Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
Int J Mol Sci ; 24(24)2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38139016

RESUMEN

Müller cells play a critical role in the closure of macular holes, and their proliferation and migration are facilitated by the internal limiting membrane (ILM). Despite the importance of this process, the underlying molecular mechanism remains underexplored. This study investigated the effects of ILM components on the microRNA (miRNA) profile of Müller cells. Rat Müller cells (rMC-1) were cultured with a culture insert and varying concentrations of ILM component coatings, namely, collagen IV, laminin, and fibronectin, and cell migration was assessed by measuring cell-free areas in successive photographs following insert removal. MiRNAs were then extracted from these cells and analyzed. Mimics and inhibitors of miRNA candidates were transfected into Müller cells, and a cell migration assay and additional cell viability assays were performed. The results revealed that the ILM components promoted Müller cell migration (p < 0.01). Among the miRNA candidates, miR-194-3p was upregulated, whereas miR-125b-1-3p, miR-132-3p, miR-146b-5p, miR-152-3p, miR-196a-5p, miR-542-5p, miR-871-3p, miR-1839-5p, and miR-3573-3p were significantly downregulated (p < 0.05; fold change > 1.5). Moreover, miR-152-3p and miR-196a-5p reduced cell migration (p < 0.05) and proliferation (p < 0.001), and their suppressive effects were reversed by their respective inhibitors. In conclusion, miRNAs were regulated in ILM component-activated Müller cells, with miR-152-3p and miR-196a-5p regulating Müller cell migration and proliferation. These results serve as a basis for understanding the molecular healing process of macular holes and identifying potential new target genes in future research.


Asunto(s)
MicroARNs , Perforaciones de la Retina , Animales , Ratas , Colágeno Tipo IV/farmacología , Células Ependimogliales , Membranas , MicroARNs/genética , MicroARNs/farmacología , Perforaciones de la Retina/genética
2.
J Cancer Res Clin Oncol ; 149(8): 4477-4487, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36125535

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs) are major components of the tumor microenvironment (TME). Hypoxic TME is known to promote tumor progression. However, how a hypoxic condition regulates CAFs remains elusive. METHODS: To investigate the underlying mechanism involved in the regulation of gastric cancer (GC) progression by hypoxic CAFs, we performed secretome profiling. Normoxic or hypoxic CAFs conditioned media (CM) were filter-concentrated and in-gel trypsin digested. Resulting peptides were analyzed with LC-MS/MS. RESULTS: We observed that CM derived from hypoxic CAFs could promote migration of a panel of GC cell lines (AGS, SNU668, SNU638). Mass spectrometry analysis of hypoxic or normoxic CAFs CM identified 1595 proteins, of which 19 proteins (10 upregulated and 9 downregulated) were differentially expressed in the hypoxic secretome. We focused on COL4A2, whose expression was significantly decreased in hypoxic CAFs in HIF-1α-independent manner. Silencing of COL4A2 expression in normoxic CAFs phenocopied the effect of hypoxic CAFs in promoting GC cell migration. CONCLUSIONS: The reduced expression of COL4A2 in a hypoxic environment might be associated with the tumor-promoting role of hypoxic CAFs in GC.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Gástricas , Humanos , Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias Gástricas/patología , Línea Celular Tumoral , Cromatografía Liquida , Secretoma , Espectrometría de Masas en Tándem , Microambiente Tumoral , Fibroblastos/metabolismo , Proliferación Celular , Colágeno Tipo IV/metabolismo , Colágeno Tipo IV/farmacología
3.
Int J Mol Sci ; 23(16)2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-36012259

RESUMEN

Angiogenetic inhibitors are crucial in tumor therapy, and endogenous angiogenesis inhibitors have attracted considerable attention due to their effectiveness, safety, and multi-targeting ability. Arresten and canstatin, which have anti-angiogenesis effects, are the c-terminal fragments of the α1 and α2 chains of type IV collagen, respectively. In this study, human arresten and canstatin were recombinantly expressed in Escherichia coli (E. coli), and their effects on the proliferation, migration and tube formation of human umbilical vein endothelial cells (HUVECs) were evaluated. Regarding the cell cycle distribution test and 5-ethynyl-2'-deoxyuridine (EdU) assays, arresten and canstatin could repress the proliferation of HUVECs at a range of concentrations. Transwell assay indicated that the migration of HUVECs was significantly decreased in the presence of arresten and canstatin, while tube formation assays suggested that the total tube length and junction number of HUVECs were significantly inhibited by these two proteins; moreover, they could also reduce the expression of vascular endothelial growth factor (VEGF) and the phosphorylation levels of PI3K and Akt, which indicated that the activation of the 3-kinase/serine/threonine-kinase (PI3K/Akt) signaling pathway was inhibited. These findings may have important implications for the soluble recombinant expression of human arresten and canstatin, and for the related therapy of cancer.


