Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 412
Filtrar
1.
Sci Rep ; 11(1): 6067, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33727583

RESUMEN

Dismal prognosis of glioblastoma (GBM) prompts for the identification of response predictors and therapeutic resistance mechanisms of current therapies. The authors investigated the impact of c-Met, HGF, VEGFR2 expression and microvessel density (MVD) in GBM patients submitted to second-line chemotherapy with bevacizumab. Immunohistochemical expression of c-Met, HGF, VEGFR2, and MVD was assessed in tumor specimens of GBM patients treated with bevacizumab, after progression under temozolomide. Survival analysis was evaluated according to the expression of the aforementioned biomarkers. c-Met overexpression was associated with a time-to-progression (TTP) after bevacizumab of 3 months (95% CI, 1.5-4.5) compared with a TTP of 7 months (95% CI, 4.6-9.4) in patients with low or no expression of c-Met (p = 0.05). VEGFR2 expression was associated with a TTP after bevacizumab of 3 months (95% CI, 1.8-4.2) compared with a TTP of 7 months (95% CI, 5.7-8.3) in patients with no tumoral expression of VEGFR2 (p = 0.009). Concomitant c-Met/VEGFR2 overexpression was associated with worse overall survival (13 months) compared with concomitant c-Met/VEGFR2 negative expression (19 months; p = 0.025). Our data support the hypothesis that c-Met and VEGFR2 overexpression have a role in the development of glioblastoma early resistance and might predict poorer responses to anti-angiogenic therapies.


Asunto(s)
Bevacizumab/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma , Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Glioblastoma/irrigación sanguínea , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tasa de Supervivencia
2.
Angiogenesis ; 24(3): 519-531, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33449298

RESUMEN

Receptor-interacting protein kinase 3 (RIPK3) is a multifunctional intracellular protein that was first recognized as an important component of the necroptosis programmed cell death pathway. RIPK3 is also highly expressed in non-necroptotic murine embryonic endothelial cells (ECs) during vascular development, indicating its potential contribution to angiogenesis. To test this hypothesis, we generated mice lacking endothelial RIPK3 and found non-lethal embryonic and perinatal angiogenesis defects in multiple vascular beds. Our in vitro data indicate that RIPK3 supports angiogenesis by regulating growth factor receptor degradation in ECs. We found that RIPK3 interacted with the membrane trafficking protein myoferlin to sustain expression of vascular endothelial growth factor receptor 2 (VEGFR2) in cultured ECs following vascular endothelial growth factor A (VEGFA) stimulation. Restoration of myoferlin, which was diminished after RIPK3 knockdown, rescued decreased VEGFR2 expression and vascular sprouting in RIPK3-deficient ECs after VEGFA treatment. In addition, we found that RIPK3 modulated expression of genes involved in endothelial identity by inhibiting ERK signaling independently of growth factor receptor turnover. Altogether, our data reveal unexpected non-necroptotic roles for RIPK3 in ECs and evidence that RIPK3 promotes developmental angiogenesis in vivo.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Neovascularización Fisiológica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Ratones , Ratones Transgénicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
3.
Urol Oncol ; 39(1): 78.e9-78.e16, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32988712

RESUMEN

OBJECTIVES: Few studies have independently investigated the population of patients with synchronous metastatic renal cell carcinoma (smRCC). In this study, we evaluated programmed death protein-ligand 1 (PD-L1) and vascular endothelial growth factor receptor 2 (VEGFR-2) expression in primary tumor tissue of smRCC. METHODS: A total of 96 patients with smRCC who were treated with cytoreductive nephrectomy followed by targeted therapy from January 2006 to January 2013 were identified. PD-L1 and VEGFR-2 expression were evaluated by immunohistochemistry. Kaplan-Meier and Cox methods were used for analysis. RESULTS: PD-L1 and VEGFR-2 protein immunopositivity were observed in 39.6% (38 of 96) and 58.3% (56 of 96) of patients, respectively. A significant correlation was detected between VEGFR-2 and PD-L1 expression (P = 0.030). Based on PD-L1 and VEGFR-2 expression, patients with intermediate-risk disease (n = 63) were divided into 4 subgroups including patients who were PD-L1 (+) VEGFR-2 (+) (n = 21), PD-L1 (+) VEGFR-2 (-) (n = 11), PD-L1 (-) VEGFR-2 (+) (n = 15) and PD-L1 (-) VEGFR-2 (-) (n = 16). Compared to the PD-L1 (-) VEGFR-2 (+), PD-L1 (+) VEGFR-2 (+) and PD-L1 (-) VEGFR-2 (-) groups, patients in the PD-L1 (+) VEGFR-2 (-) group had shorter progression-free survival (median, 9.0 vs. 20.0, 16.0 and 15.5 months, P < 0.05) and overall survival (median, 14.0 vs. 33.0, 24.0 and 26.5 months, P < 0.05). CONCLUSIONS: Intermediate-risk smRCC patients with PD-L1-positive and VEGFR-2-negative expression who were treated with targeted therapy following cytoreductive nephrectomy had poor prognoses. We suggest that other treatments beyond sunitinib or sorafenib may be explored in this subgroup.


Asunto(s)
Antineoplásicos/uso terapéutico , Antígeno B7-H1/biosíntesis , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/terapia , Procedimientos Quirúrgicos de Citorreducción , Neoplasias Renales/metabolismo , Neoplasias Renales/terapia , Nefrectomía/métodos , Sorafenib/uso terapéutico , Sunitinib/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Anciano , Carcinoma de Células Renales/secundario , Terapia Combinada , Femenino , Humanos , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
4.
J Biol Chem ; 295(34): 12086-12098, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32636303

RESUMEN

Disorganized vessels in the tumor vasculature lead to impaired perfusion, resulting in reduced accessibility to immune cells and chemotherapeutic drugs. In the breast tumor-stroma interplay, paracrine factors such as interleukin-6 (IL-6) often facilitate disordered angiogenesis. We show here that epigenetic mechanisms regulate the crosstalk between IL-6 and vascular endothelial growth factor receptor 2 (VEGFR2) signaling pathways in myoepithelial (CD10+) and endothelial (CD31+, CD105+, CD146+, and CD133-) cells isolated from malignant and nonmalignant tissues of clinically characterized human breast tumors. Tumor endothelial (Endo-T) cells in 3D cultures exhibited higher VEGFR2 expression levels, accelerated migration, invasion, and disorganized sprout formation in response to elevated IL-6 levels secreted by tumor myoepithelial (Epi-T) cells. Constitutively, compared with normal endothelial (Endo-N) cells, Endo-T cells differentially expressed DNA methyltransferase isoforms and had increased levels of IL-6 signaling intermediates such as IL-6R and signal transducer and activator of transcription 3 (STAT3). Upon IL-6 treatment, Endo-N and Endo-T cells displayed altered expression of the DNA methyltransferase 1 (DNMT1) isoform. Mechanistic studies revealed that IL-6 induced proteasomal degradation of DNMT1, but not of DNMT3A and DNMT3B and subsequently led to promoter hypomethylation and expression/activation of VEGFR2. IL-6-induced VEGFR2 up-regulation was inhibited by overexpression of DNMT1. Transfection of a dominant-negative STAT3 mutant, but not of STAT1, abrogated VEGFR2 expression. Our results indicate that in the breast tumor microenvironment, IL-6 secreted from myoepithelial cells influences DNMT1 stability, induces the expression of VEGFR2 in endothelial cells via a promoter methylation-dependent mechanism, and leads to disordered angiogenesis.


Asunto(s)
Neoplasias de la Mama , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Interleucina-6/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-6/genética , Células MCF-7 , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
5.
Int J Mol Sci ; 21(15)2020 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-32722551

RESUMEN

The vascular endothelial growth factor (VEGF), a homodimeric vasoactive glycoprotein, is the key mediator of angiogenesis. Angiogenesis, the formation of new blood vessels, is responsible for a wide variety of physio/pathological processes, including cardiovascular diseases (CVD). Cardiomyocytes (CM), the main cell type present in the heart, are the source and target of VEGF-A and express its receptors, VEGFR1 and VEGFR2, on their cell surface. The relationship between VEGF-A and the heart is double-sided. On the one hand, VEGF-A activates CM, inducing morphogenesis, contractility and wound healing. On the other hand, VEGF-A is produced by CM during inflammation, mechanical stress and cytokine stimulation. Moreover, high concentrations of VEGF-A have been found in patients affected by different CVD, and are often correlated with an unfavorable prognosis and disease severity. In this review, we summarized the current knowledge about the expression and effects of VEGF-A on CM and the role of VEGF-A in CVD, which are the most important cause of disability and premature death worldwide. Based on clinical studies on angiogenesis therapy conducted to date, it is possible to think that the control of angiogenesis and VEGF-A can lead to better quality and span of life of patients with heart disease.


Asunto(s)
Regulación de la Expresión Génica , Cardiopatías/metabolismo , Miocitos Cardíacos/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Cardiopatías/patología , Humanos , Miocitos Cardíacos/patología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
6.
ASN Neuro ; 12: 1759091420926836, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32423231

RESUMEN

Nuclear-distribution element-like 1 (NDEL1) is associated with the proliferation and migration of neurons. Vascular endothelial growth factor (VEGF) in combination with VEGF receptor-2 (VEGFR-2) regulates the proliferation and migration of neurons. This study was performed to explore undefined alterations in the expression levels of NDEL1 and VEGF/VEGFR-2 within the hippocampus after status epilepticus (SE). Following the creation of pilocarpine-induced epilepsy models using adolescent male C57BL/6 mice, Western blotting and reverse transcription quantitative polymerase chain reaction were applied to assess the levels of NDEL1, VEGF, and VEGFR-2 expression in whole hippocampi at 1, 2, 3, and 4 weeks post-SE, respectively. Immunofluorescent labeling was also employed to detect the colocalization of NDEL1 and VEGF in the hippocampus. Our results indicated that NDEL1 and VEGF have similar patterns of upregulation throughout the hippocampus. Upregulation of VEGFR-2 occurred only in the early stages, and the expression decreased shortly afterward. NDEL1 and VEGF were coexpressed in the cornu ammonis 3 pyramidal cell, granular, and polymorph layers of the dentate gyrus in the hippocampus. This study revealed that NDEL1, VEGF, and VEGFR-2 may work together and are involved in the pathophysiology in the hippocampus after SE.


Asunto(s)
Proteínas Portadoras/biosíntesis , Hipocampo/metabolismo , Estado Epiléptico/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pilocarpina/toxicidad , Estado Epiléptico/inducido químicamente , Estado Epiléptico/patología
7.
Mol Biol Rep ; 47(6): 4263-4272, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32436041

RESUMEN

Tachykinins such as Substance P (SP) are a group of neuropeptides that are involved in cancer development. Neurokinin-1 receptor (NK-1R) is the main tachykinin receptor mediating the effects of SP, which is overexpressed in human esophageal squamous cell carcinoma (ESCC) and other malignant tissues. However, the effects of SP/NK-1R system on the migration of esophageal cancer cells and angiogenesis is not clear yet. This study seeks to obtain data to address these research gaps. In order to assess the effects of the FDA-approved aprepitant drug, a commercially available NK-1R antagonist, on the viability of KYSE-30 ESCC cells, resazurin assay was performed. The influence of SP/NK-1R system on the migration potential of these cells was examined using scratch assay. The effects of this system on the expression levels of metastatic factors were also examined by RT-PCR and western blot analyses. The half-maximal inhibitory concentration (IC50) value for KYSE-30 cells treated with aprepitant found to be 29.88 µM. Treatment with SP significantly promoted KYSE-30 esophageal cancer cell migration, and aprepitant blocked this effect. In addition, SP significantly induced the expression of matrix metalloproteinase-2 (MMP-2), MMP-9, vascular endothelial growth factor-A (VEGF-A), and VEGF receptor1 (VEGFR1) in the cells, whereas aprepitant inhibited the up-regulation effects caused by SP. SP plays important roles in the development of human esophageal squamous cell carcinoma by promoting cancer cell invasion and enhancing the expression of factors involved in cellular migration and angiogenesis, which can be blocked by the NK-1R antagonist, aprepitant.


Asunto(s)
Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Sustancia P/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Apoptosis/efectos de los fármacos , Aprepitant/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Antagonistas del Receptor de Neuroquinina-1/farmacología , Receptores de Neuroquinina-1/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
8.
Int J Dev Neurosci ; 80(4): 335-346, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32198947

RESUMEN

Early responses to a neurological excitotoxic process include blood-brain barrier (BBB) impairment and overexpression of vascular endothelial growth factor (VEGF), but the long-term effects of excitotoxicity on the BBB properties remain unknown. To assess this, we induced an excitotoxic process on male rats by neonatal monosodium glutamate (MSG) treatment. At postnatal day (PD) 60, we measured the expression level of structural proteins of the BBB and the VEGF type-2 receptor (VEGFR-2) protein in the cerebral motor cortex (CMC), striatum (STR), hippocampus (Hp), entorhinal cortex (Ent), and hypothalamus (Hyp). We also measured BBB permeability in the same cerebral regions. Neonatal MSG treatment significantly reduced the protein expression level of claudin-5 in the CMC, and of ZO-1 in the CMC and Hp, and increased the expression level of plasmalemmal vesicle-associated protein in the CMC, and of VEGFR-2 in all regions except for the Hyp. BBB permeability was significantly higher in all studied regions of MSG-treated animals after hypertonic shock (HS). The increased BBB permeability observed in the MSG-treated animals after HS was reversed by VEGFR-2 inhibition with SU5416. We conclude that neonatal excitotoxicity leads to lasting impairment on BBB properties in adulthood, increasing its susceptibility to HS that could be regulated by VEGFR-2 activity inhibition.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Glutamato de Sodio/toxicidad , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Indoles/farmacología , Masculino , Presión Osmótica/efectos de los fármacos , Pirroles/farmacología , Ratas , Ratas Wistar , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
9.
Cell Mol Life Sci ; 77(5): 885-901, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31278420

RESUMEN

Purinergic P2 receptors are critical regulators of several functions within the vascular system, including platelet aggregation, vascular inflammation, and vascular tone. However, a role for ATP release and P2Y receptor signalling in angiogenesis remains poorly defined. Here, we demonstrate that blood vessel growth is controlled by P2Y2 receptors. Endothelial sprouting and vascular tube formation were significantly dependent on P2Y2 expression and inhibition of P2Y2 using a selective antagonist blocked microvascular network generation. Mechanistically, overexpression of P2Y2 in endothelial cells induced the expression of the proangiogenic molecules CXCR4, CD34, and angiopoietin-2, while expression of VEGFR-2 was decreased. Interestingly, elevated P2Y2 expression caused constitutive phosphorylation of ERK1/2 and VEGFR-2. However, stimulation of cells with the P2Y2 agonist UTP did not influence sprouting unless P2Y2 was constitutively expressed. Finally, inhibition of VEGFR-2 impaired spontaneous vascular network formation induced by P2Y2 overexpression. Our data suggest that P2Y2 receptors have an essential function in angiogenesis, and that P2Y2 receptors present a therapeutic target to regulate blood vessel growth.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/crecimiento & desarrollo , Neovascularización Fisiológica/fisiología , Receptores Purinérgicos P2Y2/metabolismo , Angiopoyetina 2/biosíntesis , Antígenos CD34/biosíntesis , Células Cultivadas , Humanos , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Fosforilación/fisiología , Agregación Plaquetaria/fisiología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores CXCR4/biosíntesis , Receptores Purinérgicos P2Y2/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
10.
BMJ Case Rep ; 12(11)2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31776151

RESUMEN

The angiogenesis inhibitor ramucirumab (IMC-1121B) is a fully humanised IgG1 monoclonal antibody targeting the extracellular domain of vascular endothelial growth factor receptor 2. Ramucirumab has been approved as a second-line treatment for lung cancer. Pyogenic granuloma is an acquired, benign vascular tumour of the skin or mucous membrane. We encountered a patient with pyogenic granuloma who was treated with ramucirumab. The patient was a 48-year-old Japanese woman with advanced lung cancer who had been heavily pretreated using several lines of chemotherapy. Ramucirumab was administered as the fifth-line treatment with docetaxel. After 10 days, a painless rice-coloured or pink papule appeared on her finger. One month later, it increased in size to 20 mm. We examined the pathological condition by immunostaining using the resected specimen diagnosed as pyogenic granuloma. Paradoxically, this vascular tumour arose during the administration of an angiogenesis inhibitor.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/efectos adversos , Granuloma Piogénico/etiología , Granuloma Piogénico/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , Ramucirumab
11.
Anesthesiology ; 131(5): 1125-1147, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31567367

RESUMEN

BACKGROUND: Cancer pain is a pervasive clinical symptom impairing life quality. Vascular endothelial growth factor A has been well studied in tumor angiogenesis and is recognized as a therapeutic target for anti-cancer treatment. This study tested the hypothesis that vascular endothelial growth factor A and vascular endothelial growth factor receptor 2 contribute to bone cancer pain regulation associated with spinal central sensitization. METHODS: This study was performed on female rats using a metastatic breast cancer bone pain model. Nociceptive behaviors were evaluated by mechanical allodynia, thermal hyperalgesia, spontaneous pain, and CatWalk gait analysis. Expression levels were measured by real-time quantitative polymerase chain reaction, western blot, and immunofluorescence analysis. Excitatory synaptic transmission was detected by whole-cell patch-clamp recordings. The primary outcome was the effect of pharmacologic intervention of spinal vascular endothelial growth factor A/vascular endothelial growth factor receptor 2-signaling on bone cancer pain behaviors. RESULTS: The mRNA and protein expression of vascular endothelial growth factor A and vascular endothelial growth factor receptor 2 were upregulated in tumor-bearing rats. Spinal blocking vascular endothelial growth factor A or vascular endothelial growth factor receptor 2 significantly attenuated tumor-induced mechanical allodynia (mean ± SD: vascular endothelial growth factor A, 7.6 ± 2.6 g vs. 5.3 ± 3.3 g; vascular endothelial growth factor receptor 2, 7.8 ± 3.0 g vs. 5.2 ± 3.4 g; n = 6; P < 0.0001) and thermal hyperalgesia (mean ± SD: vascular endothelial growth factor A, 9.0 ± 2.4 s vs. 7.4 ± 2.7 s; vascular endothelial growth factor receptor 2, 9.3 ± 2.5 s vs. 7.5 ± 3.1 s; n = 6; P < 0.0001), as well as spontaneous pain and abnormal gaits. Exogenous vascular endothelial growth factor A enhanced excitatory synaptic transmission in a vascular endothelial growth factor receptor 2-dependent manner, and spinal injection of exogenous vascular endothelial growth factor A was sufficient to cause pain hypersensitivity via vascular endothelial growth factor receptor 2-mediated activation of protein kinase C and Src family kinase in naïve rats. Moreover, spinal blocking vascular endothelial growth factor A/vascular endothelial growth factor receptor 2 pathways suppressed protein kinase C-mediated N-methyl-D-aspartate receptor activation and Src family kinase-mediated proinflammatory cytokine production. CONCLUSIONS: Vascular endothelial growth factor A/vascular endothelial growth factor receptor 2 contributes to central sensitization and bone cancer pain via activation of neuronal protein kinase C and microglial Src family kinase pathways in the spinal cord.


Asunto(s)
Neoplasias Óseas/metabolismo , Dolor en Cáncer/metabolismo , Dimensión del Dolor/métodos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Neoplasias Óseas/patología , Dolor en Cáncer/patología , Femenino , Inyecciones Espinales , Dimensión del Dolor/efectos de los fármacos , Quinazolinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
12.
Mol Biotechnol ; 61(11): 860-872, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31531759

RESUMEN

Angiogenesis is a biological process finely tuned by a plethora of pro- and anti-angiogenic molecules, among which vascular endothelial growth factors (VEGFs). Their biological activity is expressed through the interaction with three cognate receptor tyrosine kinases, VEGFR1, 2, and 3. VEGFR2 is the primary regulator of angiogenesis. Ligand-induced VEGFR2 dimerization and activation depend on direct ligand binding to extracellular domains 2 and 3 of receptor and in the establishment of interactions between proximal membrane domains. VEGFR2 domain 7 has been shown to play a crucial role in receptor dimerization and regulation, therefore, representing a convenient target for the allosteric modulation of VEGFR2 activity. The ability to prepare a functional VEGFR2D7 domain represents the starting point to the development of novel VEGFR2 binders acting as allosteric inhibitors of receptor activity. Here, we describe a robust and efficient procedure for the preparation in E. coli of the VEGFR2 domain 7. The protein was obtained with a good yield and was properly folded. It was investigated in a biochemical and structural study, providing information on its conformational arrangement and in solution properties.


Asunto(s)
Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Regulación Alostérica , Dicroismo Circular , Escherichia coli/metabolismo , Espectroscopía de Resonancia Magnética , Dominios Proteicos/genética , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Dispersión de Radiación , Espectrometría de Fluorescencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/aislamiento & purificación
13.
J Cancer Res Clin Oncol ; 145(4): 1063-1073, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30810838

RESUMEN

OBJECTIVE: The impact of angiogenesis on long-term survival of high-grade serous ovarian cancer (HGSOC) patients remains unclear. This study investigated whether angiogenic markers correlated with 5-year progression-free survival (PFS) in a large cohort of matched advanced HGSOC tissue samples. METHODS: Tumor samples from 124 primary HGSOC patients were retrospectively collected within the Tumor Bank Ovarian Cancer ( http://www.toc-network.de ). All patients were in advanced stages (FIGO stage III-IV). No patient had received anti-angiogenesis therapy. The cohort contains 62 long-term survivors and 62 controls matched by age and post-surgical tumor residuals. Long-term survivors were defined as patients with no relapse within 5 years after the end of first-line chemotherapy. Controls were patients who suffered from first relapse within 6-36 months after primary treatment. Samples were assessed for immunohistochemical expression of vascular endothelial growth factor (VEGF) A and VEGF receptor 2 (VEGFR2). Expression profiles of VEGFA and VEGFR2 were compared between the two groups. RESULTS: Significant correlation between VEGFA and VEGFR2 expression was observed (p < 0.0001, Spearman coefficient 0.347). A high expression of VEGFR2 (VEGFR2high) was found more frequently in long-term survivors (77.4%, 48/62) than in controls (51.6%, 30/62, p = 0.001), independent of FIGO stage and VEGFA expression in multivariate analysis (p = 0.005). Also, VEGFR2high was found the most frequently in women with PFS ≥ 10 years (p = 0.001) among all 124 patients. However, no significant association was detected between VEGFA expression and 5-year PFS (p = 0.075). CONCLUSIONS: VEGFR2 overexpression significantly correlated with long-term PFS in HGSOC patients, independent of age, FIGO stage, tumor residual and VEGFA expression.


Asunto(s)
Cistadenocarcinoma Seroso/metabolismo , Neoplasias Ováricas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Supervivientes de Cáncer , Cistadenocarcinoma Seroso/irrigación sanguínea , Cistadenocarcinoma Seroso/mortalidad , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Supervivencia sin Progresión , Factor A de Crecimiento Endotelial Vascular/biosíntesis
14.
J Neuropathol Exp Neurol ; 78(2): 172-180, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30615171

RESUMEN

Significant angiogenesis is one of the malignant features in astrocytomas. Cotransfactor Yes-associated protein/transcriptional coactivator with PDZ-binding motif (YAP/TAZ) is a major regulator of embryonic angiogenesis, in which it plays an essential role in vascular tip cell migration, blood vessel formation, and vascular barrier maturation. We quantified TAZ expression on blood vessels and parenchyma of astrocytomas of varying malignancy to investigate its role in tumor angiogenesis. Replicating others' findings, we observed that TAZ is expressed in tumor cells but also in vascular cells. TAZ expression in both cell types was correlated with malignant grade. Immunofluorescence staining for TAZ, smooth muscle actin, and CD31 verified that TAZ-expressing vascular cells are endothelial cells, not pericytes. Analysis of blood vessel density using CD31 immunolabeling revealed that endothelial cell TAZ immunoreactivity was positively correlated with blood vessel density. MRI-acquired tumor blood perfusion measurements in 12 pre-excision glioblastomas and subsequent postexcision TAZ staining supported that TAZ immunoreactivity-blood vessel density correlation with blood perfusion. In glioblastoma, TAZ staining was denser in glomerular neovascularization than that in the thin-walled neovascularization. TAZ expression was also correlated with vascular endothelial growth factor 2 (VEGFR2) immunoreactivity on endothelial cells. Our results indicate that VEGFR2/TAZ signaling pathway plays an important role in angiogenesis in astrocytomas.


Asunto(s)
Astrocitoma/patología , Neoplasias Encefálicas/patología , Neovascularización Patológica/patología , Factores de Transcripción/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/análisis , Niño , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Estudios Retrospectivos , Transactivadores , Factores de Transcripción/análisis , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Adulto Joven
15.
FASEB J ; 33(3): 4626-4637, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30576228

RESUMEN

Aggregated amyloid ß (Aß) peptides in the Alzheimer's disease (AD) brain are hypothesized to trigger several downstream pathologies, including cerebrovascular dysfunction. Previous studies have shown that Aß peptides can have antiangiogenic properties, which may contribute to vascular dysfunction in the early stages of the disease process. We have generated data showing that brain endothelial cells (ECs) exposed to toxic Aß1-42 oligomers can readily enter a senescence phenotype. To determine the effect of Aß oligomers on brain ECs, we treated early passaged human brain microvascular ECs and HUVECs with high MW Aß1-42 oligomers (5 µM, for 72 h). For controls, we used no peptide treatment, 5 µM Aß1-42 monomers, and 5 µM Aß1-42 fibrils, respectively. Brain ECs treated with Aß1-42 oligomers showed increased senescence-associated ß-galactosidase staining and increased senescence-associated p21/p53 expression. Treatment with either Aß1-42 monomer or Aß1-42 fibrils did not induce senescence in this assay. We then measured vascular endothelial growth factor receptor (VEGFR) expression in the Aß1-42 oligomer-treated ECs, and these cells showed significantly increased VEGFR-1 expression and decreased VEGFR-2 levels. Overexpression of VEGFR-1 in brain ECs readily induced senescence, suggesting a direct role of VEGFR-1 signaling events in this paradigm. More importantly, small interfering RNA-mediated knockdown of VEGFR-1 expression in brain ECs was able to prevent up-regulation of p21 protein expression and significantly reduced induction of senescence following Aß1-42 oligomer treatment. Our studies show that exposure to Aß1-42 oligomers may impair vascular functions by altering VEGFR-1 expression and causing ECs to enter a senescent phenotype. Altered VEGFR expression has been documented in brains of AD patients and suggests that this pathway may play a role in AD disease pathogenesis. These studies suggest that modulating VEGFR-1 expression and signaling events could potentially prevent senescence and rejuvenate EC functions, and provides us with a novel target to pursue for prevention and treatment of cerebrovascular dysfunction in AD.-Angom, R. S., Wang, Y., Wang, E., Pal, K., Bhattacharya, S., Watzlawik, J. O., Rosenberry, T. L., Das, P., Mukhopadhyay, D. VEGF receptor-1 modulates amyloid ß 1-42 oligomer-induced senescence in brain endothelial cells.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Senescencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Encéfalo/irrigación sanguínea , Capilares/citología , Supervivencia Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Células Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
16.
Mol Reprod Dev ; 86(2): 156-165, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30431677

RESUMEN

In the present study, we investigated the temporal relationship between angiogenic and antiangiogenic vascular endothelial growth factor isoforms (VEGFxxxa and VEGFxxxb, respectively), the receptors VEGFR1 and VEGFR2, their soluble forms, and the kinases and the splicing factors regulating the synthesis of VEGF isoforms in healthy and atretic antral follicles. The results show a higher (p < 0.05) messenger RNA (mRNA) expression of VEGF120a, VEGF164a, and VEGF120b in healthy than in atretic follicles, but the mRNA expression of VEGF164b was not detected. The mRNA of serine-arginine protein kinase 1 ( SRPK1) was higher ( p < 0.05) in large healthy follicles than in large atretic follicles. In contrast, atretic follicles had higher mRNA expression of a soluble form of the receptor 2 of VEGF ( sVEGFR2) than healthy follicles ( p < 0.05). Additionally, we observed a positive relationship ( p < 0.05) between SRPK1 and serine-arginine-rich splicing factor 1 ( SRSF1) with the angiogenic isoforms VEGF120a and VEGF164a and between CDC-like kinases-1 ( CLK1) and SRSF6 with the antiangiogenic VEGF120b isoform. Principal components analysis (PCA) resulted in two PC explaining 71% of the variation, which was formed by the VEGF isoforms, the kinases and the splicing factor (PC1) and by the VEGF receptors (PC2). When PC analysis was carried out within follicular health status, there were no differences for PC1 between follicular status, whereas PC2 differed between healthy and atretic follicles. In conclusion, the higher mRNA expression for VEGF120a and VEGF164a, the low expression of sVEGFR2, and absent expression of mRNA for VEGF164b provide evidence of a proangiogenic autocrine milieu to support granulosa cells during follicle development.


Asunto(s)
Comunicación Autocrina/fisiología , Regulación de la Expresión Génica/fisiología , Células de la Granulosa/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Bovinos , Femenino , Células de la Granulosa/citología
17.
Curr Mol Med ; 18(5): 273-286, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30289073

RESUMEN

BACKGROUND: Whereas retinal pigment epithelial (RPE) cells are known to secrete VEGF-A and VEGFR2, the functions of the autocrine VEGF signaling remain unclear. Meanwhile, anti-VEGF therapies have been applied routinely to treat ocular vascular diseases. OBJECTIVE: The aim of this study was to determine the functions of the VEGF signaling in RPE cells and evaluate the consequences of its interruption. METHODS: The genes involved in the VEGF and Hippo signal pathways were knocked down with siRNAs in both ARPE-19 cell line and human primary RPE cells via transient transfection whereas overexpression of VEGFR2 was mediated via adenovirus transduction. Expression of the epithelial-mesenchymal transition (EMT) markers and the downstream genes of YAP were determined by real-time PCR and Western Blot analysis. Immunofluorescence staining was utilized to determine gene expression in tissue and mouse samples. RESULTS: Knockdown of VEGFR2 results in epithelial-mesenchymal transition in vitro and in vivo. Overexpression of VEGFR2 suppresses TGF ß-mediated EMT in RPE cells. Loss of VEGF-C rather than VEGF-A induces EMT. Mechanistically, the VEGFR2 ablation-induced EMT in RPE cells is mediated by activation of YAP, an effector of the Hippo pathway. Finally, the immunohistochemical analysis of VEGFR2 and YAP in human proliferative vitreoretinopathy (PVR) membranes indicates a tendency of an inverse correlation between VEGFR2-positive and YAP-positive cells. CONCLUSIONS: Our results disclose unexpected novel roles of VEGFR2 and VEGF-C in the process of EMT of RPE cells and in the Hippo pathway. The data shown here demonstrated that VEGFR2 and VEGF-C are important to maintain the normal physiological state of RPE cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica , Fosfoproteínas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Fosfoproteínas/genética , Epitelio Pigmentado de la Retina/citología , Factores de Transcripción , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas Señalizadoras YAP
18.
Sci Rep ; 8(1): 11844, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30087428

RESUMEN

To investigate the ability of CT texture analysis to assess and predict the expression statuses of E-cadherin, Ki67, VEGFR2 and EGFR in gastric cancers, the enhanced CT images of 139 patients with gastric cancer were retrospectively reviewed. The region of interest was manually drawn along the margin of the lesion on the largest slice in the arterial and venous phases, which yielded a series of texture parameters. Our results showed that the standard deviation, width, entropy, entropy (H), correlation and contrast from the arterial and venous phases were significantly correlated with the E-cadherin expression level in gastric cancers (all P < 0.05). The skewness from the arterial phase and the mean and autocorrelation from the venous phase were negatively correlated with the Ki67 expression level in gastric cancers (all P < 0.05). The width, entropy and contrast from the venous phase were positively correlated with the VEGFR2 expression level in gastric cancers (all P < 0.05). No significant correlation was found between the texture features and EGFR expression level. CT texture analysis, which had areas under the receiver operating characteristic curve (AUCs) ranging from 0.612 to 0.715, holds promise in predicting E-cadherin, Ki67 and VEGFR2 expression levels in gastric cancers.


Asunto(s)
Biomarcadores/metabolismo , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/metabolismo , Tomografía Computarizada por Rayos X/métodos , Cadherinas/biosíntesis , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/biosíntesis , Masculino , Persona de Mediana Edad , Pronóstico , Curva ROC , Estudios Retrospectivos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
19.
J Tissue Eng Regen Med ; 12(11): 2203-2220, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30062712

RESUMEN

Oxidative stress, induced by harmful levels of reactive oxygen species, is a common occurrence that impairs proper bone defect vascular healing through the impairment of endothelial cell function. Ionic silicon released from silica-based biomaterials, can upregulate hypoxia-inducible factor-1α (HIF-1α). Yet it is unclear whether ionic Si can restore endothelial cell function under oxidative stress conditions. Therefore, we hypothesized that ionic silicon can help improve human umbilical vein endothelial cells' (HUVECs') survival under toxic oxidative stress. In this study, we evaluated the ionic jsilicon effect on HUVECs viability, proliferation, migration, gene expression, and capillary tube formation under normal conditions and under harmful hydrogen peroxide levels. We demonstrated that 0.5-mM Si4+ significantly enhanced angiogenesis in HUVECs under normal condition (p < 0.05). HUVECs exposed to 0.5-mM Si4+ presented a morphological change, even without the bed of Matrigel, and formed significantly more tube-like structures than the control (p < 0.001). In addition, 0.5-mM Si4+ enhanced cell viability in HUVECs under harmful H2 O2 levels. HIF-1α, vascular endothelial growth factor-A, and vascular endothelial growth factor receptor-2 were overexpressed more than twofold in silicon-treated HUVECs, under normal and toxic H2 O2 conditions. Moreover, the HUVECs were treated with 0.5-mM Si4+ overexpressed superoxide dismutase-1 (SOD-1), catalase-1 (Cat-1), and nitric oxide synthase-3 (NOS3) under normal and oxidative stress environment (p < 0.01). A computational model was used for explaining the antioxidant effect of Si4+ in endothelial cells and human periosteum cells by SOD-1 enhancement. In conclusion, we demonstrated that 0.5-mM Si4+ can recover the HUVECs' viability under oxidative stress conditions by reducing cell death and upregulating expression of angiogenic and antioxidant factors.


Asunto(s)
Materiales Biocompatibles , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Peróxido de Hidrógeno/efectos adversos , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/biosíntesis , Silicatos , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Apoptosis/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Mitocondriales , Proteínas de Neoplasias/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Silicatos/química , Silicatos/farmacología , Silicio/química , Silicio/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis
20.
PLoS One ; 13(8): e0201395, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30125282

RESUMEN

Various angiogenic factors have been shown to play important roles in intraplaque angiogenesis, while little is known about the dynamic expression change and interplay between various angiogenic factors and intraplaque angiogenesis under high cholesterol conditions. New Zealand rabbits underwent balloon injury of the abdominal artery and then were assigned to a control group (n = 15, normal chow) or high cholesterol group (n = 25, 1% high cholesterol diet). At weeks 4, 6, 8, 10, and 12 after acclimation, rabbits (high cholesterol group, n = 5; control group, n = 3) were euthanized. No lesions were observed in the control group. From week 4 to week 12, the expression of vascular endothelial growth factor A (VEGF-A), VEGF receptor 2 (VEGFR-2), fibroblast growth factor 2 (FGF-2), FGF receptor 1 (FGFR-1), platelet-derived growth factor-BB (PDGF-BB), and tumor necrosis factor alpha (TNF-α), the vulnerability index (VI) and the microvessel density (MVD) were significantly elevated in the high cholesterol group; however, PDGF receptor ß (PDGFR-ß) expression showed little change. Analysis by double-label immunofluorescence (CD31 and Ng2) and FITC-dextran indicated that the neovessels within the plaque were leaky due to a lack of pericytes. As indicated by Pearson's correlation analysis, there was a highly positive correlation between the VI, MVD, macrophage content, and TNF-α level, and the levels of VEGF-A/VEGFR-2 and FGF-2/FGFR-1. However, no correlations were observed between PDGFR-ß levels and the VI or MVD. High expression of VEGF-A/VEGFR-2 and FGF-2/FGFR-1 but not of PDGF-BB/PDGFR-ß may contribute to immature and inflammatory intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Neovascularización Patológica/metabolismo , Placa Aterosclerótica/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Animales , Aterosclerosis/patología , Becaplermina/biosíntesis , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/patología , Masculino , Neovascularización Patológica/patología , Pericitos/metabolismo , Pericitos/patología , Placa Aterosclerótica/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA