markers: CD31 and vascular endothelial-cadherin (VE-Cad). We also compared protein and mRNA expression of CD31 and VE-Cad between NT and PE placentas, and determined the alternatively spliced expression of CD31 using PCR. We found that CD31 and VE-Cad were immunolocalized predominantly in villous endothelial cells. However, capillary number density (total capillary number per unit villous area) and capillary area density (total capillary lumen area per unit villous area) as well as CD31 and VE-Cad protein and mRNA levels were similar between NT and PE placentas. PCR in combination with sequence analysis revealed a single, full-length CD31, suggesting that there are no alternatively spliced isoform of CD31 expressed in placentas. These data indicate that preeclampsia does not significantly affect vascular growth or the expression of endothelial junction proteins in human placentas.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Regulación de la Expresión Génica , Placenta/irrigación sanguínea , Placenta/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Preeclampsia/metabolismo , Empalme Alternativo , Antígenos CD/genética , Cadherinas/genética , Capilares/crecimiento & desarrollo , Femenino , Humanos , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Preeclampsia/genética , Preeclampsia/patología , Preeclampsia/fisiopatología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo
17.
J Histochem Cytochem ; 62(5): 347-54, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24423937

RESUMEN

G-proteins mediate cellular function upon interaction with G-protein coupled receptors. Of the 16 mammalian G-protein α subunits identified, G-protein subunit α-11 (GNA11) and -14 (GNA14) have been implicated in modulating hypertension and endothelial function. However, little is known about their expression and roles in human placentas. Here, we examined GNA11 and GNA14 protein expression in first trimester (FT), normal term (NT), and severe preeclamptic (sPE) human placentas as well as in NT human umbilical cords. We found that GNA11 and GNA14 were immunolocalized primarily in trophoblasts, villous stromal cells, and endothelial cells in placentas as well as in endothelial and/or smooth muscle cells of the umbilical cord artery and vein. Western blotting revealed that the GNA14, but not GNA11, protein levels were increased (2.5-2.9 fold; p<0.01) in sPE vs. NT placentas. GNA11 protein was detected only in NT, but not FT, placentas, whereas GNA14 protein levels were increased (7.7-10.6 fold; p<0.01) in NT vs. FT placentas. Thus, GNA11 and GNA14 may mediate the function of several cell types in placentas. Moreover, the high expression of GNA14 in sPE placentas may also imply its importance in sPE pregnancies as in the other hypertension-related disorders.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Anticuerpos/inmunología , Western Blotting , Femenino , Subunidades alfa de la Proteína de Unión al GTP/inmunología , Humanos , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...