Asunto(s)
Inhibidores de la Angiogénesis , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Inhibidores de la Angiogénesis/farmacología , Movimiento Celular , Proliferación Celular , Colágeno Tipo IV/farmacología , Escherichia coli/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética
4.
Autophagy ; 18(9): 2229-2245, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34964701

RESUMEN

Deficient bone regeneration causes bone defects or nonunion in a substantial proportion of trauma patients that urges for novel therapies. To develop a reliable therapy, we investigated the effect of negative pressure wound therapy (NPWT) on bone regeneration in vivo in a rat calvarial defect model. Negative pressure (NP) treatment in vitro was mimicked to test its effect on osteoblast differentiation in rat mesenchymal stem cells (MSCs) and MC3T3-E1 cells. Transcriptomic analyses, pharmaceutical interventions, and shRNA knockdowns were conducted to explore the underlying mechanism and their clinical relevance was investigated in samples from patients with nonunion. The potential application of a combined therapy of MSCs in hydrogels with negative pressure was tested in the rat critical-size calvarial defect model. We found that NPWT promoted bone regeneration in vivo and NP treatment induced osteoblast differentiation in vitro. NP induced osteogenesis via activating macroautophagy/autophagy by AMPK-ULK1 signaling that was impaired in clinical samples from patients with nonunion. More importantly, the combined therapy involving MSCs in hydrogels with negative pressure significantly improved bone regeneration in rat critical-size calvarial defect model. Thus, our study identifies a novel AMPK-ULK1-autophagy axis by which negative pressure promotes osteoblast differentiation of MSCs and bone regeneration. NPWT treatment can potentially be adopted for therapy of bone defects.Abbreviations: ADP, adenosine diphosphate; AICAR/Aic, acadesine; ALP, alkaline phosphatase; ALPL, alkaline phosphatase, biomineralization associated; AMP, adenosine monophosphate; AMPK, AMP-activated protein kinase; ARS, alizarin red S staining; ATG7, autophagy related 7; ATP, adenosine triphosphate; BA1, bafilomycin A1; BGLAP/OCN, bone gamma-carboxyglutamate protein; BL, BL-918; BS, bone surface; BS/TV, bone surface per tissue volume; BV/TV, bone volume per tissue volume; C.C, compound C; CCN1, cellular communication network factor 1; COL1A1, collagen type I alpha 1 chain; COL4A3, collagen type IV alpha 3 chain; COL4A4, collagen type IV alpha 4 chain; COL18A1, collagen type XVIII alpha 1 chain; CQ, chloroquine; GelMA, gelatin methacryloyl hydrogel; GO, Gene Ontology; GSEA, gene set enrichment analysis; HIF1A, hypoxia inducible factor 1 subunit alpha; HPLC, high-performance liquid chromatography; ITGAM/CD11B, integrin subunit alpha M; ITGAX/CD11C, integrin subunit alpha X; ITGB1/CdD9, integrin subunit beta 1; KEGG, Kyoto Encyclopedia of Genes and Genomes; MAP1LC3B/LC3B, microtubule associated protein 1 light chain 3 beta; micro-CT, microcomputed tomography; MSCs, mesenchymal stem cells; MTOR, mechanistic target of rapamycin kinase; NP, negative pressure; NPWT, negative pressure wound therapy; PRKAA1/AMPKα1, protein kinase AMP-activated catalytic subunit alpha 1; PRKAA2, protein kinase AMP-activated catalytic subunit alpha 2; PTPRC/CD45, protein tyrosine phosphatase receptor type C; ROS, reactive oxygen species; RUNX2, RUNX family transcription factor 2; SBI, SBI-0206965; SPP1/OPN, secreted phosphoprotein 1; THY1/CD90, Thy-1 cell surface antigen; SQSTM1, sequestosome 1; TGFB3, transforming growth factor beta 3; ULK1/Atg1, unc-51 like autophagy activating kinase 1.


Asunto(s)
Autofagia , Terapia de Presión Negativa para Heridas , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato , Fosfatasa Alcalina/farmacología , Animales , Autofagia/fisiología , Homólogo de la Proteína 1 Relacionada con la Autofagia , Regeneración Ósea , Colágeno Tipo IV/farmacología , Gelatina , Hidrogeles/farmacología , Integrinas , Metacrilatos , Osteogénesis , Ratas , Microtomografía por Rayos X
5.
Pathol Res Pract ; 223: 153463, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33971545

