Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Phytomedicine ; 118: 154966, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37487254

RESUMEN

BACKGROUND: A proprietary Chinese herbal product called Dan-Deng-Tong-Nao softgel capsule (DDTNC) is used to treat ischemic stroke. However, the preventive mechanisms of DDTNC against cerebral ischemia reperfusion injury (CIRI) haven not been characterized. OBJECTIVE: To explore the mechanisms of protective effects of DDTNC against CIRI from both internal and external levels. METHODS: Chemical characterization was performed using UPLC. The potential protective mechanisms of DDTNC against CIRI were predicted using network pharmacology. Model of middle cerebral artery occlusion/reperfusion (MCAO/R) was established in rats. An model of brain microvascular endothelial cells (BMECs) induced by oxygen-glucose deprivation/reoxygenation (OGD/R) was also established. We evaluated neurological deficits, cerebral infarct volume, cortical neuron damage, and mitochondrial swelling in vivo. We evaluated the expression of VEGFR2, VEGFA, HIF-1α, CD31, and CD34 in ischemic cortex, and VEGF, bFGF, BDNF, angiostatin, and endostatin in serum of rats and in BMEC supernatants. We also evaluated cell viability, cytotoxicity, intracellular ROS, apoptosis, and migration ability in vitro. RESULTS: Seven components were detected in DDTNC. KEGG enrichment analysis showed that DDTNC may modulate angiogenesis via the HIF-1 signaling pathway. DDTNC treatment reduced neurological score and infarct volume, and improved cell morphology of damaged neurons. Transmission electron microscopy showed that DDTNC reduced mitochondria swelling in cortical neurons. Furthermore, DDTNC reduced intracellular ROS and inhibited apoptosis. DDTNC boosted the expression of CD31, CD34, VEGFR2, VEGFA and HIF-1α, highlighting its involvement in angiogenesis, according to immunofluorescence studies. Furthermore, DDTNC enhanced tube formation and migration of BMECs in vitro. ELISA and western blotting indicated that DDTNCCSF induced the expression of VEGF, BDNF and bFGF, reduced the level of angiostatin and endostatin, increased the protein expression of VEGFA, Notch1 and HIF-1α in vitro and in vivo. CONCLUSIONS: DDTNC promoted angiogenesis to protect brain tissue against MCAO/R, and exerted protective effects against OGD/R in BMECs via activating HIF-1α-VEGFA-NOTCH1 signal transduction pathway.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Ratas , Animales , Células Endoteliales , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiostatinas/metabolismo , Angiostatinas/farmacología , Angiostatinas/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Endostatinas/metabolismo , Endostatinas/farmacología , Endostatinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Microvasos/metabolismo , Receptor Notch1/metabolismo
2.
Metab Brain Dis ; 36(8): 2263-2271, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34003412

RESUMEN

Vascular endothelial growth factor (VEGF) regulates angio/neurogenesis and also tightly links to the pathogenesis of Alzheimer's disease (AD). Although exercise has a beneficial effect on neurovascular function and cognitive function, the direct effect of exercise on VEGF-related signaling and cognitive deficit in AD is incompletely understood. Therefore, the purpose of this study was to investigate the protective effect of exercise on angiostatin/VEGF cascade and cognitive function in AD model rats. Wistar male rats were randomly divided into five groups: control (CON), injection of DMSO (Sham-CON), CON-exercise (sham-EX), intrahippocampal injection of Aß (Aß), and Aß-exercise (Aß-EX). Rats in EX groups underwent treadmill exercise for 4 weeks, then the cognitive function was measured by the Morris Water Maze (MWM) test. mRNA levels of hypoxia-induced factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 2 (VEGFR2), and angiostatin were determined in hippocampus by RT-PCR. We found that spatial learning and memory were impaired in Aß-injected rats, but exercise training improved it. Moreover, exercise training increased the reduced mRNA expression level of VEGF signaling, including HIF1α, VEGF, and VEGFR2 in the hippocampus from Aß-injected rats. Also, the mRNA expression level of angiostatin was elevated in the hippocampus from Aß-injected rats, and exercise training abrogated its expression. Our findings suggest that exercise training improves cognitive function in Aß-injected rats, possibly through enhancing VEGF signaling and reducing angiostatin.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/farmacología , Angiostatinas/metabolismo , Angiostatinas/farmacología , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/terapia , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
J Pharmacol Sci ; 129(3): 200-4, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26603529

RESUMEN

This study aimed to elucidate the protective potential of angiostatin in inflamed endothelial cells in culture. We assessed the effect of angiostatin on the expression of ICAM-1 and eNOS. Angiostatin prevented IL-1ß-induced down-regulation of eNOS expression, but produced no significant changes on IL-1ß-induced up-regulation of ICAM-1. We then explored the effect of angiostatin on IL-1ß-mediated inflammatory signaling and found that angiostatin inhibited IL-1ß-mediated nuclear translocation of NF-κB. Thus, our results suggest that angiostatin prevents IL-1ß-induced down-regulation of eNOS expression via inhibition of the NF-κB cascade; this may be the anti-inflammatory mechanism of angiostatin.


Asunto(s)
Angiostatinas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Interleucina-1beta/efectos adversos , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Células Cultivadas , Depresión Química , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Transducción de Señal
4.
Am J Physiol Lung Cell Mol Physiol ; 306(1): L58-68, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24213918

RESUMEN

Acute lung injury is marked by profound influx of activated neutrophils, which have delayed apoptosis, along with fluid accumulation that impairs lung function and causes high mortality. Inflammatory and antimicrobial molecules, such as reactive oxygen species from activated neutrophils with prolonged lifespan, cause tissue damage and contribute to lung dysfunction. Angiostatin, an endogenous antiangiogenic molecule, is expressed in the lavage fluid of patients with acute respiratory distress syndrome and modifies neutrophil infiltration in a mouse model of peritonitis. Our aim was to investigate the therapeutic role of angiostatin in acute lung injury. We analyzed bronchoalveolar lavage and lung tissues from C57BL/6 mouse model of Escherichia coli LPS-induced acute lung injury to assess the effects of angiostatin treatment. Subcutaneous angiostatin administered at 5 h after LPS treatment reduces histological signs of inflammation, protein accumulation, lung Gr1+ neutrophils, myeloperoxidase activity, and expression of phosphorylated p38 MAPK in lung tissues and peripheral blood neutrophils, while increasing the number of apoptotic cells in the lungs without affecting the levels of macrophage inflammatory protein-1 α, IL-1ß, keratinocyte chemoattractant, and monocyte chemoattractant protein-1 in lavage and lung homogenates at 9 and 24 h after LPS treatment. In contrast, angiostatin administered intravenously 5 h after LPS treatment did not reduce histological sign of inflammation, BAL cell recruitment, and protein concentration at 9 h of LPS treatment. We conclude that angiostatin administered subcutaneously after LPS challenge inhibits acute lung inflammation up to 24 h after LPS treatment.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Angiostatinas/administración & dosificación , Antiinflamatorios/administración & dosificación , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Angiostatinas/farmacología , Animales , Antiinflamatorios/farmacología , Apoptosis , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Inyecciones Subcutáneas , Lipopolisacáridos/farmacología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/inmunología , Peroxidasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Cell Tissue Res ; 355(2): 375-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24297047

RESUMEN

There is a critical need to identify molecules that modulate the biology of neutrophils because activated neutrophils, though necessary for host defense, cause exuberant tissue damage through production of reactive oxygen species and increased lifespan. Angiostatin, an endogenous anti-angiogenic cleavage product of plasminogen, binds to integrin αvß3, ATP synthase and angiomotin and its expression is increased in inflammatory conditions. We test the hypothesis that angiostatin inhibits neutrophil activation, induces apoptosis and blocks recruitment in vivo and in vitro. The data show immuno-reactivity for plasminogen/angiostatin in resting neutrophils. Angiostatin conjugated to FITC revealed that angiostatin was endocytozed by activated mouse and human neutrophils in a lipid raft-dependent fashion. Co-immunoprecipitation of human neutrophil lysates, confocal microscopy of isolated mouse and human neutrophils and functional blocking experiments showed that angiostatin complexes with flotillin-1 along with integrin αvß3 and ATP synthase. Angiostatin inhibited fMLP-induced neutrophil polarization, as well as caused inhibition of hsp-27 phosphorylation and stabilization of microtubules. Angiostatin treatment, before or after LPS-induced neutrophil activation, inhibited phosphorylation of p38 and p44/42 MAPKs, abolished reactive oxygen species production and released the neutrophils from suppressed apoptosis, as indicated by expression of activated caspase-3 and morphological evidence of apoptosis. Finally, intravital microscopy and myeloperoxidase assay showed inhibition of neutrophil recruitment in post-capillary venules of TNFα-treated cremaster muscle in mouse. These in vitro and in vivo data demonstrate angiostatin as a broad deactivator and silencer of neutrophils and an inhibitor of their migration. These data potentially open new avenues for the development of anti-inflammatory drugs.


Asunto(s)
Angiostatinas/farmacología , Movimiento Celular/efectos de los fármacos , Activación Neutrófila/efectos de los fármacos , Neutrófilos/citología , Animales , Apoptosis/efectos de los fármacos , Antígenos CD18/metabolismo , Polaridad Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Humanos , Inflamación/patología , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Músculos/efectos de los fármacos , Músculos/metabolismo , Músculos/patología , N-Formilmetionina Leucil-Fenilalanina/farmacología , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Oxidación-Reducción/efectos de los fármacos , Subunidades de Proteína/metabolismo , ATPasas de Translocación de Protón/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
6.
Neoplasia ; 15(6): 591-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23730207

RESUMEN

Oncolytic herpes simplex virus (oHSV) can potentially spread throughout the tumor, reach isolated infiltrating cells, kill them, and deliver anticancer agents. However, the host responds to oHSV by inducing intratumoral infiltration of macrophages that can engulf the virus, limiting the potential of this therapeutic strategy. Hypervascularity is a pathognomonic feature of glioblastoma (GBM) and is a promising therapeutic target. Antiangiogenic treatments have multiple benefits, including the capacity to increase oHSV efficacy by suppressing macrophage extravasation and infiltration into the tumor. Angiostatin is an antiangiogenic polypeptide, and interleukin-12 (IL-12) is an immunostimulatory cytokine with strong antiangiogenic effects. Clinical use of each has been limited by delivery issues and systemic toxicity. We tested a combination treatment strategy using oHSVs expressing angiostatin (G47Δ-mAngio) and IL-12 (G47Δ-mIL12) in two orthotopic human GBM models. Intratumoral injection of G47Δ-mAngio and G47Δ-mIL12 in mice bearing intracranial U87 or tumors derived from glioblastoma stem cells significantly prolonged survival compared to each armed oHSV alone. This was associated with increased antiangiogenesis and virus spread and decreased macrophages. These data support the paradigm of using oHSV expressing different antiangiogenic agents and show for the first time that oHSVs expressing angiostatin and IL-12 can improve efficacy in human GBM models.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiostatinas/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Interleucina-12/farmacología , Virus Oncolíticos/genética , Simplexvirus/genética , Angiostatinas/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/virología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Glioblastoma/metabolismo , Glioblastoma/virología , Humanos , Inyecciones Intralesiones , Interleucina-12/genética , Ratones , Ratones Desnudos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 8(3): e59281, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555012

RESUMEN

Platelets play an important role in thrombosis and in neo-vascularisation as they release and produce factors that both promote and suppress angiogenesis. Amongst these factors is the angiogenesis inhibitor angiostatin, which is released during thrombus formation. The impact of anti-thrombotic agents and the kinetics of platelet angiostatin release are unknown. Hence, our objectives were to characterize platelet angiostatin release temporally and pharmacologically and to determine how angiostatin release influences endothelial cell migration, an early stage of angiogenesis. We hypothesized anti-platelet agents would suppress angiostatin release but not generation by platelets. Human platelets were aggregated and temporal angiostatin release was compared to vascular endothelial growth factor (VEGF). Immuno-gold electron microscopy and immunofluorescence microscopy identified α-granules as storage organelles of platelet angiostatin. Acetylsalicylic acid, MRS2395, GPIIb/IIIa blocking peptide, and aprotinin were used to characterize platelet angiostatin release and generation. An endothelial cell migration assay was performed under hypoxic conditions to determine the effects of pharmacological platelet and angiostatin inhibition. Compared to VEGF, angiostatin generation and release from α-granules occurred later temporally during platelet aggregation. Consequently, collagen-activated platelet releasates stimulated endothelial cell migration more potently than maximally-aggregated platelets. Platelet inhibitors prostacyclin, S-nitroso-glutathione, acetylsalicylic acid, and GPIIb/IIIa blocking peptide, but not a P2Y12 inhibitor, suppressed angiostatin release but not generation. Suppression of angiostatin generation in the presence of acetylsalicylic acid enhanced platelet-stimulated endothelial migration. Hence, the temporal and pharmacological modulation of platelet angiostatin release may have significant consequences for neo-vascularization following thrombus formation.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiostatinas/farmacología , Plaquetas/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Aspirina/farmacología , Plaquetas/metabolismo , Hipoxia de la Célula , Línea Celular , Colágeno/farmacología , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/citología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Epoprostenol/farmacología , Humanos , Péptidos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , S-Nitrosoglutatión/farmacología , Valeratos/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
8.
Curr Alzheimer Res ; 10(3): 252-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23534534

RESUMEN

We have examined the anti-angiogenic compound, angiostatin as a modulator of inflammatory reactivity and vascular responses and for neuroprotection in an animal model of Alzheimer's disease (AD). Intra-hippocampal amyloidbeta (Aß1₋42) injection, relative to controls phosphate buffer saline (PBS) or reverse peptide Aß42₋1, increased gliosis in the molecular layer (ML) of rat hippocampus. Vascular remodeling was indicated from increased microvessel immunoreactivity (ir) in ML suggesting the possibility of an angiogenic response to peptide injection. Administration of Aß1₋42 also induced a loss of neurons in the granule cell region of hippocampus relative to controls. Treatment of peptide-injected rats with angiostatin was associated with a spectrum of modulatory effects including reduced microgliosis (by 34%), diminished microvessel ir (by 36%) and increased neuronal viability (by 31%) compared with peptide injection alone. Angiostatin treatment was ineffective in reducing astrogliosis induced by Aß1₋42 and applied alone the compound had no significant effect to alter gliosis, microvessel ir or neuronal viability compared with PBS control. In vitro, angiostatin significantly attenuated secretion of the pro-angiogenic agent, vascular endothelial growth factor (VEGF) in lipopolysaccharide (LPS)-stimulated THP-1 cells. Our findings provide novel evidence for a broad spectrum of angiostatin effects in an animal model of AD including actions to reduce inflammatory reactivity, stabilize vascular remodeling and confer neuroprotection. The overall effects of angiostatin are consistent with actions of the compound to inhibit microglial secretion of VEGF.


Asunto(s)
Enfermedad de Alzheimer/patología , Inhibidores de la Angiogénesis/farmacología , Angiostatinas/farmacología , Encéfalo/efectos de los fármacos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Bull Math Biol ; 75(2): 274-87, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23292363

RESUMEN

The present work formulates and analyzes the inhibitory effect of anti-angiogenic factor angiostatin excreted by the primary tumor on metastatic tumor angiogenesis, blood perfusion, and interstitial fluid flow in the tumor microenvironment by means of a numerical experiment. The simulation results demonstrate that angiostatin has an obvious impact on the morphology, growth rate, and the number of branches of microvascular network inside and outside the metastatic tumor, and angiostatin has the capacity to regulate and inhibit the formation of new blood vessels. Heterogeneous blood perfusion, widespread interstitial hypertension, and low convection within the metastatic tumor have obviously improved under the inhibitory effect of angiostatin, which are consistent with physiological observed facts. The simulation results may provide beneficial information and theoretical models for clinical research of anti-angiogenic therapy strategies.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiostatinas/farmacología , Modelos Biológicos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Angiostatinas/uso terapéutico , Simulación por Computador , Humanos , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico
10.
Mol Imaging Biol ; 15(3): 239-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23054555

RESUMEN

PURPOSE: There is a real need to adapt simple and reproducible imaging methodologies to evaluate noninvasively pro- and antiangiogenic activities of new treatments in a physiological context in mice. PROCEDURE: The angiogenic response to fibroblast growth factor 2 (FGF-2) in a model of subcutaneously implanted cellulose sponges was measured in parallel after an intravenous injection of a fluorescent αvß3 integrin-targeting molecule (Angiolone(TM)) and an fluorescence diffuse optical tomography optical imaging system and by measuring the hemoglobin content in the sponges. RESULTS: Optical measurements of angiogenesis correlated perfectly with the values obtained using hemoglobin quantification. This assay can be used to follow the activity of a pro- or antiangiogenic treatment like demonstrated after FGF-2 or angiostatin, respectively. CONCLUSION: The perfectly controlled quality of cellulose sponges combined to this noninvasive optical method allow rapid, accurate, and reproducible measurements of angiogenic activities in vivo at the preclinical level.


Asunto(s)
Imagen Molecular/métodos , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Oligopéptidos/farmacología , Tejido Subcutáneo/irrigación sanguínea , Tapones Quirúrgicos de Gaza , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Angiostatinas/farmacología , Angiostatinas/uso terapéutico , Animales , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fluorescencia , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Tejido Subcutáneo/efectos de los fármacos
11.
Rheumatol Int ; 33(4): 879-85, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22811011

RESUMEN

Angiogenesis is controlled by a variety of angiogenesis stimulators and inhibitors. The increased power Doppler (PD) signals determined by ultrasonography is an indirect marker of synovial vascularity in arthritis. We aimed to investigate relationship between ultrasonographic findings and synovial angiogenesis modulators. Thirteen Behcet's disease (BD), 15 spondyloarthropathy, 21 rheumatoid arthritis (RA), and 15 osteoarthritis (OA) patients with knee arthritis were included. Cumulative effusion, synovial hypertrophy, and PD signal scores were calculated in arthritic joints. In synovial fluid samples, angiogenesis inhibitors (angiostatin, thrombospondin-1, and endostatin) and stimulators [bFGF (basic fibroblast growth factor), angiopoietin-1] were studied. The comparisons between groups were made by Kruskal-Wallis test, and correlation analysis was calculated with Pearson and Spearman tests. Effusion scores were significantly higher in inflammatory arthritis than in OA. Synovial hypertrophy scores were higher in RA and spondylarthritis than in OA and BD. PD scores were not different between the groups. Synovial angiostatin and bFGF levels were significantly higher in patients with inflammatory arthritis than in OA. Cumulative effusion scores were positively correlated with angiopoietin-1, angiostatin, and bFGF and negatively correlated with thrombospondin-1 levels. Synovial hypertrophy scores were positively correlated with angiostatin and bFGF levels and negatively correlated with thrombospondin-1. No correlation was found between PD scores and modulators of angiogenesis. In large joints like knee, detecting PD signals alone was not sufficient to assess the angiogenesis. However, cumulative activity scores were positively correlated with angiogenesis stimulators. Therefore, when investigating the angiogenesis, PD technique should be added to gray-scale examinations.


Asunto(s)
Artritis Reumatoide/diagnóstico por imagen , Síndrome de Behçet/diagnóstico por imagen , Articulación de la Rodilla/diagnóstico por imagen , Osteoartritis de la Rodilla/diagnóstico por imagen , Espondiloartropatías/diagnóstico por imagen , Líquido Sinovial/efectos de los fármacos , Adulto , Inductores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/farmacología , Angiopoyetina 1/farmacología , Angiostatinas/farmacología , Endostatinas/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Masculino , Persona de Mediana Edad , Trombospondina 1/farmacología , Ultrasonografía
12.
Toxicol Ind Health ; 29(6): 490-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22393105

RESUMEN

Antiangiogenic therapy is supposed to be an attractive approach for antitumor treatment. Human plasminogen-derived angiostatin K1-3 is one of the most potent antiangiogenic agents known currently. However, it is unclear whether angiostatin has got protective effects on colon cancer. So we investigated the protective effects of angiostatin on 1,2-dimethylhydrazine (DMH)-induced colon cancer in mice. Thirty Balb/C male mice, weighing 25-30 g and 8 weeks of age, were used. Twenty of the mice were treated with DMH subcutaneously (20 mg/kg) once a week for 12 weeks. Six mice died during the DMH injection and surviving mice were divided into two groups (7 mice in DMH and 7 mice in DMH + angiostatin groups). In the angiostatin group, 6 weeks after the last DMH injection the animals were first treated with angiostatin (20 µg/mouse) intraperitoneally and then subcutaneously every 48 h (5 µg/mouse) throughout a period of 12 weeks. The animals were killed after 30 weeks for histopathological examination. When we look at the distribution of lesions in the colon, they mainly occurred in the distal colon. The incidence of mean colonic lesions in a tumor-bearing mouse was 9.85 ± 4.91 in those treated with DMH and 8.71 ± 3.49 in those treated with angiostatin. The incidence of colon tumors was not significantly affected by low dose of angiostatin, and we noticed that the number of lesions decreased by 12% in DMH + angiostatin group compared to the number of the lesions in DMH group, but this decrease was not statistically significant (p > 0.05). The administration period of angiostatin corresponds to the precancerous period and the reduction in the number of lesions could be important for the protective function of angiostatin in DMH + angiostain group. We assume that therapeutic effects of angiostatin are related to its doses, route of administration, frequency and administration period. In addition, we believe that combination of high doses of angiostatin with radiation, gene therapy or chemotherapy might be successful in proper tumor model.


Asunto(s)
Angiostatinas/farmacología , Neoplasias del Colon/tratamiento farmacológico , 1,2-Dimetilhidrazina , Análisis de Varianza , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Histocitoquímica , Masculino , Ratones , Ratones Endogámicos BALB C
13.
Vestn Oftalmol ; 128(4): 62-5, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-22994111

RESUMEN

Increased plasminogen level in tear fluid was found within 28 days and increased plasmin activity in 1-3 and 21 days after alkali burn of cornea, this is the time of cornel ulcers development. Increased plasminogen level and plasmin activity in cornea, conjunctiva and intraocular fluid was found in three days after trauma. Subconjunctival injections of angiostatin K1-4,5 (a product of plasminogen metabolism) during 3 weeks resulted in significant suppression of corneal neovascularization within 14 days and of active branching of the vessels in the following. The use of angiostatin reduced depth and area of corneal ulcers. Obtained data shows the promising potential of development of medications based on angiostatin K1-4,5 for suppression of corneal neovascularization and for treatment of diseases associated with corneal ulceration.


Asunto(s)
Angiostatinas , Córnea/metabolismo , Neovascularización de la Córnea/metabolismo , Úlcera de la Córnea , Quemaduras Oculares , Neovascularización Patológica/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Angiostatinas/administración & dosificación , Angiostatinas/metabolismo , Angiostatinas/farmacología , Animales , Humor Acuoso/metabolismo , Conjuntiva/metabolismo , Córnea/irrigación sanguínea , Córnea/fisiopatología , Neovascularización de la Córnea/etiología , Neovascularización de la Córnea/fisiopatología , Úlcera de la Córnea/tratamiento farmacológico , Úlcera de la Córnea/etiología , Úlcera de la Córnea/metabolismo , Úlcera de la Córnea/fisiopatología , Descubrimiento de Drogas , Quemaduras Oculares/inducido químicamente , Quemaduras Oculares/complicaciones , Quemaduras Oculares/fisiopatología , Modelos Animales , Neovascularización Patológica/etiología , Neovascularización Patológica/fisiopatología , Plasminógeno/metabolismo , Conejos
14.
Bioorg Khim ; 37(3): 319-26, 2011.
Artículo en Ruso | MEDLINE | ID: mdl-21899046

RESUMEN

The influence of angiostatin K1-4.5--a fragment of the heavy chain of plasmin and a powerful inhibitor of angiogenesis--on kinetic parameters (k(Pg) and K(Pg)) of human Glu-plasminogen activation under the action of urokinase (uPA) not having affinity for fibrin and fibrin-specific tissue plasminogen activator (tPA) was investigated. Angiostatin does not affect the k(Pg) value, but increases the value K(Pg) urokinase plasminogen activation. A decrease in the k(Pg) value and an increase in the K(Pg) value were found for fibrin-stimulated plasminogen activation by tPA with increasing concentrations of angiostatin. The obtained results show that angiostatin is competitive inhibitor of the uPA activator activity, while it inhibits the activator activity of tPA by mixed type. Such an influence ofangiostatin on the kinetic constants ofthe urokinase plasminogen activation suggests that angiostatin dose dependent manner replaces plasminogen in the binary enzyme-substrate complex uPA-Pg. In case of fibrin-stimulated plasminogen activation by tPA, both zymogen and tPA are bound to fibrin with formation of the effective triple tPA-Pg-fibrin complex. Angiostatin replaces plasminogen both from the fibrin surface and from the enzyme-substrate tPA-Pg complex that leads to a decrease in k(Pg) and an increase in K(Pg) of plasminogen activation. Inhibition constants by angioststin (Ki) of plasminogen-activator activities of uPA and tPA determined by Dixon method were found to be 0.59 +/- 0.04 and 0.12 +/- 0.05 microM, respectively.


Asunto(s)
Angiostatinas/fisiología , Fibrinolisina/antagonistas & inhibidores , Plasminógeno/antagonistas & inhibidores , Angiostatinas/farmacología , Fibrina/farmacología , Fibrinolisina/fisiología , Humanos , Neovascularización Fisiológica , Plasminógeno/fisiología , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología
15.
J. bras. patol. med. lab ; 47(4): 465-472, ago. 2011. ilus, graf, tab
Artículo en Inglés | LILACS | ID: lil-599781

RESUMEN

INTRODUCTION AND OBJECTIVE: Kint3-4 protein, originated from a genetic recombination of K1-3 and K1-4 human plasminogen segments, is recognized for its antiangiogenic and anti-inflammatory potential. This study aimed to evaluate the effect of Kint3-4 protein on tumor development in Swiss mice previously inoculated with Ehrlich tumor cells. METHODS: The protein fragment was obtained from Pichia pastoris cloning and transformation. After tumor cell inoculation three different protocols were used to assess tumor growth: beginning (0-6 days), peak (0-12 days) and after peak (0-18 days). We analyzed tumor growth, histomorphological characteristics and immunohistochemistry by use of CDC47 (cellular proliferation marker) and CD31 (blood vessel marker). RESULTS: Animals treated with Kint3-4 protein (150 µg/kg/48 h) showed lower tumor growth in all protocols. Based on histological assessment, inflammation and tumor areas were also reduced. Moreover, both the lowest rate of tumor cell proliferation and low microvessel density were observed in animals treated with Kint3-4 protein compared with the untreated control group. CONCLUSION: The effect of Kint3-4 recombinant protein on tumor angiogenesis and control of malignant cell proliferation enhances the prospects of its use in clinical and antiangiogenic treatment.


INTRODUÇÃO E OBJETIVO: A proteína Kint3-4 originou-se a partir de uma recombinação genética dos segmentos K1-3 e K1--4 do plasminogênio humano e é reconhecida por seu potencial anti-inflamatório e antiangiogênico. Este estudo teve como objetivo avaliar o efeito da proteína Kint3-4 no desenvolvimento de tumores em camundongos inoculados com células do tumor de Ehrlich. MÉTODOS: O fragmento de proteína foi obtido por uma técnica de clonagem e transformação de Pichia pastoris. Três diferentes protocolos foram avaliados após a inoculação das células tumorais: no início (0-6 dias), no pico (0-12 dias) e após o pico (0-18 dias) de crescimento do tumor. Foram analisados o crescimento do tumor e as características histomorfológica e imuno-histoquímica com CDC47 (marcador de proliferação celular) e CD31 (marcador de vasos sanguíneos). RESULTADOS: Os animais tratados com a proteína Kint3-4 (150 µg/kg/48 h) nos três diferentes protocolos apresentaram menor crescimento do tumor. Áreas de inflamação e tumor também foram reduzidas, avaliadas por exame histológico. Além disso, a menor taxa de proliferação das células tumorais e a baixa densidade de microvasos foram observadas nos animais tratados com proteína Kint3-4 em comparação com o grupocontrole. CONCLUSÃO: A participação da proteína recombinante Kint3-4 na angiogênese tumoral e no controle da proliferação de células malignas abre perspectivas para seu uso no tratamento clínico como antiangiogênico.


Asunto(s)
Animales , Ratones , Angiostatinas/farmacología , Neoplasias , Neovascularización Patológica , Proliferación Celular
16.
Hum Gene Ther ; 22(5): 523-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21443427

RESUMEN

Age-related macular degeneration (AMD) is a complex disease that has two phases: a degenerative phase often referred to as nonneovascular AMD (non-NVAMD) or dry AMD and a phase dominated by growth of new blood vessels in the subretinal space, referred to as NVAMD or wet AMD. Advances in the understanding of the molecular pathogenesis of NVAMD have led to new drug therapies that have provided major benefits to patients. However, those treatments require frequent intraocular injections that in many patients must be continued indefinitely to maintain visual benefits. Gene transfer to augment expression of endogenous antiangiogenic proteins is an alternative approach that has the potential to provide long-term stability in patients with NVAMD. Studies in animal models that mimic aspects of NVAMD have identified several possible transgenes, and a clinical trial in patients with advanced NVAMD has suggested that the approach may be feasible. Many important questions remain, but the rationale and preliminary data are compelling. The results of two ongoing clinical trials may answer several of the questions and help direct future research.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Degeneración Macular Húmeda/fisiopatología , Degeneración Macular Húmeda/terapia , Angiostatinas/administración & dosificación , Angiostatinas/farmacología , Dependovirus , Endostatinas/administración & dosificación , Endostatinas/farmacología , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/farmacología , Vectores Genéticos , Humanos , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/farmacología , Serpinas/administración & dosificación , Serpinas/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 1 de Factores de Crecimiento Endotelial Vascular/farmacología
17.
Biol Chem ; 392(4): 347-56, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21194375

RESUMEN

Many proteins in the fibrinolysis pathway contain antiangiogenic kringle domains. Owing to the high degree of homology between kringle domains, there has been a safety concern that antiangiogenic kringles could interact with common kringle proteins during fibrinolysis leading to adverse effects in vivo. To address this issue, we investigated the effects of several antiangiogenic kringle proteins including angiostatin, apolipoprotein(a) kringles IV(9)-IV(10)-V (LK68), apolipoprotein(a) kringle V (rhLK8) and a derivative of rhLK8 mutated to produce a functional lysine-binding site (Lys-rhLK8) on the entire fibrinolytic process in vitro and analyzed the role of lysine binding. Angiostatin, LK68 and Lys-rhLK8 increased clot lysis time in a dose-dependent manner, inhibited tissue-type plasminogen activator-mediated plasminogen activation on a thrombin-modified fibrinogen (TMF) surface, showed binding to TMF and significantly decreased the amount of plasminogen bound to TMF. The inhibition of fibrinolysis by these proteins appears to be dependent on their functional lysine-binding sites. However, rhLK8 had no effect on these processes owing to an inability to bind lysine. Collectively, these results indicate that antiangiogenic kringles without lysine binding sites might be safer with respect to physiological fibrinolysis than lysine-binding antiangiogenic kringles. However, the clinical significance of these findings will require further validation in vivo.


Asunto(s)
Apolipoproteínas A/química , Apolipoproteínas A/farmacología , Fibrinólisis/efectos de los fármacos , Kringles , Lisina , Plasminógeno/química , Plasminógeno/farmacología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Angiostatinas/efectos adversos , Angiostatinas/química , Angiostatinas/metabolismo , Angiostatinas/farmacología , Apolipoproteínas A/efectos adversos , Apolipoproteínas A/metabolismo , Sitios de Unión , Relación Dosis-Respuesta a Droga , Fibrina/metabolismo , Fibrinógeno/química , Fibrinógeno/metabolismo , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Plasminógeno/efectos adversos , Plasminógeno/metabolismo , Trombina/química , Trombina/metabolismo , Activador de Tejido Plasminógeno/metabolismo
18.
J Microbiol Biotechnol ; 20(10): 1430-5, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21030828

RESUMEN

Two angiostatic fusion proteins (hAE and hEA) of human angiostatin (hAS) and endostatin (hES) proteins differed in tandem connection manner were constructed and evaluated for synergistic anti-angiogenic effects. The 65 kDa secreted fusion proteins from Pichia pastoris expression were verified by mass-spec analysis and western blotting assay. Luciferase reporter gene assay using VEGF promoter revealed that angiostatin-endostatin fusion protein (hAE) and its corresponding fusion gene delivery on Human Microvascular Endothelial Cells (HMEC-1) resulted in more potent synergistic anti-angiogenic effects than endostatin-angiostatin fusion protein (hEA). These facts suggest that the orientation of fusion genes between hAS and hES might be an important factor for developing therapeutic proteins.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiostatinas/genética , Angiostatinas/farmacología , Endostatinas/genética , Endostatinas/farmacología , Expresión Génica , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Angiostatinas/metabolismo , Sinergismo Farmacológico , Endostatinas/metabolismo , Células Endoteliales/efectos de los fármacos , Humanos , Pichia/genética , Pichia/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología
19.
J Pharmacol Sci ; 112(4): 432-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20308796

RESUMEN

Angiostatin (AS), a proteolytic fragment of plasminogen, is a potent antiangiogenic factor. It was reported that AS attenuates the vasodilatory response to vascular endothelial growth factor (VEGF) in isolated interventricular arterioles. Here, we investigated the effect of AS on nitric oxide (NO) production in human umbilical vein endothelial cells (HUVECs). AS inhibited VEGF-stimulated NO production in a dose-dependent manner, whereas AS alone did not affect basal NO production. Disruption of kringle structures by reduction of disulfide bonds resulted in the loss of the inhibitory effect of AS on VEGF-stimulated NO production. To elucidate how AS might impair VEGF activation of endothelial NO synthase (eNOS), we further examined whether AS would affect Ca(2+)-dependent and -independent pathways of eNOS activation. AS had no effect on the transient increase in cytosolic Ca(2+) levels elicited by VEGF. In contrast, AS prevented VEGF-potentiated eNOS phosphorylation at Ser1177. These results clearly indicate that AS inhibits VEGF-stimulated NO production in HUVECs without affecting basal NO production. The kringle structures of AS are required for this effect, and impairment of Ser1177 phosphorylation of eNOS might be involved in the inhibition of VEGF-stimulated NO production by AS.


Asunto(s)
Angiostatinas/farmacología , Endotelio Vascular/efectos de los fármacos , Óxido Nítrico/biosíntesis , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Calcio/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Activación Enzimática , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Factor A de Crecimiento Endotelial Vascular/fisiología
20.
J Cell Mol Med ; 14(10): 2519-30, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20050964

RESUMEN

Kringle 5(K5) is the fifth kringle domain of human plasminogen and its anti-angiogenic activity is more potent than angiostatin that includes the first four kringle fragment of plasminogen. Our recent study demonstrated that K5 suppressed hepatocarcinoma growth by anti-angiogenesis. To find high efficacy and minimal peptide sequence required for the anti-angiogenic and anti-tumour activities of K5, two deletion mutants of K5 were generated. The amino acid residues outside kringle domain of intact K5 (Pro452-Ala542) were deleted to form K5mut1(Cys462-Cys541). The residue Cys462 was deleted again to form K5mut2(Met463-Cys541). K5mut1 specifically inhibited proliferation, migration and induced apoptosis of endothelial cells, with an apparent two-fold enhanced activity than K5. Intraperitoneal injection of K5mut1 resulted in more potent tumour growth inhibition and microvessel density reduction than K5 both in HepA-grafted and Bel7402-xenografted hepatocarcinoma mouse models. These results suggested that K5mut1 has more potent anti-angiogenic activity than intact K5. K5mut2, which lacks only the amino terminal cysteine of K5mut1, completely lost the activity, suggesting that the kringle domain is essential for the activity of K5. The activity was enhanced to K5mut1 level when five acidic amino acids of K5 in NH(2) terminal outside kringle domain were replaced by five serine residues (K5mut3). The shielding effect of acidic amino acids may explain why K5mut1 has higher activity. K5, K5mut1 and K5mut3 held characteristic ß-sheet spectrum while K5mut2 adopted random coil structure. These results suggest that K5mut1 with high efficacy is the minimal active peptide sequence of K5 and may have therapeutic potential in liver cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Angiostatinas/farmacología , Antineoplásicos/farmacología , Kringles , Plasminógeno/farmacología , Secuencia de Aminoácidos , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neovascularización Patológica/tratamiento farmacológico , Distribución Aleatoria , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA