Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
J Dent Res ; 103(1): 101-110, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38058134

RESUMEN

Adding dental pulp stem cells (DPSCs) to vascular endothelial cell-formed vessel-like structures can increase the longevity of these vessel networks. DPSCs display pericyte-like cell functions and closely assemble endothelial cells (ECs). However, the mechanisms of DPSC-derived pericyte-like cells in stabilizing the vessel networks are not fully understood. In this study, we investigated the functions of E-DPSCs, which were DPSCs isolated from the direct coculture of human umbilical vein endothelial cells (HUVECs) and DPSCs, and T-DPSCs, which were DPSCs treated by transforming growth factor beta 1 (TGF-ß1), in stabilizing blood vessels in vitro and in vivo. A 3-dimensional coculture spheroid sprouting assay was conducted to compare the functions of E-DPSCs and T-DPSCs in vitro. Dental pulp angiogenesis in the severe combined immunodeficiency (SCID) mouse model was used to explore the roles of E-DPSCs and T-DPSCs in vascularization in vivo. The results demonstrated that both E-DPSCs and T-DPSCs possess smooth muscle cell-like cell properties, exhibiting higher expression of the mural cell-specific markers and the suppression of HUVEC sprouting. E-DPSCs and T-DPSCs inhibited HUVEC sprouting by activating TEK tyrosine kinase (Tie2) signaling, upregulating vascular endothelial (VE)-cadherin, and downregulating vascular endothelial growth factor receptor 2 (VEGFR2). In vivo study revealed more perfused and total blood vessels in the HUVEC + E-DPSC group, HUVEC + T-DPSC group, angiopoietin 1 (Ang1) pretreated group, and vascular endothelial protein tyrosine phosphatase (VE-PTP) inhibitor pretreated group, compared to HUVEC + DPSC group. In conclusion, these data indicated that E-DPSCs and T-DPSCs could stabilize the newly formed blood vessels and accelerate their perfusion. The critical regulating pathways are Ang1/Tie2/VE-cadherin and VEGF/VEGFR2 signaling.


Asunto(s)
Células Madre , Factor A de Crecimiento Endotelial Vascular , Animales , Ratones , Humanos , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Madre/fisiología , Angiopoyetina 1/farmacología , Angiopoyetina 1/metabolismo , Pulpa Dental , Células Endoteliales de la Vena Umbilical Humana , Cadherinas/metabolismo , Células Cultivadas
2.
Drug Des Devel Ther ; 17: 519-534, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36845667

RESUMEN

Background: Bone dysfunction is a crucial problem that occurs during rheumatoid arthritis (RA) disease. Osteoclast plays a significant role in bone resorption and osteoclast differentiation and its enhancement of bone destruction. Edaravone remarkably exhibited free radical scavenging and anti-inflammatory effects. The objective of the current investigation is to comfort the inhibitory effect of Edaravone (ED) against complete Freund adjuvant (CFA) rat model via inhibition of angiogenesis and inflammation. Methods: Subcutaneous injection of CFA (1%) was used to induce arthritis; the rats were divided into different groups and received the oral administration of ED. Paw edema, body weight, and arthritis score were regularly estimated. Biochemical parameters were estimated, respectively. We also estimate the level of hypoxia-inducible factor-1α (HIF-1α), angiopoietin 1 (ANG-1), and vascular endothelial growth factor (VEGF). We also checked into how ED affected the differentiation of osteoclasts utilising a co-culture system with monocytes and synovial fibroblasts in arthritis rats. Results: ED treatment significantly (P<0.001) suppressed the arthritis score and paw edema and improved the body weight. ED treatment significantly (P<0.001) altered the antioxidant parameters and pro-inflammatory cytokines: inflammatory mediator nuclear kappa B factor (NF-κB), cyclooxygenase-2 (COX-2), and prostaglandin E2 (PGE2), respectively. Furthermore, ED treatment significantly (P<0.001) suppressed the level of ANG-1, HIF-1α, and VEGF, respectively. The results suggest that ED suppressed osteoclast differentiation and also decreased the level of cytokines and osteopontin (OPN), receptor activator for nuclear factor-κ B Ligand (RANKL) and macrophage colony stimulating factor (M-CSF) in the co-culture supernatant of monocytes and synovial fibroblasts. Conclusion: Edaravone could mitigate CFA via inhibiting angiogenesis and inflammatory reactions, which may be linked with the HIF-1α-VEGF-ANG-1 axis and also enhance the bone destruction of murine arthritis via suppression of osteoclast differentiation and inflammatory reaction.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Ratas , Ratones , Animales , Osteoclastos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adyuvante de Freund/metabolismo , Adyuvante de Freund/farmacología , Edaravona/farmacología , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Citocinas/metabolismo , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo
3.
Oncogene ; 42(2): 124-137, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36385374

RESUMEN

Drug resistance from BCR-ABL tyrosine kinase inhibitors (TKIs) and other chemotherapeutics results in treatment failure and disease progression in chronic myeloid leukemia (CML). However, the mechanism is still uncertain. In this study, we investigated the role of angiopoietin-1 (ANG-1) as a potential prognostic factor for drug resistance in CML. Both intracellular and secretory ANG-1 (iANG-1 and sANG-1) were overexpressed in multidrug-resistant CML samples. The IC50 value was higher in primary CD34+ CD38- cells with more ANG-1. Silencing ANG-1significantly sensitized three TKI-resistant CML cell lines to imatinib (IM) while recombinant human ANG-1 failed to retain cell survival in vitro. This indicated the important role of iANG-1 as opposed to sANG-1 in CML drug resistance. Moreover, a similar effect was observed in xenograft mice models bearing ANG-1-silenced CML cells. Subsequently, pathway analysis and protein validation experiments showed activation of the JAK/STAT pathway and augmentation of STAT5a phosphorylation in ANG-1 restored CML cells. Upstream Src phosphorylation, which plays a crucial role in CML drug resistance, was also upregulated as a key event in iANG-1-related JAK/STAT pathway activation. In conclusion, our study elucidated a new BCR-ABL independent molecular mechanism induced by intracytoplasmic ANG-1 overexpression as a potential strategy for overcoming CML resistance.


Asunto(s)
Proteínas de Fusión bcr-abl , Leucemia Mielógena Crónica BCR-ABL Positiva , Animales , Humanos , Ratones , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Apoptosis , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Quinasas Janus , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal , Factores de Transcripción STAT/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo
4.
Curr Neurovasc Res ; 20(1): 140-148, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35418285

RESUMEN

AIMS: To explore angiopoietin-1 (Ang-1) involved in cerebral vasospasm (CVS) after aneurysmal subarachnoid hemorrhage (aSAH) through its effect on endoplasmic reticulum stress (ERS) and apoptosis of vascular endothelial cells (VECs). BACKGROUND: CVS accounts for high morbidity and mortality of aSAH. Abnormal cellular physiological processes of VECs play a critical role in aSAH-induced CVS. In addition, Ang-1 is involved in regulating vascular structure and function. OBJECTIVE: To study the role of Ang-1 played in CVS and the underlying mechanism. METHODS: Blood samples of 130 aSAH patients were collected from 2016 to 2020 at West China Hospital of Sichuan University. A two-hemorrhage rodent model was employed to structure an aSAH-induced CVS rat model. Moreover, oxyHb was used to treat VECs to construct a CVS cell model in vitro. ELISA was used to measure the level of Ang-1 and HE staining to assess the rat's basilar arteries. Subsequently, CCK-8 was used to detect cell viability ability, and flow cytometry was used to test the cell apoptosis rate. Western blotting was used to determine the expression level of ERS marker and apoptosis-related proteins. RESULTS: There was an abnormally low expression of Ang-1 in CVS patients and CVS rats; besides, oxyHb treatment decreased Ang-1 in VECs in a concentration-dependent manner. Ang-1 treatment led to the thinner basilar artery wall and lumen circumference in CVS rats; moreover, in oxyHbtreated VECs, Ang-1 treatment inhibited ERS and apoptosis. In addition, the expression of p-PI3K and p-Akt in the CVS group decreased, while the expression of p53 in the CVS group increased. The expression of p-PI3K and p-Akt in 8 CVS rats negatively correlates with the expression of Ang- 1, but the correlation between p53 and Ang-1 was positive. Furthermore, the results suggested that Ang-1 suppressed ERS and apoptosis of VECs through the regulated PI3K/Akt/p53 pathway. CONCLUSION: Elevated Ang-1 inhibited p53-mediated ERS and apoptosis of VECs through the activated PI3K/Akt pathway; Ang-1 might be an attractive treatment strategy for CVS.


Asunto(s)
Estrés del Retículo Endoplásmico , Hemorragia Subaracnoidea , Vasoespasmo Intracraneal , Animales , Ratas , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis , Células Endoteliales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Hemorragia Subaracnoidea/tratamiento farmacológico , Proteína p53 Supresora de Tumor/metabolismo
5.
J Pharmacol Sci ; 150(3): 135-145, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36184118

RESUMEN

Histamine is a major neurotransmitter and alleviates neuronal damage after ischemic injury via H2 receptors. Herein, we investigated the effects of H2 receptor agonists on the blood-brain barrier (BBB) disruption after traumatic brain injury (TBI). Male ddY mice were used to generate the TBI model, in which a fluid percussion injury (FPI) was induced by a hydraulic impact. The BBB disruption was evaluated using Evans blue extravasation. H2 receptor agonists, amthamine and dimaprit, were administered into the lateral cerebroventricle (i.c.v.) or tail vein (i.v.) from 3 hours to 3 days after FPI. The i.c.v. or i.v. administration of amthamine and dimaprit reduced FPI-induced Evans blue extravasation and promoted mRNA expression of vascular protective factors, including angiopoietin-1 and sonic hedgehog. The co-administration of ranitidine, a H2 receptor antagonist, inhibited these effects. Expression of the H2 receptor was observed in astrocytes and brain microvascular endothelial cells (BMECs) in the injured cortex. Treatment with amthamine and dimaprit promoted mRNA expression of vascular protective factors in astrocytes and BMECs. These results suggest that H2 receptor agonists alleviate TBI-induced BBB disruption by increasing the expression of vascular protective factors in astrocytes and BMECs.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Agonistas de los Receptores Histamínicos , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Dimaprit/metabolismo , Dimaprit/farmacología , Células Endoteliales/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Proteínas Hedgehog , Histamina/farmacología , Agonistas de los Receptores Histamínicos/metabolismo , Agonistas de los Receptores Histamínicos/farmacología , Masculino , Ratones , Factores Protectores , ARN Mensajero/metabolismo , Ranitidina/metabolismo , Ranitidina/farmacología , Receptores Histamínicos H2/genética , Receptores Histamínicos H2/metabolismo , Tiazoles
6.
Cells ; 11(18)2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36139399

RESUMEN

Numerous studies have demonstrated that biological compounds and trace elements such as dopamine (DA) and copper ions (Cu) could be modified onto the surfaces of scaffolds using a one-step immersion process which is simple, inexpensive and, most importantly, non-cytotoxic. The development and emergence of 3D printing technologies such as selective laser melting (SLM) have also made it possible for us to fabricate bone scaffolds with precise structural designs using metallic compounds. In this study, we fabricated porous titanium scaffolds (Ti) using SLM and modified the surface of Ti with polydopamine (PDA) and Cu. There are currently no other reported studies with such a combination for osteogenic and angiogenic-related applications. Results showed that such modifications did not affect general appearances and microstructural characteristics of the porous Ti scaffolds. This one-step immersion modification allowed us to modify the surfaces of Ti with different concentrations of Cu ions, thus allowing us to fabricate individualized scaffolds for different clinical scenarios. The modification improved the hydrophilicity and surface roughness of the scaffolds, which in turn led to promote cell behaviors of Wharton's jelly mesenchymal stem cells. Ti itself has high mechanical strength, therefore making it suitable for surgical handling and clinical applications. Furthermore, the scaffolds were able to release ions in a sustained manner which led to an upregulation of osteogenic-related proteins (bone alkaline phosphatase, bone sialoprotein and osteocalcin) and angiogenic-related proteins (vascular endothelial growth factor and angiopoietin-1). By combining additive manufacturing, Ti6Al4V scaffolds, surface modification and Cu ions, the novel hybrid 3D-printed porous scaffold could be fabricated with ease and specifically benefited future bone regeneration in the clinic.


Asunto(s)
Titanio , Oligoelementos , Fosfatasa Alcalina , Aleaciones , Angiopoyetina 1/farmacología , Regeneración Ósea , Cobre/farmacología , Dopamina , Indoles , Sialoproteína de Unión a Integrina , Osteocalcina , Polímeros , Porosidad , Impresión Tridimensional , Titanio/química , Titanio/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Sci Rep ; 12(1): 15531, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109537

RESUMEN

Community acquired pneumonia, mainly caused by Streptococcus pneumoniae (S.pn.), is a common cause of death worldwide. Despite adequate antibiotic therapy, pneumococcal pneumonia can induce pulmonary endothelial hyperpermeability leading to acute lung injury, which often requires mechanical ventilation (MV) causing ventilator-induced lung injury (VILI). Endothelial stabilization is mediated by angiopoietin-1 induced Tie2 activation. PEGylated (polyethylene glycol) Tie2-agonist Vasculotide (VT) mimics Angiopietin-1 effects. Recently, VT has been shown to reduce pulmonary hyperpermeability in murine pneumococcal pneumonia. The aim of this study was to determine whether VT reduces lung damage in S.pn. infected and mechanically ventilated mice. Pulmonary hyperpermeability, immune response and bacterial load were quantified in S.pn. infected mice treated with Ampicillin + /-VT and undergoing six hours of MV 24 h post infection. Histopathological lung changes, Tie2-expression and -phosphorylation were evaluated. VT did not alter immune response or bacterial burden, but interestingly combination treatment with ampicillin significantly reduced pulmonary hyperpermeability, histological lung damage and edema formation. Tie2-mRNA expression was reduced by S.pn. infection and/or MV but not restored by VT. Moreover, Tie2 phosphorylation was not affected by VT. These findings indicate that VT may be a promising adjunctive treatment option for prevention of VILI in severe pneumococcal pneumonia.


Asunto(s)
Neumonía Neumocócica , Receptor TIE-2/agonistas , Lesión Pulmonar Inducida por Ventilación Mecánica , Ampicilina/farmacología , Angiopoyetina 1/farmacología , Animales , Antibacterianos/farmacología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Permeabilidad , Neumonía Neumocócica/tratamiento farmacológico , Polietilenglicoles/farmacología , ARN Mensajero/farmacología , Respiración Artificial , Streptococcus pneumoniae , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control
8.
Mater Sci Eng C Mater Biol Appl ; 135: 112673, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35581065

RESUMEN

Scaffolds combined with bioactive agents can enhance bone regeneration at therapeutic sites. We explore whether combined supplementation with coumaric acid and recombinant human-cartilage oligomeric matrix protein-angiopoietin 1 (rhCOMP-Ang1) is an ideal approach for bone tissue engineering. We developed coumaric acid-conjugated absorbable collagen scaffold (CA-ACS) and investigated whether implanting CA-ACS in combination with rhCOMP-Ang1 facilitates ACS- or CA-ACS-mediated bone formation using a rat model of critically sized mandible defects. We examined the mechanisms by which coumaric acid and rhCOMP-Ang1 regulate behaviors of human periodontal ligament fibroblasts (hPLFs). The CA-ACS exhibits greater anti-degradation and mechanical strength properties than does ACS alone. Implanting CA-ACS loaded with rhCOMP-Ang1 greatly enhances bone regeneration at the defect via the activation of angiogenic, osteogenic, and anti-osteoclastic responses compared with other rat groups implanted with an ACS alone or CA-ACS. Treatment with both rhCOMP-Ang1 and coumaric acid increases proliferation, mineralization, and migration of cultured hPLFs via activation of the Ang1/Tie2 signaling axis at a greater rate than treatment with either of them alone. Collectively, this study demonstrates that CA-ACS impregnated with rhCOMP-Ang1 enhances bone regeneration at therapeutic sites, and this enhancement is associated with a synergistic interaction between rhCOMP-Ang1-mediated angiogenesis and coumaric acid-related antioxidant responses.


Asunto(s)
Angiopoyetina 1 , Antioxidantes , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Animales , Antioxidantes/farmacología , Proteína de la Matriz Oligomérica del Cartílago , Colágeno/farmacología , Ácidos Cumáricos , Mandíbula , Ratas
9.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055129

RESUMEN

Tissue regeneration is often impaired in patients with metabolic disorders such as diabetes mellitus and obesity, exhibiting reduced wound repair and limited regeneration capacity. We and others have demonstrated that wound healing under normal metabolic conditions is potentiated by the secretome of human endothelial cell-differentiated mesenchymal stem cells (hMSC-EC). However, it is unknown whether this effect is sustained under hyperglycemic conditions. In this study, the wound healing effect of secretomes from undifferentiated human mesenchymal stem cells (hMSC) and hMSC-EC in a type-2 diabetes mouse model was analyzed. hMSC were isolated from human Wharton's jelly and differentiated into hMSC-EC. hMSC and hMSC-EC secretomes were analyzed and their wound healing capacity in C57Bl/6J mice fed with control (CD) or high fat diet (HFD) was evaluated. Our results showed that hMSC-EC secretome enhanced endothelial cell proliferation and wound healing in vivo when compared with hMSC secretome. Five soluble proteins (angiopoietin-1, angiopoietin-2, Factor de crecimiento fibroblástico, Matrix metallopeptidase 9, and Vascular Endothelial Growth Factor) were enriched in hMSC-EC secretome in comparison to hMSC secretome. Thus, the five recombinant proteins were mixed, and their pro-healing property was evaluated in vitro and in vivo. Functional analysis demonstrated that a cocktail of these proteins enhanced the wound healing process similar to hMSC-EC secretome in HFD mice. Overall, our results show that hMSC-EC secretome or a combination of specific proteins enriched in the hMSC-EC secretome enhanced wound healing process under hyperglycemic conditions.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Células Madre Mesenquimatosas/citología , Proteínas Recombinantes/farmacología , Cicatrización de Heridas/efectos de los fármacos , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Angiopoyetina 2/metabolismo , Angiopoyetina 2/farmacología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados/química , Diabetes Mellitus Tipo 2/inducido químicamente , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/farmacología , Células Madre Mesenquimatosas/metabolismo , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Gelatina de Wharton/citología , Gelatina de Wharton/metabolismo
10.
Pediatr Res ; 91(6): 1405-1415, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33980990

RESUMEN

BACKGROUND: Sepsis in premature newborns is a risk factor for bronchopulmonary dysplasia (BPD), but underlying mechanisms of lung injury remain unclear. Aberrant expression of endothelial cell (EC) angiopoietin 2 (ANGPT2) disrupts angiopoietin 1 (ANGPT1)/TIE2-mediated endothelial quiescence, and is implicated in sepsis-induced acute respiratory distress syndrome in adults. We hypothesized that recombinant ANGPT1 will mitigate sepsis-induced ANGPT2 expression, inflammation, acute lung injury (ALI), and alveolar remodeling in the saccular lung. METHODS: Effects of recombinant ANGPT1 on lipopolysaccharide (LPS)-induced endothelial inflammation were evaluated in human pulmonary microvascular endothelial cells (HPMEC). ALI and long-term alveolar remodeling were assessed in newborn mice exposed to intraperitoneal LPS and recombinant ANGPT1 pretreatment. RESULTS: LPS dephosphorylated EC TIE2 in association with increased ANGPT2 in vivo and in vitro. ANGPT1 suppressed LPS and ANGPT2-induced EC inflammation in HPMEC. Neonatal mice treated with LPS had increased lung cytokine expression, neutrophilic influx, and cellular apoptosis. ANGPT1 pre-treatment suppressed LPS-induced lung Toll-like receptor signaling, inflammation, and ALI. LPS-induced acute increases in metalloproteinase 9 expression and elastic fiber breaks, as well as a long-term decrease in radial alveolar counts, were mitigated by ANGPT1. CONCLUSIONS: In an experimental model of sepsis-induced BPD, ANGPT1 preserved endothelial quiescence, inhibited ALI, and suppressed alveolar simplification. IMPACT: Key message: Angiopoietin 1 inhibits LPS-induced neonatal lung injury and alveolar remodeling. Additions to existing literature: Demonstrates dysregulation of angiopoietin-TIE2 axis is important for sepsis- induced acute lung injury and alveolar simplification in experimental BPD. Establishes recombinant Angiopoietin 1 as an anti-inflammatory therapy in BPD. IMPACT: Angiopoietin 1-based interventions may represent novel therapies for mitigating sepsis-induced lung injury and BPD in premature infants.


Asunto(s)
Lesión Pulmonar Aguda , Displasia Broncopulmonar , Sepsis , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/prevención & control , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Angiopoyetina 2/metabolismo , Angiopoyetina 2/farmacología , Animales , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/prevención & control , Células Endoteliales/metabolismo , Endotoxinas/metabolismo , Endotoxinas/farmacología , Humanos , Recién Nacido , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Pulmón , Ratones
11.
Aging (Albany NY) ; 13(14): 19048-19063, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34326273

RESUMEN

Dystonia is a disorder associated with abnormalities in many brain regions including the basal ganglia and cerebellum. The toxin 3-Nitropropionic acid (3-NP) can induce neuropathologies in the mice striatum and nigra substance, including excitotoxicity, neuroinflammation, and extensive neuronal atrophy, characterized by progressive motor dysfunction, dystonia, and memory loss, mimicking those observed in humans. We established a mouse model of dystonia by administering 3-NP. Given the reported neuroprotective effects of the endothelial growth factor angiopoietin-1 (Ang-1) and the anti-inflammatory integrin αvß3 binding peptide C16, we performed this study to evaluate their combined effects on 3-NP striatal toxicity and their therapeutic potential with multiple methods using an in vivo mouse model. Sixty mice were equally and randomly divided into three groups: control, 3-NP treatment, and 3-NP+C16+Ang-1 treatment. Behavioral and electrophysiological tests were conducted and the effect of the combined C16+Ang-1 treatment on neural function recovery was determined. We found that C16+Ang-1 treatment alleviated 3-NP-induced behavioral, biochemical, and cellular alterations in the central nervous system and promoted function recovery by restoring vascular permeability and reducing inflammation in the micro-environment. In conclusion, our results confirmed the neuroprotective effect of combined C16+Ang-1 treatment and suggest their potential as a complementary therapeutic against 3-NP-induced dystonia.


Asunto(s)
Angiopoyetina 1/uso terapéutico , Encéfalo/efectos de los fármacos , Distonía/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Neuroprotección , Péptidos/uso terapéutico , Angiopoyetina 1/farmacología , Animales , Antiinflamatorios , Encéfalo/patología , Encéfalo/fisiopatología , Permeabilidad Capilar , Sistema Nervioso Central , Cuerpo Estriado , Modelos Animales de Enfermedad , Quimioterapia Combinada , Distonía/inducido químicamente , Distonía/patología , Distonía/fisiopatología , Factores de Crecimiento Endotelial , Masculino , Ratones Endogámicos C57BL , Neuronas , Nitrocompuestos , Péptidos/farmacología , Propionatos , Distribución Aleatoria
12.
Sci Rep ; 11(1): 13558, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193893

RESUMEN

In vitro differentiation of human induced pluripotent stem cells (iPSCs) into functional islets holds immense potential to create an unlimited source of islets for diabetes research and treatment. A continuous challenge in this field is to generate glucose-responsive mature islets. We herein report a previously undiscovered angiopoietin signal for in vitro islet development. We revealed, for the first time, that angiopoietins, including angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) permit the generation of islets from iPSCs with elevated glucose responsiveness, a hallmark of mature islets. Angiopoietin-stimulated islets exhibited glucose synchronized calcium ion influx in repetitive glucose challenges. Moreover, Ang2 augmented the expression of all islet hormones, including insulin, glucagon, somatostatin, and pancreatic polypeptide; and ß cell transcription factors, including NKX6.1, MAFA, UCN3, and PDX1. Furthermore, we showed that the Ang2 stimulated islets were able to regulate insulin exocytosis through actin-filament polymerization and depolymerization upon glucose challenge, presumably through the CDC42-RAC1-gelsolin mediated insulin secretion signaling pathway. We also discovered the formation of endothelium within the islets under Ang2 stimulation. These results strongly suggest that angiopoietin acts as a signaling molecule to endorse in vitro islet development from iPSCs.


Asunto(s)
Angiopoyetina 1/farmacología , Angiopoyetina 2/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Islotes Pancreáticos/metabolismo , Transducción de Señal/efectos de los fármacos , Humanos
13.
Sci Rep ; 11(1): 14021, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34234265

RESUMEN

Activation of the tyrosine kinase with Ig and epidermal growth factor homology domain 2 (Tie2) receptor by angiopoietin-1 (Ang1) is critical for vascular stabilization: it promotes survival signal transduction via auto-phosphorylation and reduces vascular permeability by strengthening tight junctions between endothelial cells. Thus, Tie2/Ang1 signaling is a promising therapeutic target for vascular diseases. However, in vivo use of existing Tie2 signaling modulators, such as recombinant Ang1, is restricted by limitations in manufacturability and stability. Here, we present a novel engineered tetra-valent agonistic antibody, ASP4021, which can specifically and fully activate the Tie2 receptor in an equivalent manner to Ang1. ASP4021 induced Tie2 self-phosphorylation and inhibited apoptosis in a human primary endothelial cell line. Additionally, single administration of ASP4021 significantly suppressed mustard-oil-induced vascular permeability in rats. ASP4021 may thus be a potential therapeutic candidate for diseases associated with vascular weakness such as diabetic retinopathy, diabetic macular edema and critical limb ischemia.


Asunto(s)
Angiopoyetina 1/metabolismo , Anticuerpos/farmacología , Receptor TIE-2/agonistas , Proteínas Recombinantes de Fusión/farmacología , Angiopoyetina 1/farmacología , Animales , Anticuerpos/genética , Apoptosis/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Ingeniería Genética , Humanos , Ligandos , Masculino , Ratones , Fosforilación , Ratas , Proteínas Recombinantes de Fusión/genética
14.
Biotechnol Bioeng ; 118(1): 423-432, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32970320

RESUMEN

Vascular leak is a key driver of organ injury in diseases, and strategies that reduce enhanced permeability and vascular inflammation are promising therapeutic targets. Activation of the angiopoietin-1 (ANG1)-Tie2 tyrosine kinase signaling pathway is an important regulator of vascular quiescence. Here we describe the design and construction of a new soluble ANG1 mimetic that is a potent activator of endothelial Tie2 in vitro and in vivo. Using a chimeric fusion strategy, we replaced the extracellular matrix (ECM) binding and oligomerization domain of ANG1 with a heptameric scaffold derived from the C-terminus of serum complement protein C4-binding protein α. We refer to this new fusion protein biologic as Hepta-ANG1, which forms a stable heptamer and induces Tie2 phosphorylation in cultured cells, and in the lung following intravenous injection of mice. Injection of Hepta-ANG1 ameliorates vascular endothelial growth factor- and lipopolysaccharide-induced vascular leakage, in keeping with the known functions of Angpt1-Tie2 in maintaining quiescent vascular stability. The new Hepta-ANG1 fusion is easy to produce and displays remarkable stability with high multimericity that can potently activate Tie2. It could be a new candidate ANG1 mimetic therapy for treatments of inflammatory vascular leak, such as acute respiratory distress syndrome and sepsis.


Asunto(s)
Angiopoyetina 1 , Proteína de Unión al Complemento C4b , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas Recombinantes de Fusión , Enfermedades Vasculares/tratamiento farmacológico , Angiopoyetina 1/biosíntesis , Angiopoyetina 1/genética , Angiopoyetina 1/farmacología , Animales , Proteína de Unión al Complemento C4b/biosíntesis , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/farmacología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Dominios Proteicos , Conejos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Sepsis/patología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
15.
CNS Neurosci Ther ; 27(1): 48-59, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33346402

RESUMEN

AIM: Vasculotide (VT), an angiopoietin-1 mimetic peptide, exerts neuroprotective effects in type one diabetic (T1DM) rats subjected to ischemic stroke. In this study, we investigated whether delayed VT treatment improves long-term neurological outcome after stroke in T1DM rats. METHODS: Male Wistar rats were induced with T1DM, subjected to middle cerebral artery occlusion (MCAo) model of stroke, and treated with PBS (control), 2 µg/kg VT, 3 µg/kg VT, or 5.5 µg/kg VT. VT treatment was initiated at 24 h after stroke and administered daily (i.p) for 14 days. We evaluated neurological function, lesion volume, vascular and white matter remodeling, and inflammation in the ischemic brain. In vitro, we evaluated the effects of VT on endothelial cell capillary tube formation and inflammatory responses of primary cortical neurons (PCN) and macrophages. RESULTS: Treatment of T1DM-stroke with 3 µg/kg VT but not 2 µg/kg or 5.5 µg/kg significantly improves neurological function and decreases infarct volume and cell death compared to control T1DM-stroke rats. Thus, 3 µg/kg VT dose was employed in all subsequent in vivo analysis. VT treatment significantly increases axon and myelin density, decreases demyelination, decreases white matter injury, increases number of oligodendrocytes, and increases vascular density in the ischemic border zone of T1DM stroke rats. VT treatment significantly decreases MMP9 expression and decreases the number of M1 macrophages in the ischemic brain of T1DM-stroke rats. In vitro, VT treatment significantly decreases endothelial cell death and decreases MCP-1, endothelin-1, and VEGF expression under high glucose (HG) and ischemic conditions and significantly increases capillary tube formation under HG conditions when compared to non-treated control group. VT treatment significantly decreases inflammatory factor expression such as MMP9 and MCP-1 in macrophages subjected to LPS activation and significantly decreases IL-1ß and MMP9 expression in PCN subjected to ischemia under HG conditions. CONCLUSION: Delayed VT treatment (24 h after stroke) significantly improves neurological function, promotes vascular and white matter remodeling, and decreases inflammation in the ischemic brain after stroke in T1DM rats.


Asunto(s)
Angiopoyetina 1/uso terapéutico , Materiales Biomiméticos/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Angiopoyetina 1/farmacología , Animales , Materiales Biomiméticos/farmacología , Células Cultivadas , Diabetes Mellitus Experimental/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/farmacología , Embarazo , Ratas , Ratas Wistar , Recuperación de la Función/fisiología , Accidente Cerebrovascular/patología , Resultado del Tratamiento
16.
Exp Biol Med (Maywood) ; 245(18): 1683-1696, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32915636

RESUMEN

C16 peptide and angiopoietin-1 (Ang-1) have been found to have anti-inflammatory activity in various inflammation-related diseases. However, their combined role in acute respiratory distress syndrome (ARDS) has not been investigated yet. The objective of this study was to investigate the effects of C16 peptide and Ang-1 in combination with lipopolysaccharide (LPS)-induced inflammatory insult in vitro and in vivo. Human pulmonary microvascular endothelial cells and human pulmonary alveolar epithelial cells were used as cell culture systems, and an ARDS rodent model was used for in vivo studies. Our results demonstrated that C16 and Ang-1 in combination significantly suppressed inflammatory cell transmigration by 33% in comparison with the vehicle alone, and decreased the lung tissue wet-to-dry lung weight ratio to a maximum of 1.53, compared to 3.55 in the vehicle group in ARDS rats. Moreover, C + A treatment reduced the histology injury score to 60% of the vehicle control, enhanced arterial oxygen saturation (SO2), decreased arterial carbon dioxide partial pressure (PCO2), and increased oxygen partial pressure (PO2) in ARDS rats, while also improving the survival rate from 47% (7/15) to 80% (12/15) and diminishing fibrosis, necrosis, and apoptosis in lung tissue. Furthermore, when C + A therapy was administered 4 h following LPS injection, the treatment showed significant alleviating effects on pulmonary inflammatory cell infiltration 24 h postinsult. In conclusion, our in vitro and in vivo studies show that C16 and Ang-1 exert protective effects against LPS-induced inflammatory insult. C16 and Ang-1 hold promise as a novel agent against LPS-induced ARDS. Further studies are needed to determine the potential for C16 and Ang-1 in combination in treating inflammatory lung diseases.


Asunto(s)
Angiopoyetina 1/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Péptidos/uso terapéutico , Sustancias Protectoras/uso terapéutico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Secuencia de Aminoácidos , Angiopoyetina 1/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Inflamación/patología , Lipopolisacáridos , Lesión Pulmonar/complicaciones , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/patología , Masculino , Monocitos/efectos de los fármacos , Monocitos/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Péptidos/química , Péptidos/farmacología , Sustancias Protectoras/farmacología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Intercambio Gaseoso Pulmonar , Ratas Wistar , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/patología
17.
Life Sci ; 256: 117894, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32502544

RESUMEN

AIMS: Pathological alterations in the brain can cause microglial activation (MA). Thus, inhibiting MA could provide a new approach for treating neurodegenerative disorders. MAIN METHODS: To investigate the effect of C16 peptide and angiopoietin-1 (Ang1) on inflammation following MA, we stimulated microglial BV-2 cells with lipopolysaccharide (LPS) and used dexmedetomidine (DEX) as a positive control. Specific inhibitors of Tie2, αvß3 and α5ß1 integrins, and PI3K/Akt were applied to investigate the neuron-protective and anti-inflammatory effects and signaling pathway of C16 + Ang1 treatment in the LPS-induced BV-2 cells. KEY FINDINGS: Our results showed that C16 + Ang1 treatment reduced the microglia M1 phenotype but promoted the microglia M2 phenotype. In addition, C16 + Ang1 treatment suppressed leukocyte migration across human pulmonary microvascular endothelial cells, reduced the levels of pro-inflammatory factors [inducible nitric oxide synthase (iNOS), interleukin (IL)-1ß, tumor necrosis factor (TNF-α)], and cellular apoptosis factors (caspase-3 and p53), and decreased lactate dehydrogenase (LDH) release, but promoted anti-inflammatory cytokine (IL-10) expression and cell proliferation in the LPS-activated BV-2 cells. The signaling pathways underlying the neuron-protective and anti-inflammatory effects of C16 + Ang1 may be mediated by Tie2-PI3K/Akt, Tie2-integrin and integrin-PI3K/Akt. SIGNIFICANCE: The neuron-protective and anti-inflammatory effects of C16 + Ang1 treatment included M1 to M2 microglia phenotype switching, blocking leukocyte transmigration, decreasing apoptotic and inflammatory factors, and promoting cellular viability.


Asunto(s)
Angiopoyetina 1/farmacología , Inflamación/patología , Lipopolisacáridos/farmacología , Microglía/patología , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Pulmón/irrigación sanguínea , Ratones , Microglía/efectos de los fármacos , Microvasos/patología , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/efectos de los fármacos , Células THP-1
18.
Neuroreport ; 31(11): 825-832, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32576769

RESUMEN

Angiopoietin-1 (Ang-1) is a new neuroprotective agent, which can protect neurons from apoptosis. Increased autophagy in neurons subjected to oxygen-glucose deprivation/recovery (OGD/R) injury may lead to autophagic cell death; therefore, the present study investigated the effect of Ang-1 on neurons subjected to OGD/R injury. Neuronal viability was detected by using the Cell Counting Kit-8, which was then used to select the appropriate concentration of Ang-1 and rapamycin used in the OGD/R injury model. The mechanistic role of Ang-1 was observed by detecting the survival rate of neurons and the level of autophagy. Results showed that Ang-1 significantly reduced neuronal cell injury induced by OGD/R and the expression of the autophagy-related proteins LC3 II/I and Beclin-1, and increased the expression of P62/SQSTM1. However, the neuroprotective effects of Ang-1 were counteracted by rapamycin, an autophagy activating agent. The changes of autophagy intensity were further confirmed by transmission electron microscopy observation of autophagosomes. Ang-1 appears to have a neuroprotective role by inhibiting autophagy expression in OGD/R. Thus, these findings could be useful for the treatment of OGD/R injury.


Asunto(s)
Angiopoyetina 1/farmacología , Autofagia/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/metabolismo , Animales , Células Cultivadas , Neuronas/patología , Ratas , Ratas Sprague-Dawley
19.
Nutr Res ; 76: 20-28, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32146252

RESUMEN

Cross talk between endothelial cells and adipocytes is vital to adipocyte functions, but little is known about the mechanisms or factors controlling the process. Angiogenesis is a critical component linking the endothelium to healthy adipogenesis, yet it is not known if or how it is involved in adipocyte physiology. Therefore, the purpose of this study was to determine the effect of angiopoietin-1 (Ang-1) and -2 (Ang-2) as well as their receptor, Tie-2, on adipocyte physiology. 3T3-L1 pre- and mature adipocytes were found to express Ang-1, Ang-2, and Tie-2, which decrease upon polyunsaturated fatty acid treatment. Furthermore, 3T3-L1 cells treated with recombinant Ang-1 or Ang-2 increased expression of the antiapoptotic gene Bcl-x and decreased expression of the proapoptotic gene Casp-8. Next, preadipocytes were treated with saturated fatty acids (SFAs) to induce cell stress. SFA-mediated splicing of X-box-binding protein-1 was reduced by co-treatment with Ang-1, and cell viability was improved in the presence of SFAs + Ang-1. Taken together, these results indicate that Ang-1 may protect preadipocytes from SFA-induced apoptosis and endoplasmic reticulum stress.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis , Tejido Adiposo/citología , Angiopoyetina 1/farmacología , Grasas de la Dieta/farmacología , Ácidos Grasos/farmacología , Neovascularización Fisiológica , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/fisiología , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/fisiología , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Angiopoyetina 2/farmacología , Animales , Apoptosis , Caspasa 8/metabolismo , Supervivencia Celular , Estrés del Retículo Endoplásmico , Células Endoteliales , Ácidos Grasos Insaturados/farmacología , Humanos , Macrófagos , Ratones , Ratones Endogámicos C57BL , Receptor TIE-2/metabolismo , Receptor TIE-2/farmacología , Proteína 1 de Unión a la X-Box/metabolismo , Proteína bcl-X/metabolismo
20.
Cells ; 9(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31936361

RESUMEN

Angiopoietin-1 (Ang-1) is an important proangiogenic factor also involved in the maintenance of endothelial-barrier integrity. The small GTPase Rap1 is involved in the regulation of adherens junctions through VE-cadherin-mediated adhesion, and in endothelial permeability. While many studies established that Rap1 activation is critical for endothelial cell-cell adhesions, its roles in the antipermeability effects of Ang-1 are ill-defined. Thus, we determined the contribution of Rap1 to Ang-1-stimulated angiogenic effects on endothelial cells (ECs). We found that Rap1 is activated following Ang-1 stimulation and is required for the antipermeability effects of Ang-1 on EC monolayers. Our results also revealed that Rap1 is necessary for EC sprouting stimulated by Ang-1 but had no significant effect on Ang-1-induced EC migration and adhesion. In contrast, downregulation of VE-cadherin markedly increased the adhesiveness of ECs to the substratum, which resulted in inhibition of Ang-1-stimulated migration. These results revealed that Rap1 is central to the effects of Ang-1 at intercellular junctions of ECs, whereas VE-cadherin is also involved in the adhesion of ECs to the extracellular matrix.


Asunto(s)
Angiopoyetina 1/farmacología , Aorta/fisiología , Adhesión Celular , Comunicación Celular , Endotelio Vascular/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Transducción de Señal , Proteínas de Unión al GTP rap1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...