RESUMEN

The presence and clinical significance of IL-17 and IL-17-expressing cells have been studied for several cancers, although their correlation with tumor development remains controversial. Peripheral blood was collected from healthy donors and glioma patients to isolate peripheral blood mononuclear cells (PBMCs). The percentage of IL-17-expressing cells and the production of inflammatory cytokines in PBMCs and tissues were measured. Human IL­17 cDNA was then inserted into the pEGFP­N1 plasmid and transfected into the glioma U87MG cell line, and tumstatin was used to block the effect of the IL-17 overexpression. Stem cell transcription factors were evaluated in each group using qRT-PCR and western blotting, and proliferation and migration were detected using colony formation and wound-healing assays. The cells were then subcutaneously inoculated into nude mice to evaluate the growth of glioma. Compared with healthy donors, the PBMCs from glioma patients showed a significant accumulation of IL-17-expressing T cells. Th17 cell differentiation-related cytokines (IL-23, TGF-ß and IL-6) were increased in the tumor microenvironment. IL­17 transfection increased the mRNA and protein expression of stem cell transcription factors in U87MG cells in vitro. The proliferation and migration of U87MG cells were also increased. Moreover, the pEGFP­N1­IL­17­U87MG cells grew more rapidly than other cells. However, tumstatin-treated U87MG cells showed significantly inhibited the effects of IL-17 overexpression. Tumstatin effectively suppressed IL-17-derived U87MG cell growth by downregulating stem cell maintenance factors and inducing proliferation and migration. These findings indicated that IL-17 represents a potential prognostic marker for glioma, while tumstatin has potential in the treatment for glioma.


Asunto(s)
Antineoplásicos/farmacología , Autoantígenos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Colágeno Tipo IV/farmacología , Glioma/tratamiento farmacológico , Interleucina-17/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Th17/metabolismo , Adulto , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Glioma/inmunología , Glioma/metabolismo , Glioma/patología , Humanos , Interleucina-17/genética , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Th17/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Drug Deliv ; 28(1): 844-855, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33928829

RESUMEN

T7 peptide is considered as an antiangiogenic polypeptide. The presents study aimed to further detect the antiangiogenic mechanisms of T7 peptide and determine whether combining T7 peptide and meloxicam (COX-2/PGE2 specific inhibitor) could offer a better therapy to combat hepatocellular carcinoma (HCC). T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3ß1 and αvß3 pathways. Cell proliferation, migration, apoptosis, or tube formation ability were detected, and the expression of integrin-associated regulatory proteins was detected. The anti-tumor activity of T7 peptide, meloxicam, and their combination were evaluated in HCC tumor models established in mice. T7 peptide suppressed the proliferation, migration, tube formation, and promoted the apoptosis of endothelial cells under both normoxic and hypoxic conditions via integrin α3ß1 and αvß3 pathways. Meloxicam enhanced the activity of T7 peptide under hypoxic condition. T7 peptide partly inhibited COX-2 expression via integrin α3ß1 not αvß3-dependent pathways under hypoxic condition. T7 peptide regulated apoptosis associated protein through MAPK-dependent and -independent pathways under hypoxic condition. The MAPK pathway was activated by the COX-2/PGE2 axis under hypoxic condition. The combination of T7 and meloxicam showed a stronger anti-tumor effect against HCC tumors in mice. The data highlight that meloxicam enhanced the antiangiogenic activity of T7 peptide in vitro and in vivo.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Colágeno Tipo IV/farmacología , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/antagonistas & inhibidores , Neoplasias Hepáticas/tratamiento farmacológico , Meloxicam/farmacología , Fragmentos de Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Humanos , Hipoxia/patología , Integrinas/efectos de los fármacos , Ratones , Neovascularización Patológica/tratamiento farmacológico , ARN Interferente Pequeño/metabolismo , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Mol Sci ; 22(3)2021 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-33498253

RESUMEN

Ventricular arrhythmia induced by ischemia/reperfusion (I/R) injury is a clinical problem in reperfusion therapies for acute myocardial infarction. Ca2+ overload through reactive oxygen species (ROS) production is a major cause for I/R-induced arrhythmia. We previously demonstrated that canstatin, a C-terminal fragment of type IV collagen α2 chain, regulated Ca2+ handling in rat heart. In this study, we aimed to clarify the effects of canstatin on I/R-induced ventricular arrhythmia in rats. Male Wistar rats were subjected to I/R injury by ligating the left anterior descending artery followed by reperfusion. Ventricular arrhythmia (ventricular tachycardia and ventricular fibrillation) was recorded by electrocardiogram. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) activity and ROS production in neonatal rat cardiomyocytes (NRCMs) stimulated with oxygen glucose deprivation/reperfusion (OGD/R) were measured by lucigenin assay and 2',7'-dichlorodihydrofluorescein diacetate staining, respectively. The H2O2-induced intracellular Ca2+ ([Ca2+]i) rise in NRCMs was measured by a fluorescent Ca2+ indicator. Canstatin (20 µg/kg) inhibited I/R-induced ventricular arrhythmia in rats. Canstatin (250 ng/mL) inhibited OGD/R-induced NOX activation and ROS production and suppressed the H2O2-induced [Ca2+]i rise in NRCMs. We for the first time demonstrated that canstatin exerts a preventive effect against I/R-induced ventricular arrhythmia, perhaps in part through the suppression of ROS production and the subsequent [Ca2+]i rise.


Asunto(s)
Antiarrítmicos/uso terapéutico , Colágeno Tipo IV/uso terapéutico , Daño por Reperfusión Miocárdica/complicaciones , Fragmentos de Péptidos/uso terapéutico , Taquicardia/prevención & control , Fibrilación Ventricular/prevención & control , Animales , Antiarrítmicos/farmacología , Calcio/metabolismo , Células Cultivadas , Colágeno Tipo IV/farmacología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Taquicardia/tratamiento farmacológico , Taquicardia/etiología , Fibrilación Ventricular/tratamiento farmacológico , Fibrilación Ventricular/etiología
8.
Int J Biol Macromol ; 173: 541-553, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33493562

RESUMEN

Tissue regeneration and wound healing are still serious clinical complications globally and lack satisfactory cures. Inspired by the impressive regeneration ability of the post-injury earthworms and their widely accepted medicinal properties, we screened and identified a novel collagen-like peptide from the amputated earthworms using high-throughput techniques, including transcriptomics, proteomics, and mass spectrum. The identified collagen-like peptide col4a1 was cloned and expressed to comprehensively investigate the wound healing effect and underlying mechanism. It exerted significant effects on wound healing both in vitro and in vivo, including enhanced viability, proliferation, migration of fibroblasts, granulation, and collagen deposition. Moreover, the col4a1 functioned via binding with integrin α2ß1 and upregulating the RAS/MAPK signaling pathway. This work demonstrates that the novel collagen-like peptide col4a1 obtained from the amputated earthworms enables enhanced wound healing and provides new opportunities for wound care.


Asunto(s)
Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Perfilación de la Expresión Génica/métodos , Integrina alfa1beta1/metabolismo , Oligoquetos/fisiología , Proteómica/métodos , Cicatrización de Heridas/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Clonación Molecular , Colágeno Tipo IV/farmacología , Modelos Animales de Enfermedad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Espectrometría de Masas , Ratones , Células 3T3 NIH , Oligoquetos/genética , Oligoquetos/metabolismo , Análisis de Secuencia de ARN
9.
Drug Dev Res ; 82(3): 430-439, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33244794

RESUMEN

Complication of arsenic trioxide (ATO) and other drugs in cancer treatment has attracted much focus, but is limitedly investigated in hepatocellular carcinoma (HCC). This study aimed to explore the role of ATO combined with canstatin in HCC. HepG2 cells were treated with different concentrations of ATO with or without canstatin, CCK-8, flow cytometry, Transwell assays were conducted to determine cell proliferation, apoptosis, adhesion, migration, and invasion abilities. Besides, the protein expression or mRNA level of caspase-3, PCNA, and MMP-2 was measured using western blotting or qRT-PCR. BALB/c-nu/nu mice were used to establish nude mouse transplantation tumor model, and received ATO or canstatin treatment for 3 weeks. The results showed that ATO inhibited cell proliferation, adhesion, migration and invasion, and promoted cell apoptosis with a concentration-dependent way. Canstatin had a significantly inhibitory effect on cell proliferation, but had limited effects on the other cellular behaviors. Besides, combination with ATO and canstatin strengthened the effects of ATO alone on cell proliferation inhibition and cell apoptosis promotion. Moreover, both of ATO and canstatin increased the protein expression of caspase-3, while decreased PCNA and MMP-2, which was further strengthened upon their combination. Furthermore, both of ATO and canstatin inhibited tumor growth in vivo, which was also strengthened upon their combination. Collectively, we found that combined canstatin and ATO significantly inhibited cell proliferation, migration and adhesion abilities, and promoted cell apoptosis, and inhibited tumor growth, thus suppressed the progression of HCC.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Colágeno Tipo IV/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Vis Exp ; (165)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33283783

RESUMEN

Brain microvascular endothelial cells (BMECs) can be differentiated from human induced pluripotent stem cells (iPSCs) to develop ex vivo cellular models for studying blood-brain barrier (BBB) function. This modified protocol provides detailed steps to derive, expand, and cryopreserve BMECs from human iPSCs using a different donor and reagents than those reported in previous protocols. iPSCs are treated with essential 6 medium for 4 days, followed by 2 days of human endothelial serum-free culture medium supplemented with basic fibroblast growth factor, retinoic acid, and B27 supplement. At day 6, cells are sub-cultured onto a collagen/fibronectin matrix for 2 days. Immunocytochemistry is performed at day 8 for BMEC marker analysis using CLDN5, OCLN, TJP1, PECAM1, and SLC2A1. Western blotting is performed to confirm BMEC marker expression, and absence of SOX17, an endodermal marker. Angiogenic potential is demonstrated with a sprouting assay. Trans-endothelial electrical resistance (TEER) is measured using chopstick electrodes and voltohmmeter starting at day 7. Efflux transporter activity for ATP binding cassette subfamily B member 1 and ATP binding cassette subfamily C member 1 is measured using a multi-plate reader at day 8. Successful derivation of BMECs is confirmed by the presence of relevant cell markers, low levels of SOX17, angiogenic potential, transporter activity, and TEER values ~2000 Ω x cm2. BMECs are expanded until day 10 before passaging onto freshly coated collagen/fibronectin plates or cryopreserved. This protocol demonstrates that iPSC-derived BMECs can be expanded and passaged at least once. However, lower TEER values and poorer localization of BMEC markers was observed after cryopreservation. BMECs can be utilized in co-culture experiments with other cell types (neurons, glia, pericytes), in three-dimensional brain models (organ-chip and hydrogel), for vascularization of brain organoids, and for studying BBB dysfunction in neuropsychiatric disorders.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/citología , Criopreservación , Células Endoteliales/citología , Células Madre Pluripotentes Inducidas/citología , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Biomarcadores/metabolismo , Barrera Hematoencefálica/citología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Colágeno Tipo IV/farmacología , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibronectinas/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos
11.
Sci Rep ; 10(1): 12881, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32732948

RESUMEN

Myocardial infarction (MI) still remains a leading cause of mortality throughout the world. An adverse cardiac remodeling, such as hypertrophy and fibrosis, in non-infarcted area leads to uncompensated heart failure with cardiac dysfunction. We previously demonstrated that canstatin, a C-terminus fragment of type IV collagen α2 chain, exerted anti-remodeling effect against isoproterenol-induced cardiac hypertrophy model rats. In the present study, we examined whether a long-term administration of recombinant canstatin exhibits a cardioprotective effect against the adverse cardiac remodeling in MI model rats. Left anterior descending artery of male Wistar rats was ligated and recombinant mouse canstatin (20 µg/kg/day) was intraperitoneally injected for 28 days. Long-term administration of canstatin improved survival rate and significantly inhibited left ventricular dilatation and dysfunction after MI. Canstatin significantly inhibited scar thinning in the infarcted area and significantly suppressed cardiac hypertrophy, nuclear translocation of nuclear factor of activated T-cells, interstitial fibrosis and increase of myofibroblasts in the non-infarcted area. Canstatin significantly inhibited transforming growth factor-ß1-induced differentiation of rat cardiac fibroblasts into myofibroblasts. The present study for the first time demonstrated that long-term administration of recombinant canstatin exerts cardioprotective effects against adverse cardiac remodeling in MI model rats.


Asunto(s)
Cardiomegalia , Cardiotónicos/farmacología , Colágeno Tipo IV/farmacología , Infarto del Miocardio , Miocardio , Fragmentos de Péptidos/farmacología , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/prevención & control , Masculino , Ratones , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología
12.
Int J Mol Sci ; 21(14)2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32708100

RESUMEN

Persistent inflammation is a complication associated with many ocular diseases. Changes in ocular vessels can amplify disease responses and contribute to vision loss by influencing the delivery of leukocytes to the eye, vascular leakage, and perfusion. Here, we report the anti-inflammatory activity for AXT107, a non-RGD, 20-mer αvß3 and α5ß1 integrin-binding peptide that blocks vascular endothelial growth factor (VEGF)-signaling and activates tyrosine kinase with immunoglobulin and EGF-like domains 2 (Tie2) using the normally inhibitory ligand angiopoietin 2 (Ang2). Tumor necrosis factor α (TNFα), a central inflammation mediator, induces Ang2 release from endothelial cells to enhance its stimulation of inflammation and vascular leakage. AXT107 resolves TNFα-induced vascular inflammation in endothelial cells by converting the endogenously released Ang2 into an agonist of Tie2 signaling, thereby disrupting both the synergism between TNFα and Ang2 while also preventing inhibitor of nuclear factor-κB α (IκBα) degradation directly through Tie2 signaling. This recovery of IκBα prevents nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) nuclear localization, thereby blocking NF-κB-induced inflammatory responses, including the production of VCAM-1 and ICAM-1, leukostasis, and vascular leakage in cell and mouse models. AXT107 also decreased the levels of pro-inflammatory TNF receptor 1 (TNFR1) without affecting levels of the more protective TNFR2. These data suggest that AXT107 may provide multiple benefits in the treatment of retinal/choroidal and other vascular diseases by suppressing inflammation and promoting vascular stabilization.


Asunto(s)
Angiopoyetina 2/metabolismo , Colágeno Tipo IV/farmacología , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Inflamación/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Receptor TIE-2/metabolismo , Angiopoyetina 1/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Enfermedades de la Coroides/tratamiento farmacológico , Colágeno Tipo IV/uso terapéutico , Endotelio Vascular/citología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucostasis/tratamiento farmacológico , Leucostasis/metabolismo , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/uso terapéutico , Receptor TIE-2/agonistas , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Enfermedades de la Retina/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Eur J Pharmacol ; 871: 172849, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-31843516

RESUMEN

Pathological cardiac hypertrophy associated with cardiac dysfunction is an independent risk factor for arrhythmia, myocardial infarction and sudden death. Canstatin, a C-terminal fragment of type IV collagen α2 chain, is abundantly expressed in normal heart tissue. We previously demonstrated that canstatin inhibits isoproterenol (ISO)-induced dephosphorylation of nuclear factor of activated T-cells (NFAT)c4, which plays an important role in cardiac hypertrophy, in differentiated H9c2 cardiomyoblasts. Thus, we investigated whether in vivo canstatin administration prevents ISO-induced cardiac hypertrophy through the inhibition of NFATc4 pathway. Rats were subcutaneously injected with ISO (5 mg/kg) or saline (Cont) for 7 days. Simultaneously, recombinant mouse canstatin (20 µg/kg) or vehicle was intraperitoneally administered. After left ventricular wall thickness and cardiac function were measured by echocardiography, the hearts were isolated and left ventricular weight (LVW) was weighed. Azan staining was performed to measure cross-sectional diameter of cardiomyocytes. Activity of calcineurin, which dephosphorylates NFATc4, was measured by calcineurin phosphatase activity assay. Immunohistochemical staining was performed to evaluate nuclear translocation of NFATc4. Intracellular Ca2+ concentration in neonatal rat cardiomyocytes (NRCMs) was measured by using a calcium indicator. Canstatin significantly inhibited ISO-induced increase of LVW, left ventricular posterior wall thickness at end-diastole and diameter of cardiomyocytes. Canstatin significantly inhibited ISO-induced activation of calcineurin, nuclear translocation of NFATc4, increased mRNA expression of ß-myosin heavy chain and α-skeletal actin, and intracellular Ca2+ rise in NRCMs. In summary, we for the first time demonstrated that canstatin administration suppresses ISO-induced cardiac hypertrophy possibly through the blockade of calcineurin/NFATc4 pathway in rats.


Asunto(s)
Calcineurina/metabolismo , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Colágeno Tipo IV/farmacología , Isoproterenol/efectos adversos , Factores de Transcripción NFATC/metabolismo , Fragmentos de Péptidos/farmacología , Animales , Calcio/metabolismo , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Relación Dosis-Respuesta a Droga , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Masculino , Ratones , Monoéster Fosfórico Hidrolasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar
14.
Int J Mol Med ; 44(2): 523-534, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31173192

RESUMEN

The T7 peptide, an active fragment of full­length tumstatin [the non­collagenous 1 domain of the type IV collagen α3 chain, α3 (IV) NC1], has exhibited potential antitumor effects in several types of cancer cells. However, the mechanism underlying its action against human hepatocellular carcinoma (HCC) remains unclear. The present study aimed to investigate the role of autophagy in T7 peptide­induced cytotoxicity in HCC cells in vitro and in vivo. The results revealed that the T7 peptide significantly reduced cell viability and induced cell cycle arrest in HCC cells. The T7 peptide induced apoptosis in HCC cells through upregulation of Bax, Fas, and Fas ligand, and through upregulation of the anti­apoptotic protein Bcl­2. In addition, treatment with the T7 peptide induced protective autophagy in HCC cells. Blocking autophagy by 3­methyladenineor bafilomycin A1 enhanced T7 peptide­induced apoptosis. Furthermore, co­treatment with MK­2206 (an Akt specific inhibitor) or rapamycin (an inhibitor of mTOR) enhanced T7 peptide­induced autophagy, whereas co­treatment with insulin (an activator of the Akt/mTOR signaling pathway) alleviated T7 peptide­induced autophagy, which suggested that the T7 peptide may induce autophagy activation via inhibition of the Akt/mTOR signaling pathway. Taken together, the present results demonstrated that suppression of autophagy potentiated the cytotoxic effects of the T7 peptide, and suggested that the T7 peptide may serve as a potential alternative compound for HCC therapy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Colágeno Tipo IV/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Animales , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Colágeno Tipo IV/uso terapéutico , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Fragmentos de Péptidos/uso terapéutico
15.
J Pharmacol Sci ; 139(3): 193-200, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30827890

RESUMEN

Ischemia/reperfusion (I/R)-induced oxidative stress is a serious clinical problem in the reperfusion therapy for ischemic diseases. Tumstatin is an endogenous bioactive peptide cleaved from type IV collagen α3 chain. We previously reported that T3 peptide, an active subfragment of tumstatin, exerts cytoprotective effects on H2O2-induced apoptosis through the inhibition of intracellular reactive oxygen species (ROS) production in H9c2 cardiomyoblasts. In this study, we investigated whether T3 peptide has cardioprotective effects against I/R injury by using in vitro and ex vivo experimental models. H9c2 cardiomyoblasts were stimulated with oxygen and glucose deprivation (OGD) for 12 h followed by reoxygenation for 1-8 h (OGD/R; in vitro model). The cells were treated with T3 peptide (30-1000 ng/ml) during OGD. Ten minutes after the pre-perfusion of T3 peptide (300 ng/ml), Langendorff perfused rat hearts were exposed to ischemia for 30 min followed by reperfusion for 1 h (ex vivo model). T3 peptide inhibited OGD/R-induced apoptosis through the inhibition of mitochondrial ROS production and dysfunction in H9c2 cardiomyoblasts. T3 peptide also prevented I/R-induced cardiac dysfunction, arrhythmia and myocardial infarction in the perfused rat heart. In conclusion, we for the first time demonstrated that T3 peptide exerts cardioprotective effects against I/R injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Autoantígenos/administración & dosificación , Cardiotónicos/administración & dosificación , Colágeno Tipo IV/administración & dosificación , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/prevención & control , Autoantígenos/química , Autoantígenos/farmacología , Cardiotónicos/farmacología , Línea Celular , Colágeno Tipo IV/química , Colágeno Tipo IV/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Infarto del Miocardio/etiología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/complicaciones , Péptidos/administración & dosificación , Péptidos/farmacología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
16.
JCI Insight ; 4(4)2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30668550

RESUMEN

The angiopoietin (Ang)/Tie2 signaling pathway is essential for maintaining vascular homeostasis, and its dysregulation is associated with several diseases. Interactions between Tie2 and α5ß1 integrin have emerged as part of this control; however, the mechanism is incompletely understood. AXT107, a collagen IV-derived peptide, has strong antipermeability activity and has enabled the elucidation of this previously undetermined mechanism. Previously, AXT107 was shown to inhibit VEGFR2 and other growth factor signaling via receptor tyrosine kinase association with specific integrins. AXT107 disrupts α5ß1 and stimulates the relocation of Tie2 and α5 to cell junctions. In the presence of Ang2 and AXT107, junctional Tie2 is activated, downstream survival signals are upregulated, F-actin is rearranged to strengthen junctions, and, as a result, endothelial junctional permeability is reduced. These data suggest that α5ß1 sequesters Tie2 in nonjunctional locations in endothelial cell membranes and that AXT107-induced disruption of α5ß1 promotes clustering of Tie2 at junctions and converts Ang2 into a strong agonist, similar to responses observed when Ang1 levels greatly exceed those of Ang2. The potentiation of Tie2 activation by Ang2 even extended to mouse models in which AXT107 induced Tie2 phosphorylation in a model of hypoxia and inhibited vascular leakage in an Ang2-overexpression transgenic model and an LPS-induced inflammation model. Because Ang2 levels are very high in ischemic diseases, such as diabetic macular edema, neovascular age-related macular degeneration, uveitis, and cancer, targeting α5ß1 with AXT107 provides a potentially more effective approach to treat these diseases.


Asunto(s)
Angiopoyetina 2/metabolismo , Colágeno Tipo IV/farmacología , Inflamación/tratamiento farmacológico , Integrina alfa5beta1/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Receptor TIE-2/metabolismo , Angiopoyetina 2/genética , Animales , Permeabilidad Capilar/efectos de los fármacos , Línea Celular , Colágeno Tipo IV/uso terapéutico , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inmunología , Inflamación/patología , Integrina alfa5beta1/metabolismo , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/uso terapéutico , Péptidos/farmacología , Péptidos/uso terapéutico , Receptor TIE-2/genética , Transducción de Señal/efectos de los fármacos
17.
ACS Appl Mater Interfaces ; 11(1): 176-186, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30525386

RESUMEN

The blood-brain tumor barrier (BTB) and blood-brain barrier (BBB) have always been the major barriers in glioma therapy. In this report, we proposed D-T7 peptide-modified nanoparticles actively targeted glioma by overcoming the BBB and BTB to improve the antiglioma efficacy. Glioma-targeting experiments showed that the penetration effect of the D-T7 peptide-modified nanoparticles was 7.89-fold higher than that of unmodified nanoparticles. Furthermore, cediranib (CD) and paclitaxel (PTX) were used for the combination of the antiangiogenesis and chemotherapy for glioma. PEGylated bilirubin nanoparticles (BRNPs) were selected as a suitable drug delivery system (CD&PTX@TBRBPs) owing to the antioxidant, anti-inflammatory, and reactive oxygen species-responsive ability. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and apoptosis assays showed that CD&PTX@TBRBPs had the highest cytotoxicity and the median survival time of the CD&PTX@TBRNP group was 3.31-fold and 1.23-fold longer than that of the saline and CD&PTX@BRNP groups, respectively. All the results showed that we constructed a novel and accessible peptide-modified dual drug carrier with an enhanced antiglioma effect.


Asunto(s)
Bilirrubina , Neoplasias Encefálicas , Colágeno Tipo IV , Portadores de Fármacos , Glioma , Nanopartículas , Paclitaxel , Fragmentos de Péptidos , Quinazolinas , Animales , Bilirrubina/química , Bilirrubina/farmacología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Colágeno Tipo IV/química , Colágeno Tipo IV/farmacocinética , Colágeno Tipo IV/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/farmacología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacocinética , Fragmentos de Péptidos/farmacología , Quinazolinas/química , Quinazolinas/farmacocinética , Quinazolinas/farmacología
18.
J Pharmacol Sci ; 139(2): 59-64, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30580971

RESUMEN

Type IV collagen is a main component of basement membrane extracellular matrix. Canstatin, a non-collagenous C-terminal fragment of type IV collagen α2 chain, was firstly identified as an endogenous anti-angiogenic and anti-tumor factor, which also has an anti-lymphangiogenic effect. Then, canstatin has been widely investigated as a novel target molecule for cancer therapy. The anti-angiogenic effect of canstatin may be also useful for the treatment of ocular neovascularization. Recently, we have demonstrated that canstatin, which is abundantly expressed in the heart tissue, exerts various biological activities in cardiac cells. In rat H9c2 cardiomyoblasts, canstatin inhibits isoproterenol- or hypoxia-induced apoptosis. Canstatin plays an important role in modulating voltage-dependent calcium channel activity in rat cardiomyocytes. Canstatin also regulates various biological functions in rat cardiac fibroblasts and myofibroblasts. The expression of canstatin decreases in the infarcted area after myocardial infarction. This review focuses on a current perspective for the roles of canstatin in tumorigenesis, ocular neovascularization and cardiac pathology.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Colágeno Tipo IV/farmacología , Animales , Colágeno Tipo IV/metabolismo , Oftalmopatías/tratamiento farmacológico , Corazón/efectos de los fármacos , Humanos , Neovascularización Patológica/tratamiento farmacológico
19.
Pharmazie ; 73(12): 715-720, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30522555

RESUMEN

Tumstatin7 (CNYYSNS) is an antitumor peptide derived from the NC1 domain of Type IV collagen that has been associated with tumor angiogenesis. In this work, we generated a peptide composed of tumstatin7 fused to TAT, a cell-internalizing peptide consisting of 11 amino acids. Tumstatin7-TAT was internalized by cells and triggered cell death. The new peptide was more potent in inducing B16F10 melanoma cell apoptosis in vitro than the shorter tumstatin7. Whereas tumstatin7-TAT significantly reduced tumor cell viability, tumstatin7 showed only weak effects even at the highest treatment concentration applied. Both tumstatin7-TAT and tumstatin7 inhibited cell migration in an in vitro wound healing model, and the former was more effective than the latter in inhibiting tumor growth in vivo. Combining the cell-internalizing property of TAT with the tumor-specific property of tumstatin7 may provide a useful adjunct to tumor therapy.


Asunto(s)
Autoantígenos/farmacología , Colágeno Tipo IV/farmacología , Productos del Gen tat/metabolismo , Melanoma Experimental/tratamiento farmacológico , Péptidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Autoantígenos/administración & dosificación , Autoantígenos/química , Movimiento Celular/efectos de los fármacos , Colágeno Tipo IV/administración & dosificación , Colágeno Tipo IV/química , Femenino , Humanos , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Péptidos/administración & dosificación , Péptidos/química , Cicatrización de Heridas/efectos de los fármacos
20.
J Theor Biol ; 455: 212-221, 2018 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-30036530

RESUMEN

Cooperation between VEGFR2 and integrin αVß3 is critical for neovascularization in wound healing, cardiovascular ischemic diseases, ocular diseases, and tumor angiogenesis. In the present study, we developed a rule-based computational model to investigate the potential mechanism by which the Src-induced integrin association with VEGFR2 enhances VEGFR2 activation. Simulations demonstrated that the main function of integrin is to reduce the degradation of VEGFR2 and hence stabilize the activation signal. In addition, receptor synthesis rate and recruitment from internal compartment were found to be sensitive determinants of the activation state of VEGFR2. The model was then applied to simulate the effect of integrin-binding peptides such as tumstatin and cilengitide on VEGFR2 signaling. Further, computational modeling proposed potential molecular mechanisms for the angiogenesis-modulating activity of other integrin-binding peptides. The model highlights the complexity of the crosstalk between αVß3 integrin and VEGFR2 and the necessity of utilizing models to elucidate potential mechanisms in angiogenesis-modulating peptide therapy.


Asunto(s)
Autoantígenos , Colágeno Tipo IV , Células Endoteliales/metabolismo , Integrina alfaVbeta3/metabolismo , Modelos Biológicos , Neovascularización Patológica , Transducción de Señal/efectos de los fármacos , Venenos de Serpiente , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Autoantígenos/farmacología , Colágeno Tipo IV/farmacocinética , Colágeno Tipo IV/farmacología , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Venenos de Serpiente/farmacocinética , Venenos de Serpiente/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA