Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 237(2): 1440-1454, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34687046

RESUMEN

The bone microenvironment is one of the most hypoxic regions of the human body and in experimental models; hypoxia inhibits osteogenic differentiation of mesenchymal stromal cells (MSCs). Our previous work revealed that Mucin 1 (MUC1) was dynamically expressed during osteogenic differentiation of human MSCs and upregulated by hypoxia. Upon stimulation, its C-terminus (MUC1-CT) is proteolytically cleaved, translocases to the nucleus, and binds to promoters of target genes. Therefore, we assessed the MUC1-mediated effect of hypoxia on the proteomic composition of human osteoblast-derived extracellular matrices (ECMs) and characterized their osteogenic and angiogenic potentials in the produced ECMs. We generated ECMs from osteogenically differentiated human MSC cultured in vitro under 20% or 2% oxygen with or without GO-201, a MUC1-CT inhibitor. Hypoxia upregulated MUC1, vascular endothelial growth factor, and connective tissue growth factor independent of MUC1 inhibition, whereas GO-201 stabilized hypoxia-inducible factor 1-alpha. Hypoxia and/or MUC1-CT inhibition reduced osteogenic differentiation of human MSC by AMP-activated protein kinase/mTORC1/S6K pathway and dampened their matrix mineralization. Hypoxia modulated ECMs by transforming growth factor-beta/Smad and phosphorylation of NFκB and upregulated COL1A1, COL5A1, and COL5A3. The ECMs of hypoxic osteoblasts reduced MSC proliferation and accelerated their osteogenic differentiation, whereas MUC1-CT-inhibited ECMs counteracted these effects. In addition, ECMs generated under MUC1-CT inhibition reduced the angiogenic potential independent of oxygen concentration. We claim here that MUC1 is critical for hypoxia-mediated changes during osteoblastogenesis, which not only alters the proteomic landscape of the ECM but thereby also modulates its osteogenic and angiogenic potentials.


Asunto(s)
Mucina-1/metabolismo , Osteogénesis , Proteómica , Diferenciación Celular , Matriz Extracelular/metabolismo , Humanos , Hipoxia/metabolismo , Osteoblastos/metabolismo , Oxígeno/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Vasc Med ; 27(1): 3-12, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34570637

RESUMEN

Endothelium-derived nitric oxide (NO) is a mediator of angiogenesis. However, NO-mediated regulation of vasculogenesis remains largely unknown. In the present study, we show that the inhibition of NO significantly attenuated endothelial migration, ring formation, and tube formation. The contribution of nitric oxide synthase (NOS) enzymes during early vasculogenesis was assessed by evaluating endothelial NOS (eNOS) and inducible NOS (iNOS) mRNA expression during HH10-HH13 stages of chick embryo development. iNOS but not eNOS was expressed at HH12 and HH13 stages. We hypothesized that vasculogenic events are controlled by NOS-independent reduction of nitrite to NO under hypoxia during the very early phases of development. Semi-quantitative polymerase chain reaction analysis of hypoxia-inducible factor-1α (HIF-1α) showed higher expression at HH10 stage, after which a decrease was observed. This observation was in correlation with the nitrite reductase (NR) activity at HH10 stage. We observed a sodium nitrite-induced increase in NO levels at HH10, reaching a gradual decrease at HH13. The possible involvement of a HIF/NF-κB/iNOS signaling pathway in the process of early vasculogenesis is suggested by the inverse relationship observed between nitrite reduction and NOS activation between HH10 and HH13 stages. Further, we detected that NR-mediated NO production was inhibited by several NR inhibitors at the HH10 stage, whereas the inhibitors eventually became less effective at later stages. These findings suggest that the temporal dynamics of the NO source switches from NR to NOS in the extraembryonic area vasculosa, where both nitrite reduction and NOS activity are defined by hypoxia.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III , Óxido Nítrico , Animales , Embrión de Pollo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitritos , Transducción de Señal
3.
Cell Mol Biol (Noisy-le-grand) ; 68(4): 1-11, 2022 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-35988288

RESUMEN

Red blood cells (RBCs) carry large cholesterol fractions and imbalance in them leads to several vascular complications. RBCs band 3 protein plays an important role in maintaining membrane integrity and there are many reports on cholesterol and band 3 protein interaction. Yet, RBCs band 3 protein role in regulating cholesterol homeostasis needs to be investigated. In this study, we induced cholesterol-depletion and band 3 inhibition in RBCs; both of which cause stress by decreasing band 3 channel activity with an increase in RBCs adhesion to endothelial cells (EC) by elevating band 3 phosphorylation (Tyr21), methemoglobin level and decreasing nitric oxide level. We hypothesized that nitric oxide (NO), a prominent determinant for RBC structural stability, would protect RBCs from stressors. To estimate this, we used three NO donors (SpNO, Sildenafil citrate and 8-Bromo-cGMP) and found that all 3 NO donors were able to recover, with 8-Bromo-cGMP being the most effective as it not only increased band 3 channel activity but also decreased RBC-EC adhesiveness and methemoglobin level in both stressors. Whereas NO donor's treatment did not display an ameliorative impact when both stresses were combined. Overall, these findings may shed light on the role of 8-bromo-cGMP in regulating RBC cholesterol homeostasis by maintaining band 3 function. Further studies in this direction might help identify targets for the therapeutic use of NO donors in the treatment of blood disorders.


Asunto(s)
Donantes de Óxido Nítrico , Óxido Nítrico , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Colesterol , GMP Cíclico/análogos & derivados , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Metahemoglobina/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología
4.
Br J Clin Pharmacol ; 87(10): 3835-3850, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33609410

RESUMEN

AIMS: The SARS-coV-2 pandemic continues to cause an unprecedented global destabilization requiring urgent attention towards drug and vaccine development. Thalidomide, a drug with known anti-inflammatory and immunomodulatory effects has been indicated to be effective in treating a SARS-coV-2 pneumonia patient. Here, we study the possible mechanisms through which thalidomide might affect coronavirus disease-19 (COVID-19). METHODS: The present study explores the possibility of repurposing thalidomide for the treatment of SARS-coV-2 pneumonia by reanalysing transcriptomes of SARS-coV-2 infected tissues with thalidomide and lenalidomide induced transcriptomic changes in transformed lung and haematopoietic models as procured from databases, and further comparing them with the transcriptome of primary endothelial cells. RESULTS: Thalidomide and lenalidomide exhibited pleiotropic effects affecting a range of biological processes including inflammation, immune response, angiogenesis, MAPK signalling, NOD-like receptor signalling, Toll-like receptor signalling, leucocyte differentiation and innate immunity, the processes that are aberrantly regulated in severe COVID-19 patients. CONCLUSION: The present study indicates thalidomide analogues as a better fit for treating severe cases of novel viral infections, healing the damaged network by compensating the impairment caused by the COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Reposicionamiento de Medicamentos , Células Endoteliales , Humanos , Talidomida/farmacología
5.
Microvasc Res ; 128: 103939, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31676309

RESUMEN

A drug undergoes several in silico, in vitro, ex vivo and in vivo assays before entering into the clinical trials. In 2014, it was reported that only 32% of drugs are likely to make it to Phase-3 trials, and overall, only one in 10 drugs makes it to the market. Therefore, enhancing the precision of pre-clinical trial models could reduce the number of failed clinical trials and eventually time and financial burden in health sciences. In order to attempt the above, in the present study, we have shown that aortic ex-plants isolated from different stages of chick embryo and different regions of the aorta (pulmonary and systemic) have differential sprouting potential and response to angiogenesis modulatory drugs. Aorta isolated from HH37 staged chick embryo showed 16% (p < 0.001) and 11% (p < 0.001) increase in the number of tip cells at 72 h of culture compared to that of HH35 and HH29 respectively. The ascending order of the number of tip cells was found as central (Gen II), proximal (Gen I) and distal (Gen III) in a virtual zonal segmentation of endothelial sprouting. The HH37 staged aortas displayed differential responses to pro- and anti-angiogenic drugs like Vascular endothelial growth factor (VEGF), nitric oxide donor (spNO), and bevacizumab (avastin), thalidomide respectively. The human placenta tissue-culture however evinced endothelial sprouting only on day 12, with a gradual decrease in the number of tip cells until 21 days. In summary, this study provides an avant-garde angiogenic model emphasized on tip cells that would enhance the precision to test next-generation angiogenic drugs.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Aorta Torácica/embriología , Bioensayo , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Placenta/irrigación sanguínea , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Células Endoteliales/fisiología , Femenino , Humanos , Embarazo , Reproducibilidad de los Resultados , Factores de Tiempo , Técnicas de Cultivo de Tejidos
6.
Cell Biochem Funct ; 38(3): 319-329, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31989682

RESUMEN

Hypoxia is the most detrimental threat to humans residing at high altitudes, affecting multifaceted cellular responses that are crucial for normal homeostasis. Inhalation of nitric oxide has been successfully implemented to combat the hypoxia effect in the high altitude patients. We hypothesize that nitric oxide (NO) restores the peripheral blood mononuclear cell-matrix deadhesion during hypoxia. In the present study, we investigate the cellular action of exogenous NO in the hypoxia-mediated diminution of cell-matrix adhesion of PBMNC and NO bioavailability in vitro. The result showed that NO level and cell-matrix adhesion of PBMNC were significantly reduced in hypoxia as compared with normoxia, as assessed by the DAF-FM and cell adhesion assay, respectively. In contrast, cellular oxidative damage response was indeed upregulated in hypoxic PBMNC. Further, gene expression analysis revealed that mRNA transcripts of cell adhesion molecules (Integrin α5 and ß1) and eNOS expressions were significantly downregulated. The mechanistic study revealed that administration of NO and 8-Br-cGMP and overexpression of eNOS-GFP restored the basal NO level and recovers cell-matrix adhesion in PBMNC via cGMP-dependent protein kinase I (PKG I) signalling. In conclusion, NO-cGMP/PKG signalling may constitute a novel target to recover high altitude-afflicted cellular deadhesion. SIGNIFICANCE OF THIS STUDY: Cellular adhesion is a complex multistep process. The ability of cells to adhere to extracellular matrix is an essential physiological process for normal homeostasis and function. Hypoxia exposure in the PBMNC culture has been proposed to induce oxidative damage and cellular deadhesion and is generally believed to be the key factor in the reduction of NO bioavailability. In the present study, we demonstrated that NO donor or overexpression of eNOS-GFP has a protective effect against hypoxia-induced cellular deadhesion and greatly improves the redox balance by inhibiting the oxidative stress. Furthermore, this protective effect of NO is mediated by the NO-cGMP/PKG signal pathway, which may provide a potential strategy against hypoxia.


Asunto(s)
Hipoxia de la Célula , GMP Cíclico/metabolismo , Leucocitos Mononucleares/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Altitud , Adhesión Celular , Células Cultivadas , Medios de Cultivo/química , Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo
7.
Clin Exp Pharmacol Physiol ; 47(1): 7-15, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31549415

RESUMEN

Cardiovascular side effects of broadly used chemotherapeutic drugs such as Tamoxifen citrate (TC), Capecitabine (CP) and Epirubicin (EP) among cancer survivors are well established. Nitric oxide (NO) is known to protect cardiovascular tissues under conditions of stress. NO can act through cyclic guanosine monophosphate (cGMP)-dependent and -independent pathways. Particularly, the S-nitrosylation of SH-groups in a protein by NO falls under cGMP-independent effects of NO. TC, CP, and EP are hypothesized as interfering with cellular protein S-nitrosylation, which, in turn, may lead to endothelial dysfunctions. The results show that all three drugs attenuate nitrosylated proteins in endothelial cells. A significant reduction in endogenous S-nitrosylated proteins was revealed by Saville-Griess assay, immunofluorescence and western blot. Incubation with the drugs causes a reduction in endothelial migration, vasodilation and tube formation, while the addition of S-nitrosoglutathione (GSNO) has a reversal of this effect. In conclusion, results indicate the possibility of decreased cellular nitrosothiols as being one of the reasons for endothelial dysfunctions under TC, CP and EP treatment. Identification of the down-regulated S-nitrosylated proteins so as to correlate their implications on fundamental vascular functions could be an interesting phenomenon.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Células Endoteliales/efectos de los fármacos , Proteína S/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , GMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitrosoguanidinas/metabolismo , Vasodilatación/efectos de los fármacos
8.
Immunopharmacol Immunotoxicol ; 42(1): 48-55, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31983259

RESUMEN

Objectives: CRAC (Calcium Release Activated Calcium) channel is one of the most important channels regulating calcium influx and has been involved in many autoimmune diseases. The contribution of CRAC channel in the pathogenesis of Type 1 Diabetes (T1D) has not been described much. Thus, we aimed to study the expression of CRAC channel and inflammatory cytokines like IL-1ß (Interleukin -1ß) and TNF-α (Tumor Necrosis Factor-α) in the spleen-derived cytotoxic T cells, Bone marrow monocytes (BMM) and macrophages differentiated from BMM in the alloxan induced T1D mice.Materials and methods: BALB/c mice treated with alloxan and vehicle control for 12 and 24 h. Spleen derived T cells; Bone marrow derived monocytes were isolated from the control and diabetic BALB/c mice as well as macrophages differentiated from the control and diabetic BMM.Results: We observed increased expression of CRAC channel components like STIM1 (Stromal Interaction Molecule), ORAI1 and ORAI2 and inflammatory cytokines like IL-1ß and TNF-α in the spleen derived cytotoxic T cells and Macrophages differentiated from BMM as well as the downregulated expression of the same and CRAC channel in BMM of 12 and 24 h alloxan induced BALB/c mice.Conclusions: This study suggests that differential expression of CRAC channel correlated with the expression of inflammatory cytokines, thus CRAC channel might be responsible for the increased production of inflammatory cytokines in the alloxan induced T1D mice.


Asunto(s)
Células de la Médula Ósea/inmunología , Canales de Calcio Activados por la Liberación de Calcio/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Regulación de la Expresión Génica/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Interleucina-1beta/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Monocitos/patología , Factor de Necrosis Tumoral alfa/inmunología
9.
Calcif Tissue Int ; 104(3): 344-354, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30465120

RESUMEN

During mechanical unloading, endothelial cells reduce osteogenesis and increase bone resorption. Here we describe the feedback response of endothelial cells to unloaded osteoblasts. Primary endothelial cells, ex vivo mouse aortic rings and chicken egg yolk membranes were incubated with conditioned medium from mouse primary osteoblasts (OB-CM) subjected to unit gravity or simulated microgravity, to assess its effect on angiogenesis. In vivo injection of botulin toxin A (Botox) in the quadriceps and calf muscles of C57BL/6J mice was performed to mimic disuse osteoporosis. Unloaded osteoblasts showed strong upregulation of the pro-angiogenic factor, VEGF, and their conditioned medium increased in vitro endothelial cell viability, Cyclin D1 expression, migration and tube formation, ex vivo endothelial cell sprouting from aortic rings, and in ovo angiogenesis. Treatment with the VEGF blocker, avastin, prevented unloaded OB-CM-mediated in vitro and ex vivo enhancement of angiogenesis. Bone mechanical unloading by Botox treatment, known to reduce bone mass, prompted the overexpression of VEGF in osteoblasts. The cross talk between osteoblasts and endothelial cells plays a pathophysiologic role in the response of the endothelium to unloading during disuse osteoporosis. In this context, VEGF represents a prominent osteoblast factor stimulating angiogenesis.


Asunto(s)
Suspensión Trasera/fisiología , Neovascularización Fisiológica , Osteoblastos/fisiología , Estrés Mecánico , Animales , Toxinas Botulínicas Tipo A/farmacología , Células Cultivadas , Embrión de Pollo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Chem Res Toxicol ; 32(4): 589-602, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30834740

RESUMEN

Thalidomide is an infamous teratogen and it is continuously being explored for its anticancer properties. Fibroblast growth factor receptors (FGFRs) are implicated in embryo development and cancer pathophysiology. With striking similarities observed between FGFR implicated conditions and thalidomide embryopathy, we hypothesized thalidomide targets FGFRs. We utilized three different cell lines and chicken embryo model to investigate the effects of thalidomide and analogs on FGFR expression. We performed molecular docking, KINOMEscan analysis, and kinase activity assays to study the drug-protein interactions. The expression of FGFR1 and FGFR2 was differentially regulated by all the three drugs in cells as well as in developing organs. Transcriptome analysis of thalidomide-treated chick embryo strongly suggests the modulation of FGFR signaling and key transcription factors. Corroboration with previous studies suggests that thalidomide might affect FGFR expression through the transcription factor, E2F1. At the protein level, molecular docking predicted all three analogs to interact with lysine residue at 517th and 508th positions of FGFR2 and FGFR3, respectively. This lysine coordinates the ATP binding site of FGFR, thus hinting at the possible perturbation of FGFR activity by thalidomide. Kinome analysis revealed that kinase activities of FGFR2 and FGFR3 (G697C) reduced by 31% and 65%, respectively, in the presence of 10 µM thalidomide. Further, we checked and confirmed that the analogs inhibited the FGFR2 kinase activity in a dose-dependent manner. This study suggests that FGFRs could be potential targets of thalidomide and the two analogs, and also endorses the link between the teratogenicity and antitumor activities of the drugs.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Talidomida/análogos & derivados , Talidomida/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Pollos , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Relación Estructura-Actividad , Talidomida/química
11.
Nitric Oxide ; 82: 48-58, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30439561

RESUMEN

Angiogenesis is the formation of new blood vessels that occurs by two distinct processes following sprouting angiogenesis (SA) and intussusceptive angiogenesis (IA). Nitric oxide (NO) is known for its pro-angiogenic functions. However, no clear mechanisms are delineated on its role in promoting angiogenesis in reparative wound healing. We propose that NO regulates SA to IA transition and vice versa in wound milieu. We have used three models which include a new chick embryo extra-vasculature (CEV) burn wound model, adult Tie2-GFP transgenic Zebrafish caudal fin regeneration model and Zebrafish skin wound model to study the mechanisms underlying behind the role of NO in wound healing. Wounds created in CEV were treated with NO donor (Spermine NONOate (SPNO)), NOS inhibitor (L-nitro-l-arginine-methyl ester (l-NAME)), NaNO2, NaNO3, and beetroot juice, a nitrite-rich juice respectively and the pattern of wound healing was assessed. Morphological and histological techniques tracked the wound healing at the cellular level, and the molecular changes were investigated by using real-time RT-PCR gene expression analysis. The result concludes that NO donor promotes wound healing by activating SA at an early phase of healing while NOS inhibitor induces wound healing via IA. At the later phase of wound healing NO donor followed IA while NOS inhibitor failed to promote wound repair. The current work underpinned a differential regulation of NO on angiogenesis in wound milieu and this study would provide new insights in designing therapeutics for promoting wound repair.


Asunto(s)
Neovascularización Patológica/tratamiento farmacológico , Óxido Nítrico/farmacología , Cicatrización de Heridas/efectos de los fármacos , Pez Cebra/genética , Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Embrión de Pollo , Modelos Animales de Enfermedad , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Óxido Nítrico/metabolismo , Pez Cebra/metabolismo
12.
Microsc Microanal ; 25(4): 961-970, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072413

RESUMEN

Vasodilation occurs as a result of the relaxation of the smooth muscle cells present in the walls of blood vessels. Various suitable models are available for the analysis of the vasoactive properties of drugs with therapeutic applications. But all these models have limitations, such as ethical issues and high cost. The purpose of this study is to develop an alternative model for studying the vasoactive properties of drugs using an in-ovo chicken embryo model. In the preliminary experiment, we used a well-known vasoconstrictor (adrenaline) and a vasodilator (spermine NoNoate) in the chick embryo area vasculosa and evaluated their concentration-response curve. Adrenaline (10 µM) and spermine NoNoate (10 µM) were administered in different arteries and veins and different positions of the right vitelline artery of the chick embryo. Results showed the middle of the vessel bed of the right vitelline artery having the best vasoactive effect compared to others. Finally, anti-hypertensive drugs, calcium channel blockers, and NOS agonists were administered in the chick embryo area vasculosa to validate the model. Results demonstrate that the chick embryo area vasculosa can be an alternative, robust, and unique in-ovo model for screening of anti-hypertensive drugs in real time.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Microscopía Intravital/métodos , Vasoconstrictores/aislamiento & purificación , Vasoconstrictores/farmacología , Vasodilatadores/aislamiento & purificación , Vasodilatadores/farmacología , Animales , Embrión de Pollo
13.
Int J Mol Sci ; 20(5)2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30866404

RESUMEN

Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) plays crucial roles in cardiac homeostasis. Adult cardiomyocyte specific overexpression of eNOS confers protection against myocardial-reperfusion injury. However, the global effects of NO overexpression in developing cardiovascular system is still unclear. We hypothesized that nitric oxide overexpression affects the early migration of cardiac progenitor cells, vasculogenesis and function in a chick embryo. Vehicle or nitric oxide donor DEAN (500 mM) were loaded exogenously through a small window on the broad side of freshly laid egg and embryonic development tracked by live video-microscopy. At Hamburg Hamilton (HH) stage 8, the cardiac progenitor cells (CPC) were isolated and cell migration analysed by Boyden Chamber. The vascular bed structure and heart beats were compared between vehicle and DEAN treated embryos. Finally, expression of developmental markers such as BMP4, Shh, Pitx2, Noggin were measured using reverse transcriptase PCR and in-situ hybridization. The results unexpectedly showed that exogenous addition of pharmacological NO between HH stage 7⁻8 resulted in embryos with situs inversus in 28 out of 100 embryos tested. Embryos treated with NO inhibitor cPTIO did not have situs inversus, however 10 embryos treated with L-arginine showed a situs inversus phenotype. N-acetyl cysteine addition in the presence of NO failed to rescue situs inversus phenotype. The heart beat is normal (120 beats/min) although the vascular bed pattern is altered. Migration of CPCs in DEAN treated embryos is reduced by 60% compared to vehicle. BMP4 protein expression increases on the left side of the embryo compared to vehicle control. The data suggests that the NO levels in the yolk are important in turning of the heart during embryonic development. High levels of NO may lead to situs inversus condition in avian embryo by impairing cardiac progenitor cell migration through the NO-BMP4-cGMP axis.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Corazón/fisiología , Miocitos Cardíacos/citología , Óxido Nítrico/farmacología , Situs Inversus/inducido químicamente , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Desarrollo Embrionario , Corazón/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Microscopía por Video , Miocitos Cardíacos/efectos de los fármacos , Situs Inversus/genética , Regulación hacia Arriba
14.
Microvasc Res ; 119: 47-59, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29649432

RESUMEN

Existing animal models for screening tumor angiogenic process have various setbacks that necessitate further investigations. In this study, we developed an ex-ovo egg yolk angiogenesis model to screen the angiogenic potency of tumor cells (HeLa and SiHa cell lines). The egg yolk angiogenesis assay was applied to study the nitric oxide (NO) influence on switching from sprouting angiogenesis (SA) to intussusceptive angiogenesis (IA) under tumor microenvironment. Morphological analysis and SA-like or IA-like markers expression were determined during the development of chicken chorioallantoic membrane (CAM) from day 5 to 13. Expression of Notch1, Notch2, EphrinB2, and Tie2 were considered as SA-like while TEM8, CALD1, CXCR4 and HOMX1 were followed as IA-like markers. The HeLa and SiHa cell lines embedded CAM showed an increase in micro and macro blood vessels and vascular size, junction and length which are the pivotal morphological parameters of angiogenesis. Further, the study revealed that HeLa is more aggressive than SiHa in inducing tumor angiogenesis. To determine the NO signaling implication in tumor milieu, NO donor (Spermine NONOate (SPNO)), NOS inhibitor (L-nitro-L-arginine-methyl ester (L-NAME) and VEGF inhibitor (Avastin) were administrated to chick embryo vascular bed with and without HeLa cells. The results demonstrated that HeLa cells promote IA through NO signaling, VEGF and eNOS and it was documented by angiogenic morphological parameters and SA-like or IA-like markers expression. Therefore, our study claims that ex-ovo egg yolk angiogenesis model could be used to study tumor angiogenesis and NO plays a key role in switching of IA under tumor microenvironment.


Asunto(s)
Neovascularización Patológica , Óxido Nítrico/metabolismo , Transducción de Señal , Neoplasias del Cuello Uterino/irrigación sanguínea , Neoplasias del Cuello Uterino/metabolismo , Animales , Embrión de Pollo , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Células HeLa , Humanos , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Microambiente Tumoral , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Future Oncol ; 14(23): 2383-2401, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30141351

RESUMEN

AIM: To understand why thalidomide and lenalidomide exhibit different responses in metastatic prostate cancer (mPCa) treatment. METHODS: We analyzed the perturbation signatures of thalidomide, lenalidomide, flutamide treated mPCa cell line from Library of Integrated Network-based Cellular Signatures database and transcriptome of docetaxel-treated mPCa patients. RESULTS: Flutamide and docetaxel downregulated 'Steroid Biosynthesis', 'Cell cycle' and PCa specific transcription factor networks. Thalidomide inhibited 'Cell cycle' and 'E2F network', possibly accounting for its synergistic effects with docetaxel. Conversely, lenalidomide promoted 'Cell cycle' and 'Cholesterol biosynthesis'. CONCLUSION: Hence, we propose that lenalidomide upregulates cholesterol synthesis followed by enhanced rate of cell cycle, thereby nurturing a hyperproliferative tumor microenvironment. In summary, this study offers a possible explanation for the differential outcomes in the treatment of mPCa with thalidomide and lenalidomide.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Andrógenos/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Colesterol/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Lenalidomida/administración & dosificación , Masculino , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transducción de Señal/efectos de los fármacos , Talidomida/administración & dosificación , Transcriptoma
16.
Biochem Cell Biol ; 95(2): 263-272, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28177770

RESUMEN

Liver fibrosis is now well recognized as the causative factor for increased mortality from complications associated with liver pathologies. Activated hepatic stellate cells (HSCs) play a critical role in the progression of liver fibrosis. Therefore, targeting these activated HSCs to prevent and (or) treat liver disease is a worthwhile approach to explore. In the present in vitro study, we investigated the use of bipotential murine oval liver cells (BMOL) in regulating the functions of activated HSCs to prevent progression of liver fibrosis. We used a conditioned medium-based approach to study the effect of BMOL cells on activated HSC survival and function. Our data showed that BMOL cells block the contraction of activated HSCs by inducing apoptosis of these cells. We demonstrated that BMOL cells secrete soluble factors, such as interleukin-6 (IL-6), which induced apoptosis of activated HSCs. Using both pharmacological and molecular inhibitor approaches, we further identified that IL-6-mediated activation of NF-κB-iNOS-NO-ROS signaling in activated HSCs plays a critical role in BMOL-cell-mediated apoptosis of activated HSCs. Thus, the present study provides an alternative cell-based therapeutic approach to treat liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/efectos de los fármacos , Interleucina-6/farmacología , FN-kappa B/genética , Óxido Nítrico Sintasa de Tipo II/genética , Células Madre/metabolismo , Amidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Bencilaminas/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Células Cultivadas , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Imidazoles/farmacología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Modelos Biológicos , FN-kappa B/agonistas , FN-kappa B/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Quinoxalinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/efectos de los fármacos
17.
Chem Res Toxicol ; 30(10): 1883-1896, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28892372

RESUMEN

Since the conception of thalidomide as a teratogen, approximately 30 hypotheses have been put forward to explain the developmental toxicity of the molecule. However, no systems biology approach has been taken to understand the phenomena yet. The proposed work was aimed to explore the mechanism of thalidomide toxicity in developing chick embryo in the context of transcriptomics by using genome wide RNA sequencing data. In this study, we challenged the developing embryo at the stage of blood island formations (HH8), which is the most vulnerable stage for thalidomide-induced deformities. We observed that thalidomide affected the early vasculogenesis through interfering with the blood island formation extending the effect to organogenesis. The transcriptome analyses of the embryos collected on sixth day of incubation showed that liver, eye, and blood tissue associated genes were down regulated due to thalidomide treatment. The conserved gene coexpression module also indicated that the genes involved in lens development were heavily affected. Further, the Gene Ontology analysis explored that the pathways of eye development, retinol metabolism, and cartilage development were dampened, consistent with the observed deformities of various organs. The study concludes that thalidomide exerts its toxic teratogenic effects through interfering with early extra-embryonic vasculogenesis and ultimately gives an erroneous transcriptomic pattern to organogenesis.


Asunto(s)
Perfilación de la Expresión Génica , Neovascularización Patológica/genética , Organogénesis/genética , Talidomida/toxicidad , Animales , Embrión de Pollo , Neovascularización Patológica/inducido químicamente , Organogénesis/efectos de los fármacos , Talidomida/administración & dosificación
18.
Nitric Oxide ; 66: 30-42, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28267592

RESUMEN

Nitric oxide (NO) plays a critical role in endothelial functions such as cellular migration, vascular permeability and angiogenesis. Angiogenesis, the formation of new blood vessels from "pre-existing" ones is a carefully regulated process and essential during reproduction, development and wound healing. Previously our lab group reported that Secreted Frizzled-Related Protein 4 (sFRP4) could inhibit angiogenesis in both in vitro and in vivo conditions. sFRP4 belongs to a family of secreted glycoproteins that function as antagonists of the canonical Wnt signalling pathway. Although the pro-apoptotic role of sFRP4 is well discussed in literature, little is known in regards to its anti-angiogenic property. The objective of this study was to elucidate sFRP4 implications in NO biology of the endothelium. Results demonstrate that sFRP4 causes endothelial dysfunction by suppressing NO-cGMP signaling and elevating corresponding ROS levels. The imbalance between NO and ROS levels results in apoptosis and subsequent leakiness of endothelium as confirmed in vivo (Texas red/Annxin - CAM assay) and in vitro (Monolayer permeability assay) conditions. Furthermore utilizing peptides synthesized from the CRD domain of sFRP4, our results showed that while these peptides were able to cause endothelial dysfunctions, they did not cause apoptosis of the endothelial cells. Thereby confirming that sFRP4 can mediate its anti-angiogenic effect independent of its pro-apoptotic property. In conclusion, the current study reports that sFRP4-mediated anti-angiogenesis occurs as a result of impaired NO-cGMP signaling which in turn allow for elevation of redox levels and promotion of apoptosis of endothelial cells.


Asunto(s)
Apoptosis/fisiología , Permeabilidad de la Membrana Celular/fisiología , GMP Cíclico/metabolismo , Endotelio/metabolismo , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Humanos , Neovascularización Patológica/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal/fisiología
19.
Bioorg Chem ; 67: 139-47, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27388635

RESUMEN

A series of novel 5-benzylidene-2,4-thiazolidinediones were designed as inhibitors of angiogenesis targeting VEGFR-2. In docking study, molecules showed similar way of binding with VEGFR-2 as that of the co-crystallized ligand. Compounds were then synthesized, purified and characterized by spectroscopic techniques. Compounds 3f and 3i were found to be most active in the series showing good inhibition of angiogenesis in both CAM and in zebrafish embryo assays. Compound 3i also exhibited IC50 of 0.5µM against VEGFR-2.


Asunto(s)
Compuestos de Bencilideno/farmacología , Diseño de Fármacos , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Compuestos de Bencilideno/síntesis química , Compuestos de Bencilideno/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Neovascularización Patológica/patología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Tiazoles/síntesis química , Tiazoles/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
20.
Angiogenesis ; 18(2): 175-89, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25510468

RESUMEN

Angiogenesis, the formation of new blood vessels from pre-existing vessels, is a complex process that warrants cell migration, proliferation, tip cell formation, ring formation, and finally tube formation. Angiogenesis is initiated by a single leader endothelial cell called "tip cell," followed by vessel elongation by "stalk cells." Tip cells are characterized by their long filopodial extensions and expression of vascular endothelial growth factor receptor-2 and endocan. Although nitric oxide (NO) is an important modulator of angiogenesis, its role in angiogenic sprouting and specifically in tip cell formation is poorly understood. The present study tested the role of endothelial nitric oxide synthase (eNOS)/NO/cyclic GMP (cGMP) signaling in tip cell formation. In primary endothelial cell culture, about 40% of the tip cells showed characteristic sub-cellular localization of eNOS toward the anterior progressive end of the tip cells, and eNOS became phosphorylated at serine 1177. Loss of eNOS suppressed tip cell formation. Live cell NO imaging demonstrated approximately 35% more NO in tip cells compared with stalk cells. Tip cells showed increased level of cGMP relative to stalk cells. Further, the dissection of NO downstream signaling using pharmacological inhibitors and inducers indicates that NO uses the sGC/cGMP pathway in tip cells to lead angiogenesis. Taken together, the present study confirms that eNOS/NO/cGMP signaling defines the direction of tip cell migration and thereby initiates new blood vessel formation.


Asunto(s)
Óxido Nítrico/fisiología , Animales , Bovinos , Línea Celular Transformada , Pollos , GMP Cíclico/metabolismo , Humanos , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA