Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 327
Filtrar
1.
Int Immunopharmacol ; 127: 111362, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38103411

RESUMEN

Melanoma, the most perilous form of skin cancer, is known for its inherent resistance to chemotherapy. Even with advances in tumor immunotherapy, the survival of patients with advanced or recurrent melanomas remains poor. Over time, melanoma tumor cells may produce excessive angiogenic factors, necessitating the use of combinations of angiogenesis inhibitors, including broad-spectrum options, to combat melanoma. Among these inhibitors, Endostatin is one of the most broad-spectrum and least toxic angiogenesis inhibitors. We found Endostatin significantly increased the infiltration of CD8+ T cells and reduced the infiltration of M2 tumor-associated macrophages (TAMs) in the melanoma tumor microenvironment (TME). Interestingly, we also observed high expression levels of programmed death 1 (PD-1), an essential immune checkpoint molecule associated with tumor immune evasion, within the melanoma tumor microenvironment despite the use of Endostatin. To address this issue, we investigated the effects of a plasmid expressing Endostatin and PD-1 siRNA, wherein Endostatin was overexpressed while RNA interference (RNAi) targeted PD-1. These therapeutic agents were delivered using attenuated Salmonella in melanoma-bearing mice. Our results demonstrate that pEndostatin-siRNA-PD-1 therapy exhibits optimal therapeutic efficacy against melanoma. We found that pEndostatin-siRNA-PD-1 therapy promotes the infiltration of CD8+ T cells and the expression of granzyme B in melanoma tumors. Importantly, combined inhibition of angiogenesis and PD-1 significantly suppresses melanoma tumor progression compared with the inhibition of angiogenesis or PD-1 alone. Based on these findings, our study suggests that combining PD-1 inhibition with angiogenesis inhibitors holds promise as a clinical strategy for the treatment of melanoma.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Ratones , Animales , Endostatinas/genética , Endostatinas/uso terapéutico , Endostatinas/metabolismo , Receptor de Muerte Celular Programada 1/genética , Factor A de Crecimiento Endotelial Vascular/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Linfocitos T CD8-positivos/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Cutáneas/terapia , Neoplasias Cutáneas/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Plásmidos , Salmonella/genética , Microambiente Tumoral
2.
Cell Death Dis ; 13(12): 1019, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36470872

RESUMEN

RNA binding protein HuD plays essential roles in gene expression by regulating RNA metabolism, and its dysregulation is involved in the pathogenesis of several diseases, including tumors, neurodegenerative diseases, and diabetes. Here, we explored HuD-mediated differential expression of secretory proteins in mouse insulinoma ßTC6 cells using a cytokine array. Endostatin and Serpin E1 that play anti-angiogenic roles were identified as differentially expressed proteins by HuD. HuD knockdown increased the expression of α chain of collagen XVIII (Col18a1), a precursor form of endostatin, and Serpin E1 by associating with the 3'-untranslated regions (UTRs) of Col18a1 and Serpin E1 mRNAs. Reporter analysis revealed that HuD knockdown increased the translation of EGFP reporters containing 3'UTRs of Col18a1 and Serpin E1 mRNAs, which suggests the role of HuD as a translational repressor. Co-cultures of ßTC6 cells and pancreatic islet endothelial MS1 cells were used to assess the crosstalk between ß cells and islet endothelial cells, and the results showed that HuD downregulation in ßTC6 cells inhibited the growth and migration of MS1 cells. Ectopic expression of HuD decreased Col18a1 and Serpin E1 expression, while increasing the markers of islet vascular cells in the pancreas of db/db mice. Taken together, these results suggest that HuD has the potential to regulate the crosstalk between ß cells and islet endothelial cells by regulating Endostatin and Serpin E1 expression, thereby contributing to the maintenance of homeostasis in the islet microenvironment.


Asunto(s)
Proteína 4 Similar a ELAV , Endostatinas , Células Secretoras de Insulina , Inhibidor 1 de Activador Plasminogénico , Animales , Ratones , Regiones no Traducidas 3'/genética , Endostatinas/genética , Endostatinas/metabolismo , Células Endoteliales/metabolismo , Células Secretoras de Insulina/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Proteína 4 Similar a ELAV/genética , Proteína 4 Similar a ELAV/metabolismo
3.
Curr Gene Ther ; 22(5): 417-426, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35382718

RESUMEN

BACKGROUND: Wet age-related macular degeneration (wAMD) is characterized by the presence of choroidal neovascularization (CNV). Although there are some clinical drugs targeting vascular endothelial growth factor (VEGF) and inhibiting CNV, two major side effects limit their application, including the excessive activity of anti-VEGF and frequent intraocular injections. To explore better treatment strategies, researchers developed a hypoxic modulator retinal pigment epithelium (RPE)- specific adeno-associated virus (AAV) vector expressing endostatin to inhibit CNV. However, the mechanism of endostatin is complex. Instead, soluble fms-like tyrosine kinase-1 (sFlt-1) can inhibit VEGF-induced angiogenesis through two simple and clear mechanisms, giving rise to sequestration of VEGF and forming an inactive heterodimer with the membrane-spanning isoforms of the VEGF receptor Flt-1 and kinase insert domain-containing receptor. OBJECTIVE: In this study, we chose sFlt-1 as a safer substitute to treat wAMD by inhibiting VEGFinduced angiogenesis. METHODS: The AAV2/8-Y733F-REG-RPE-sFlt-1 vector was delivered by intravitreal injection to the eyes of mice. AAV2/8-Y733F vector is a mutant of the AAV2/8 vector, and the REG-RPE promoter is a hypoxia-regulated RPE-specific promoter. Two animal models were used to evaluate the function of the vector. RESULTS: In the cobalt chloride-induced hypoxia model, the results demonstrated that the AAV2/8- Y733F-REG-RPE-sFlt-1 vector induced the expression of the sFlt-1 gene in RPE cells through hypoxia. In the laser-induced CNV model, the results demonstrated that the AAV2/8-Y733F-REG-RPE-sFlt- 1 vector reduced laser-induced CNV. CONCLUSION: Hypoxia regulated, RPE-specific AAV vector-mediated sFlt-1 gene is a hypoxiaregulated antiangiogenic vector for wAMD.


Asunto(s)
Neovascularización Coroidal , Animales , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/terapia , Modelos Animales de Enfermedad , Endostatinas/genética , Endostatinas/metabolismo , Endostatinas/farmacología , Terapia Genética/métodos , Hipoxia/metabolismo , Hipoxia/terapia , Ratones , Epitelio Pigmentado de la Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/farmacología
4.
BMC Cardiovasc Disord ; 22(1): 97, 2022 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-35279096

RESUMEN

PURPOSE: The aim of the present study was to investigate the efficacy of recombinant human endostatin (ES) (rh-ES) combined with radiation on rat cardiomyocyte apoptosis and the regulatory mechanism of transforming growth factor beta1 (TGF-ß1)/Sma and Mad-related protein 3 (Smad3)/connective tissue growth factor (CTGF) signaling. METHOD: The primary cardiomyocytes were isolated from neonatal Sprague-Dawley rats for culture in vitro and divided into blank control group (without treatment), 10 Gy radiation + siTGF-ß1 siRNA (gene silencing) group, ES + siTGF-ß1 siRNA group, and 10 Gy radiation + ES + siTGF-ß1 siRNA group. Methyl thiazolyl tetrazolium assay was used to calculate the half-maximal inhibitory concentration (IC50) of rh-ES on cardiomyocytes. Adenoviral vector was constructed for virus packaging to silence TGF-ß1 expression in cardiomyocytes. Quantitative real-time polymerase chain reaction and Western blot were carried out to analyze TGF-ß1, Smad2, Smad3 and CTGF expression at both gene and protein levels. Flow cytometry and electron microscope were used to examine cell apoptosis. RESULTS: ES had a dose-dependent inhibitory effect on the proliferation of primary rat cardiomyocytes. ES combined with radiotherapy significantly inhibited cardiomyocyte proliferation and promoted cell apoptosis (P < 0.01). The gene and protein expression of TGF-ß1, Smad2, Smad3 and CTGF were significantly up-regulated in primary cardiomyocytes transfected with TGF-ß1 gene (P < 0.05). CONCLUSION: The combination therapy with rh-ES and radiation can promote cardiomyocyte apoptosis and aggravate myocardial cell damage via TGF-ß1/Smad3/CTGF signaling pathway.


Asunto(s)
Miocitos Cardíacos , Factor de Crecimiento Transformador beta1 , Animales , Apoptosis , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Endostatinas/genética , Endostatinas/metabolismo , Endostatinas/farmacología , Humanos , Miocitos Cardíacos/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína smad3/farmacología , Factor de Crecimiento Transformador beta1/metabolismo
5.
Biol Pharm Bull ; 45(3): 309-315, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937830

RESUMEN

Anti-angiogenic gene therapy is a promising strategy in treating cancer. Endostatin and angiostatin are widely used in tumor anti-angiogenesis therapy. Our previous studies have shown that the BDS-hEA, a baculovirus long-term expressing the fusion protein of human endostatin and angiostatin, has a favorable effect in inhibiting the growth and angiogenesis of hepatocellular carcinoma. The purpose of this study was to further investigate its synergistic antitumor efficiency in combination with low-dose chemotherapeutic gemcitabine (GEM) on the subcutaneous hepatocellular carcinoma xenograft model in nude mice. The results showed that the combined group significantly inhibited (p < 0.05 or p < 0.01 or p < 0.001) the growth of tumor weight and volume, reduced the expression of ki67 (cell proliferation marker), CD31 (angiogenic marker) and Matrix metalloproteinase 9 (MMP-9, tumor invasion and metastasis marker) and increased the apoptosis of tumor cells compared with the monotherapy and control groups, respectively. Synergistic index results showed that BDS-hEA combined with GEM had a synergistic effect in inhibiting tumor volume, proliferation, microvessel density, metastasis and promoting tumor apoptosis. Furthermore, there were no metastatic nodules and obvious pathological changes in liver tissue of the combined group, and the serum liver function indicators aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (T-BIL), alkaline phosphatase (ALP) and glutamyl transpeptidase (GGT) were significantly reduced (p < 0.05 or p < 0.01 or p < 0.001) in the BDS-hEA or GEM groups compared with the control group. Notably, the combined therapy showed lower levels of liver function indicators than the GEM group. These data support the view that the combination of BDS-hEA and GEM has a synergistic anti-tumor properties and can reduce the damage of liver to certain extent.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Inhibidores de la Angiogénesis/uso terapéutico , Angiostatinas/genética , Angiostatinas/uso terapéutico , Animales , Baculoviridae , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Desoxicitidina/análogos & derivados , Endostatinas/genética , Endostatinas/uso terapéutico , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Desnudos , Gemcitabina
6.
Horm Metab Res ; 53(7): 470-477, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34282598

RESUMEN

Angiogenesis is a multistep process implicated in the pathophysiology and progression of diabetic nephropathy (DN). Angiotensin-converting enzyme inhibitors (ACEI) and calcium channel blockers (CCB) have an important role in DN. We performed a randomized-controlled trial of lisinopril alone (an ACEI) or in combination with verapamil (a CCB) as a therapy for DN in type 2 diabetes mellitus (T2DM) patients with hypertension (HTN) and urinary albumin creatinine ratio (UACR) (30-300 mg/g) also to evaluate their effect on UACR, the angiogenic proteins: Angiopoietin 2 (Ang-2) and Endostatin (EST). Forty T2DM patients with microalbuminuria, aged 45-65 years were included. Patients were randomly assigned into group 1 receiving oral lisinopril and group 2 receiving oral lisinopril and verapamil once daily. After 3 months follow-up fasting blood glucose (FPG), HbA1c, lipid profile, UACR, serum urea and creatinine levels were assessed. EST and Ang-2 were measured using ELISA technique. Baseline Ang-2 and EST levels were elevated in both groups compared with controls (p<0.001). After follow-up, group 2 had significantly decreased FPG, HbA1c, UACR, EST and Ang-2 compared with their baseline levels (p<0.001 for all comparisons) and with group 1 (p<0.001). No adverse reactions were reported. Baseline EST and Ang-2 were positively correlated to UACR (r=0.753, p<0.001) (r=0.685, p<0.001). Lisinopril/verapamil combination enhanced glycemic control and kidney function via diminishing EST and Ang-2. This combination can be considered as a safe and effective approach for early stage nephropathy therapy in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/tratamiento farmacológico , Endostatinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipertensión/fisiopatología , Lisinopril/farmacología , Verapamilo/farmacología , Proteínas de Transporte Vesicular/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Estudios de Casos y Controles , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Quimioterapia Combinada , Endostatinas/genética , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Vasodilatadores/farmacología , Proteínas de Transporte Vesicular/genética
7.
Cancer Lett ; 509: 26-38, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33819529

RESUMEN

Oncolytic adenovirus-mediated gene therapy shows promise for cancer treatment; however, the systemic delivery of oncolytic adenovirus to tumors remains challenging. Recently, mesenchymal stem cells (MSCs) have emerged as potential vehicles for improving delivery. Yet, because the oncolytic adenovirus replicates in MSCs, balancing MSC viability with viral load is key to achieving optimal therapeutic effect. We thus developed an all-in-one Tet-on system that can regulate replication of oncolytic adenovirus. Then, we loaded the novel oncolytic adenovirus carrying interleukin (IL)-24 and/or Endostatin in human umbilical cord blood-mesenchymal stem cells (hUCB-MSCs) for glioma therapy. In vitro assays demonstrated that this novel oncolytic adenovirus could efficiently replicate and kill glioma cells while sparing normal cells. Moreover, doxycycline effectively regulated oncolytic adenovirus replication in the hUCB-MSCs. The doxycycline induction group with dual expression of IL-24 and Endostatin exhibited significantly greater antitumor effects than other groups in a xenograft model of glioma. Thus, this strategy for systemic delivery of oncolytic adenovirus with its oncolytic activity controlled by a Tet-on system is a promising method for achieving antitumor efficacy in glioma, especially for metastatic tumors.


Asunto(s)
Neoplasias Encefálicas/terapia , Trasplante de Células Madre de Sangre del Cordón Umbilical , Endostatinas/biosíntesis , Terapia Genética , Glioma/terapia , Interleucinas/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Viroterapia Oncolítica , Virus Oncolíticos/genética , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Muerte Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Endostatinas/genética , Femenino , Vectores Genéticos , Glioma/genética , Glioma/metabolismo , Glioma/virología , Humanos , Interleucinas/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Virus Oncolíticos/crecimiento & desarrollo , Carga Tumoral , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Int J Nanomedicine ; 15: 8875-8892, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33209022

RESUMEN

PURPOSE: Vascular endothelial growth factor receptor 2 (VEGFR-2) and neuropilin-1 (NRP-1) are two prominent synergistic receptors overexpressed on new blood vessels in glioma and may be promising targets for antiglioma therapy. The aim of this study was to design a dual receptor targeting and blood-brain barrier (BBB) penetrating peptide-modified polyethyleneimine (PEI) nanocomplex that can efficiently deliver the angiogenesis-inhibiting secretory endostatin gene (pVAXI-En) to treat glioma. MATERIALS AND METHODS: We first constructed the tandem peptide TAT-AT7 by conjugating AT7 to TAT and evaluated its binding affinity to VEGFR-2 and NRP-1, vasculature-targeting ability and BBB crossing capacity. Then, TAT-AT7-modified PEI polymer (PPTA) was synthesized, and a pVAXI-En-loaded PPTA nanocomplex (PPTA/pVAXI-En) was prepared. The physicochemical properties, cytotoxicity, transfection efficiency, capacities to cross the BBB and BTB (blood-tumor barrier) and glioma-targeting properties of PPTA/pVAXI-En were investigated. Moreover, the in vivo anti-angiogenic behaviors and anti-glioma effects of PPTA/pVAXI-En were evaluated in nude mice. RESULTS: The binding affinity of TAT-AT7 to VEGFR-2 and NRP-1 was approximately 3 to 10 times greater than that of AT7 or TAT. The cellular uptake of TAT-AT7 in endothelial cells was 5-fold and 119-fold greater than that of TAT and AT7 alone, respectively. TAT-AT7 also displayed remarkable efficiency in penetrating the BBB and glioma tissue in vivo. PPTA/pVAXI-En exhibited lower cytotoxicity, stronger BBB and BTB traversing abilities, higher selective glioma targeting and better gene transfection efficiency than PEI/pVAXI-En. More importantly, PPTA/pVAXI-En significantly suppressed the tube formation and migration of endothelial cells, inhibited glioma growth, and reduced the microvasculature in orthotopic U87 glioma-bearing nude mice. CONCLUSION: Our study demonstrates that PPTA/pVAXI-En can be exploited as an efficient dual-targeting nanocomplex to cross the BBB and BTB, and hence it represents a feasible and promising nonviral gene delivery system for effective glioma therapy.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Endostatinas/genética , Glioma/patología , Nanoestructuras/química , Neuropilina-1/metabolismo , Péptidos/química , Polietileneimina/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Portadores de Fármacos/química , Endostatinas/química , Terapia Genética , Glioma/genética , Glioma/terapia , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Permeabilidad , Transfección
9.
BMC Mol Cell Biol ; 21(1): 57, 2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727534

RESUMEN

BACKGROUND: Transplantation of gene transfected endothelial progenitor cells (EPCs) has provided novel methods for tumor neovascularization therapy but not for ocular disease therapy. This study aimed to investigate the efficacy of endostatin transfected EPCs in retinal neovascularization therapy. RESULTS: Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) showed the high expression of endostatin in endostatin-lentivirus-EPCs. The neovascularization leakage area and the number of preretinal neovascular cell nuclei were significantly decreased in the endostatin-lentivirus and endostatin-lentivirus-EPC groups, and the effects of these two treatments on inhibiting retinal neovascularization were almost the same. These two groups also showed the greater retinal distribution of endostatin. Intravitreal injections of endostatin-lentivirus-EPCs inhibited retinal neovascularization, vascular endothelial growth factor (VEGF) and CD31 expression, and increased endostatin expression in vivo. Endostatin-lentivirus-EPCs targeted and prevented pathologic retinal neovascularization. CONCLUSIONS: Gene-combined EPCs represent a potential new therapeutic agent for the treatment of neovascular eye diseases.


Asunto(s)
Endostatinas/genética , Endostatinas/uso terapéutico , Terapia Genética , Lentivirus/genética , Neovascularización Retiniana/terapia , Animales , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Angiografía con Fluoresceína , Fondo de Ojo , Oxígeno , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Factores de Crecimiento Endotelial Vascular/metabolismo
10.
Biomed Pharmacother ; 123: 109765, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31846843

RESUMEN

Baculovirus (BV) is a potential gene delivery vector but only mediates transient transgene expression and easily inactivated by human complement. To this end, we intend to develop a novel bivalent BV vector for complement resistance and sustained transgene expression, and evaluate its effect in anti-angiogenesis gene therapy. The results showed that the hybrid bivalent BV significantly prolonged the expression of enhanced green fluorescent protein (eGFP) in vitro for at least 90 days at over 109 a.u. total fluorescence intensity, and exhibited significantly higher complement resistance. The control BV-mediated eGFP expression gradually declined within 15 days and showed lower transduction efficiency. In vivo studies confirmed that the hybrid bivalent BV exhibited longer duration of eGFP expression and higher transduction efficacy than the control BVs. Based on these findings, we further constructed a hybrid BV expressing the antiangiogenic fusion protein containing human endostatin and angiostatin (hEA). The hybrid BV-expressed hEA significantly prolonged the expression level of hEA with enhanced anti-angiogenic activities compared to the control groups, as evidenced by ELISA, cell proliferation, migration and tubular formation assays. With the stable expression of hEA, the hybrid BV conferred hEA more significant inhibitory effect on hepatocellular carcinoma tumor growth and significantly extended the life span of mice. These data implicate that the SB-based BV surface display system may have broad prospects as a novel platform for gene therapy of tumors.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Neovascularización Patológica/terapia , Angiostatinas/genética , Animales , Baculoviridae , Carcinoma Hepatocelular/patología , Endostatinas/genética , Proteínas Fluorescentes Verdes , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión , Spodoptera , Transducción Genética , Transgenes
11.
Int J Med Sci ; 16(7): 1032-1041, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31341417

RESUMEN

AP25 is an anti-tumor peptide with a high affinity for integrins. It exerts its anti-tumor activity by inhibiting angiogenesis and by directly inhibiting the growth of tumor cells. Its half-life time in vivo is only about 50 minutes, which limits its clinical application. In order to prolong the half-life time of AP25 while preserving its anti-tumor activity, several fusion proteins of AP25 and IgG4 Fc were designed and expressed; their anti-tumor activity and pharmacokinetics properties were evaluated. Firstly, four AP25-Fc fusion protein sequences were designed, and the corresponding proteins were expressed and purified. Based on the results of HUVEC migration inhibition assay, HUVEC and tumor cell proliferation inhibition assay and yields of expression by HEK293 cells, the fusion protein designated PSG4R was selected for further evaluation. The anti-tumor effect of PSG4R was then evaluated in vivo on HCT-116 nude mice xenograft model. And the pharmacokinetics properties of PSG4R were investigated in rats. The results showed that PSG4R could inhibit the growth of xenografts of human colon cancer cell line HCT-116 in nude mice by intravenous administration of 40 mg/kg once every two days. The half-life time of PSG4R was 56.270 ± 15.398 h. This study showed that the construction of AP25-Fc fusion protein could significantly prolong the half-life of AP25 while retaining its anti-tumor activity, which provides a new direction for new drug development of AP25.


Asunto(s)
Endostatinas/farmacología , Inmunoconjugados/farmacología , Inmunoglobulina G/farmacología , Neoplasias/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Administración Intravenosa , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Endostatinas/genética , Endostatinas/uso terapéutico , Femenino , Células HCT116 , Células HEK293 , Semivida , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoconjugados/genética , Inmunoconjugados/uso terapéutico , Inmunoglobulina G/genética , Inmunoglobulina G/uso terapéutico , Masculino , Ratones , Modelos Animales , Neoplasias/patología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Orphanet J Rare Dis ; 14(1): 72, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30922357

RESUMEN

Endostatin is a naturally occurring collagen fragment with anti-angiogenic properties. We investigated the association between serum endostatin levels and DLCO in a cohort of patients with lymphangioleiomyomatosis (LAM). Associations of endostatin levels to clinical features of LAM were explored using logistic regression models. Endostatin levels were associated with DLCO and were higher in subjects with TSC-associated LAM compared to sporadic LAM. These data suggest that endostatin could be a predictive biomarker of decline in DLCO and that germline mutational inactivation of the TSC1 or TSC2 gene is associated with higher endostatin levels. These findings could offer novel insights into the pathogenesis of LAM.


Asunto(s)
Biomarcadores/sangre , Endostatinas/sangre , Linfangioleiomiomatosis/sangre , Linfangioleiomiomatosis/fisiopatología , Adulto , Estudios de Cohortes , Endostatinas/genética , Femenino , Silenciador del Gen , Mutación de Línea Germinal , Humanos , Linfangioleiomiomatosis/complicaciones , Linfangioleiomiomatosis/genética , Persona de Mediana Edad , Esclerosis Tuberosa/complicaciones , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética
13.
Thorac Cancer ; 10(1): 90-95, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30485686

RESUMEN

BACKGROUND: Recombined humanized endostatin (Rh-endostatin) exhibits a potent anti-cancer effect involving multiple molecular targets and signaling pathways. HMGB1 is a highly conserved DNA-binding protein involved in cancer development. The therapeutic effect of Rh-endostatin on HMGB1 has not been reported, thus we investigate the effect in non-small cell lung cancer (NSCLC) cells. METHODS: Quantitative real-time PCR and Western blot were used to analyze the messenger RNA and protein expression of HMGB1 in A549 cancer cells, while enzyme-linked immunosorbent assay was used to detect the release of HMGB1. Western blot was performed to evaluate HMGB1 expression in SK-MES-1 and H661 NSCLC cells. RESULTS: Rh-endostatin inhibited the proliferation of A549 cancer cells and distinctly downregulated the expression and release of HMGB1 in dose and time dependent manners. Rh-endostatin-induced HMGB1 downregulation was confirmed in different types of NSCLC cells. CONCLUSION: These results demonstrate the general phenomenon that Rh-endostatin can induce HMGB1 suppression in a variety of NSCLC cells. Rh-endostatin may suppress HMGB1 expression and release in A549 cancer cells, thus inhibiting cell proliferation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Endostatinas/farmacología , Proteína HMGB1/genética , Proteínas Recombinantes/farmacología , Células A549 , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Endostatinas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína HMGB1/antagonistas & inhibidores , Humanos , Proteínas Recombinantes/genética , Transducción de Señal/efectos de los fármacos
14.
Pathol Res Pract ; 215(2): 315-321, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30554865

RESUMEN

AIM: To construct a lentiviral vector with endostatin (ES) and staphylococcal enterotoxin C3(SEC3) gene, and investigate its capacities of inhibition on proliferation and migration of Hela cells. METHODS: By inserting ES and SEC3 gene into the plasmid and then transfect 293 T cell, the co-expressed (SEC3-ES) vector were constructed. A series of experiments in vitro were carried out to detect its anti-tumor capacity. RESULTS: SEC3 expression of the vector is about 3 times of GV365-SEC3 vector, and ES expression is over 22.5-fold compared with GV365-ES vector. Moreover, OD490 value of CO group (1.212 ± 0.003) was notably lower than NC (negative control) group (1.124 ± 0.01) (P < 0.05) in MTT assay. Cell cycle analysis showed it could block Hela cells in S phase. Meanwhile, in wound healing assay, cells of CO group migrated at a slower rate (0.59 ± 0.02) compared with NC group (0.65 ± 0.02)(P < 0.01). CONCLUSION: The successful construction of co-expressed vector lays the foundation for further studies in vivo. These promising results suggest a new strategy to treating cervical cancer.


Asunto(s)
Antineoplásicos/farmacología , Endostatinas/genética , Enterotoxinas/genética , Terapia Genética/métodos , Vectores Genéticos , Movimiento Celular , Proliferación Celular , Femenino , Células HEK293 , Células HeLa , Humanos , Lentivirus , Transfección , Neoplasias del Cuello Uterino/virología
15.
J Mol Med (Berl) ; 96(10): 1107-1118, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30105447

RESUMEN

Aberrant growth of blood vessels in the choroid layer of the eye, termed choroidal neovascularization (CNV), is the pathological hallmark of exudative age-related macular degeneration (AMD), causing irreversible blindness among the elderly. Co-localization of proangiogenic factors and hypoxia inducible factors (HIF) in neovascular membranes from AMD eyes suggests the role of hypoxia in pathogenesis of CNV. In order to utilize hypoxic conditions in RPE for therapeutic purposes, we developed an optimized hypoxia regulated, RPE cell-specific gene therapy to inhibit choroidal neovascularization. An adeno-associated virus (AAV2) vector comprising a RPE-specific promoter and HIF-1 response elements (HRE) was designed to regulate production of human endostatin (a powerful angiostatic protein) in RPE. The vector was tested in a mouse model of laser-induced CNV using subretinal delivery. Spectral domain optical coherence tomography (SD-OCT) images from live mice and confocal images from lectin stained RPE flat mount sections demonstrated reduction in CNV areas by 80% compared to untreated eyes. Quantitative real-time polymerase chain reaction (qPCR) confirmed exogenous endostatin mRNA expression from the regulated vector that was significantly elevated 3, 7, and 14 days following laser treatment, but its expression was completely shut off after 45 days. Thus, RPE-specific, hypoxia-regulated delivery of anti-angiogenic proteins could be a valuable therapeutic approach to treat neovascular AMD at the time and in the ocular space where it arises. KEY POINTS: An optimized gene therapy vector targeting hypoxia and tissue-specific expression has been designed. The inhibitory role of gene therapy vector was tested in a mouse model of laser-induced CNV. An 80% reduction in choroidal neovascularization was achieved by the optimized vector. The expression of endostatin was limited to retinal pigment epithelium and regulated by hypoxia.


Asunto(s)
Neovascularización Coroidal/terapia , Terapia Genética , Hipoxia , Animales , Dependovirus , Endostatinas/genética , Endostatinas/metabolismo , Vectores Genéticos , Ratones Endogámicos C57BL , Parvovirinae/genética , Epitelio Pigmentado de la Retina/metabolismo
16.
Exp Cell Res ; 371(1): 50-62, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30055135

RESUMEN

DESI2 is a novel pro-apoptotic gene. We previously reported that DESI2 overexpression induces S phase arrest and apoptosis by activating checkpoint kinases. This work was to test whether the combination of endostatin, an endogenous antiangiogenic inhibitor, with DESI2 could improve the therapy efficacy in vitro and in vivo. The recombinant plasmid co-expressing DESI2 and endostatin was encapsulated with DOTAP/Cholesterol cationic liposome. Mice bearing CT26 colon carcinoma and LL2 lung cancer were treated with the DNA-liposome complex. We found that, in vitro, the combination of DESI2 and endostatin more efficiently inhibited proliferation of CT26, LL2, HCT116 and A549 cancer cells via apoptosis, as assessed by MTT assay, colony-formation assays, flow cytometric analysis, hoechst staining and activation of caspase-3, respectively. In addition, DESI2 overexpression caused up-regulation of RPS7, a substrate of DESI2 deubiquitination. Furthermore, siRNA targeting RPS7 partially abrogated, whereas RPS7 overexpression enhanced DESI2-induced inhibition of cell proliferation. Importantly, the combination also caused DNA lesions accumulation, which further promotes apoptosis. Mechanistic rationale suggested that endostatin first inhibits DNA-PKcs kinase, and partly abrogated DESI2-induced phosphorylation of DNA-PKcs, leading to increase of DNA damage, then contributes to DESI2-induced apoptosis. In vivo, the combined gene therapy more significantly inhibited tumor growth and efficiently prolonged the survival of tumor bearing mice than mono therapy. The improved antitumor effect was associated with inhibition of cell proliferation via apoptosis, as analyzed by TUNEL assay and PCNA immunostaining. The combination also inhibited angiogenesis, as assessed by alginate-encapsulated tumor cell assay and CD31 staining. Our data suggest that the combined gene therapy of DESI2 and endostatin can significantly enhance the antitumor activity as a DNA lesions accumulator, apoptosis inducer and angiogenesis inhibitor. The present study may provide a novel method for the treatment of cancer.


Asunto(s)
Liasas de Carbono-Nitrógeno/genética , Neoplasias del Colon/genética , Endostatinas/genética , Regulación Neoplásica de la Expresión Génica , Terapia Genética/métodos , Neoplasias Pulmonares/genética , Plásmidos/metabolismo , Células A549 , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Animales , Apoptosis/genética , Liasas de Carbono-Nitrógeno/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Proliferación Celular , Colesterol/química , Colesterol/metabolismo , Neoplasias del Colon/mortalidad , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Fragmentación del ADN , Endostatinas/metabolismo , Ácidos Grasos Monoinsaturados/química , Ácidos Grasos Monoinsaturados/metabolismo , Femenino , Células HCT116 , Humanos , Liposomas/administración & dosificación , Liposomas/química , Liposomas/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/química , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Ribosómicas/antagonistas & inhibidores , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Transducción de Señal , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Mol Sci ; 19(7)2018 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-29937525

RESUMEN

Chronic kidney disease (CKD) is a major global health problem. Unless intensive intervention is initiated, some patients can rapidly progress to end-stage kidney disease. However, it is often difficult to predict renal outcomes using conventional laboratory tests in individuals with CKD. Therefore, many researchers have been searching for novel biomarkers to predict the progression of CKD. Angiogenesis is involved in physiological and pathological processes in the kidney and is regulated by the balance between a proangiogenic factor, vascular endothelial growth factor (VEGF)-A, and various endogenous antiangiogenic factors. In recent reports using genetically engineered mice, the roles of these antiangiogenic factors in the pathogenesis of kidney disease have become increasingly clear. In addition, recent clinical studies have demonstrated associations between circulating levels of antiangiogenic factors and renal dysfunction in CKD patients. In this review, we summarize recent advances in the study of representative endogenous antiangiogenic factors, including soluble fms-related tyrosine kinase 1, soluble endoglin, pigment epithelium-derived factor, VEGF-A165b, endostatin, and vasohibin-1, in associations with kidney diseases and discuss their predictive potentials as biomarkers of progression of CKD.


Asunto(s)
Inhibidores de la Angiogénesis/genética , Proteínas del Ojo/genética , Fallo Renal Crónico/diagnóstico , Factores de Crecimiento Nervioso/genética , Insuficiencia Renal Crónica/diagnóstico , Serpinas/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Inductores de la Angiogénesis/química , Inductores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/metabolismo , Animales , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Progresión de la Enfermedad , Endostatinas/genética , Endostatinas/metabolismo , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica , Humanos , Fallo Renal Crónico/etiología , Fallo Renal Crónico/genética , Fallo Renal Crónico/patología , Ratones , Factores de Crecimiento Nervioso/metabolismo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Serpinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
J Exp Clin Cancer Res ; 37(1): 42, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499713

RESUMEN

BACKGROUND: Anti-CD105 mAb-conjugated immunoliposomes, loaded with secreted mouse endostatin gene, were developed for targeted tumor imaging and antiangiogenic gene therapy. METHODS: The liposomes were investigated for size, zeta-potential, lipid content, antibody binding ability, and pcDNA loading capacity. The ability of immunoliposomes to target tumor-derived endothelial cells and perform gene transfer in vitro was measured and their basic biocompatibility was evaluated. A nude mouse/breast cancer xenograft model was used to examine the tumor internalization of fluorescent-labeled liposomes and the clinical potential of immnuoliposomes loaded with pcDNA3.1-CSF1-endostatin. RESULTS: Loaded immunoliposomes were homogenously distributed with a well-defined spherical shape and bilayer, diameter of 122 ± 11 nm, and zeta potential + 1.40 mV. No significant differences were observed in body weight, liver index, oxidative stress, or liver and kidney function in mice after liposomes exposure. The addition of CD105 mAb to liposomes conferred the ability to target tumor-derived endothelial cells in vitro and in vivo. Systemic intravenous administration of fluorescent immunoliposomes in the xenograft model resulted in selective and efficient internalization in tumor vasculature. Treatment of mice with pcDNA3.1-CSF1-endostatin-loaded immunoliposomes suppressed tumor growth by 71%. CONCLUSIONS: These data demonstrate the advantages of using anti-CD105 mAb-conjugated immunoliposomes to enhance tumor targeting, imaging, and gene transfer applications.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Endoglina/antagonistas & inhibidores , Endostatinas/genética , Liposomas , Imagen Molecular , Neoplasias/diagnóstico , Neoplasias/genética , Animales , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Endostatinas/biosíntesis , Femenino , Técnicas de Transferencia de Gen , Terapia Genética , Glutatión/metabolismo , Humanos , Liposomas/química , Liposomas/ultraestructura , Ratones , Neoplasias/terapia , Imagen Óptica/métodos , Plásmidos/química , Plásmidos/genética , Superóxido Dismutasa/metabolismo , Espectrometría de Masas en Tándem , Transgenes
19.
Mol Med Rep ; 17(4): 5814-5820, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29484399

RESUMEN

The viability of endothelial progenitor cells (EPCs) as a therapeutic treatment for neovascularization (NV) was subject to investigation in the present study. Furthermore, endostatin has previously been demonstrated to be an inhibitor of angiogenesis and a suppressant of vascular leakage. The aim of the present study was to generate transgenic EPCs with anti­angiogenic effects for the treatment of ocular NV. EPCs were obtained from rat peripheral blood samples and then verified. A lentiviral­endostatin­green fluorescent protein recombinant construct was generated and used to infect EPCs. Transfected cells were then subjected to puromycin selection. Reverse transcription­quantitative polymerase chain reaction and a western blot assay were then applied in order to determine both the endostatin mRNA and protein expression levels, respectively. In addition, vascular endothelial growth factor (VEGF) expression levels were also detected in order to observe the anti­angiogenic effect of the endostatin­transfected EPCs. Following puromycin (1 µg/ml) selection for 4 days, a stable endostatin­transfected EPC line was generated. In this stable endostatin-transfected EPC line, the expression levels of endostatin increased; whereas the expression levels of VEGF decreased. The results of the present study revealed that EPCs can be genetically modified to overexpress endostatin, which may provide the cells with an anti­angiogenic effect via increased expression of endostatin and decreased expression of VEGF. Thus, EPCs genetically modified to overexpress endostatin may serve as a potential therapeutic agent for ocular NV treatment.


Asunto(s)
Endostatinas/genética , Células Progenitoras Endoteliales/metabolismo , Neovascularización Patológica/genética , Neovascularización Fisiológica/genética , Transducción Genética , Animales , Biomarcadores , Línea Celular , Células Progenitoras Endoteliales/citología , Femenino , Expresión Génica , Técnicas de Transferencia de Gen , Inmunofenotipificación , Masculino , Ratas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Tumour Biol ; 39(7): 1010428317708547, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28714365

RESUMEN

Several studies have revealed the potential of normalizing tumor vessels in anti-angiogenic treatment. Recombinant human endostatin is an anti-angiogenic agent which has been applied in clinical tumor treatment. Our previous research indicated that gold nanoparticles could be a nanoparticle carrier for recombinant human endostatin delivery. The recombinant human endostatin-gold nanoparticle conjugates normalized vessels, which improved chemotherapy. However, the mechanism of recombinant human endostatin-gold nanoparticle-induced vascular normalization has not been explored. Anterior gradient 2 has been reported to be over-expressed in many malignant tumors and involved in tumor angiogenesis. To date, the precise efficacy of recombinant human endostatin-gold nanoparticles on anterior gradient 2-mediated angiogenesis or anterior gradient 2-related signaling cohort remained unknown. In this study, we aimed to explore whether recombinant human endostatin-gold nanoparticles could normalize vessels in metastatic colorectal cancer xenografts, and we further elucidated whether recombinant human endostatin-gold nanoparticles could interrupt anterior gradient 2-induced angiogenesis. In vivo, it was indicated that recombinant human endostatin-gold nanoparticles increased pericyte expression while inhibit vascular endothelial growth factor receptor 2 and anterior gradient 2 expression in metastatic colorectal cancer xenografts. In vitro, we uncovered that recombinant human endostatin-gold nanoparticles reduced cell migration and tube formation induced by anterior gradient 2 in human umbilical vein endothelial cells. Treatment with recombinant human endostatin-gold nanoparticles attenuated anterior gradient 2-mediated activation of MMP2, cMyc, VE-cadherin, phosphorylation of p38, and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) in human umbilical vein endothelial cells. Our findings demonstrated recombinant human endostatin-gold nanoparticles might normalize vessels by interfering anterior gradient 2-mediated angiogenesis in metastatic colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Endostatinas/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Neovascularización Patológica/tratamiento farmacológico , Proteínas/genética , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Endostatinas/química , Endostatinas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Oro/administración & dosificación , Oro/química , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Nanopartículas del Metal/química , Ratones , Mucoproteínas , Metástasis de la Neoplasia , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas Oncogénicas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA