Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Divers ; 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678137

RESUMO

Suppressing vascular endothelial growth factor (VEGF), its receptor (VEGFR2), and the VEGF/VEGFR2 signaling cascade system to inhibit angiogenesis has emerged as a possible cancer therapeutic target. The present work was designed to discover and evaluate bioactive phytochemicals from the Clerodendrum inerme (L.) Gaertn plant for their anti-angiogenic potential. Molecular docking of twenty-one phytochemicals against the VEGFR-2 (PDB ID: 3VHE) protein was performed, followed by ADMET profiling and molecular docking simulations. These investigations unveiled two hit compounds, cirsimaritin (- 12.29 kcal/mol) and salvigenin (- 12.14 kcal/mol), with the highest binding energy values when compared to the reference drug, Sorafenib (- 15.14 kcal/mol). Furthermore, only nine phytochemicals (cirsimaritin and salvigenin included) obeyed Lipinski's rule of five and passed ADMET filters. Molecular dynamics simulations run over 100 ns revealed that the protein-ligand complexes remained stable with minimal backbone fluctuations. The binding free energy values of cirsimaritin (- 52.35 kcal/mol) and salvigenin (- 55.89 kcal/mol), deciphered by MM-GBSA analyses, further corroborated the docking interactions. The HOMO-LUMO band energy gap (ΔE) was calculated using density-functional theory (DFT) and substantiated using density of state (DOS) spectra. The chemical reactivity analyses revealed that salvigenin exhibited the highest chemical softness value (6.384 eV), the lowest hardness value (0.07831 eV), and the lowest ΔE value (0.1566 eV), which implies salvigenin was less stable and chemically more reactive than cirsimaritin and sorafenib. These findings provide further evidence that cirsimaritin and salvigenin have the ability to prevent angiogenesis and the development of cancer. Nevertheless, more in vitro and in vivo confirmation is necessary.

2.
Biomater Adv ; 157: 213754, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38211507

RESUMO

Chronic wounds pose significant health concerns. Current treatment options include natural compounds like natural rubber latex (NRL) from Hevea brasiliensis. NRL, particularly the F1 protein fraction, has demonstrated bioactivity, biocompatibility, and angiogenic effects. So far, there is no study comparing F1 protein with total NRL serum, and the necessity of downstream processing remains unknown. Here, we evaluated the angiogenic potential of F1 protein compared to total NRL serum and the need for downstream processing. For that, ion exchange chromatography (DEAE-Sepharose), antioxidant activity, physicochemical characterization, cell culture in McCoy fibroblasts, and wound healing in Balb-C mice were performed. Also, the evaluation of histology and collagen content and the levels of inflammatory mediators were quantified. McCoy fibroblast cell assay showed that F1 protein (0.01 %) and total NRL serum (0.01 %) significantly increased cell proliferation by 47.1 ± 11.3 % and 25.5 ± 2.5 %, respectively. However, the AA of F1 protein (78.9 ± 0.8 %) did not show a significant difference compared to NRL serum (77.0 ± 1.1 %). F1 protein and NRL serum were more effective in wound management in rodents. Histopathological analysis confirmed accelerated healing and advanced tissue repair. Similarly, the F1 protein (0.01 %) increased collagen, showing that this fraction can stimulate the synthesis of collagen by fibroblastic cells. Regarding cytokines production (IL-10, TNF-α, IFN-γ), F1 protein and NRL serum did not exert an impact on the synthesis of these cytokines. Furthermore, we did not observe statistically significant changes in dosages of enzymes (MPO and EPO) among the groups. Nevertheless, Nitric Oxide dosage was reduced drastically when the F1 protein (0.01 %) protein was applied topically. These findings contribute to the understanding of F1 protein and NRL serum properties and provide insights into cost-effectiveness and practical applications in medicine and biotechnology. Therefore, further research is needed to assess the economic feasibility of downstream processing for NRL-based herbal medicine derived from Hevea brasiliensis.


Assuntos
Hevea , Borracha , Animais , Camundongos , Látex , Hevea/química , Cicatrização , Colágeno , Citocinas
3.
Nat Prod Res ; 38(4): 634-638, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-36799649

RESUMO

In connection to search for safe and alternative plant-based drugs, the wound healing mechanisms of an Indian ethnomedicine Couroupita guianensis fruit pulp was analyzed in this project work. Gas chromatography coupled with mass spectrometer (GC-MS) analysis revealed the existence of phytochemicals such as 2-furoic acid, 2,4-heptadienal, pyrazole and 8-hydroxyquinoline in the methanol extract. Methanol extract of C. guianensis exhibited remarkable radical scavenging activity against 2, 2-diphenyl-1-picrylhydrazyl (DPPH) (89.88%), superoxide (91.51%), hydrogen peroxide (24.25%) and hydroxyl radicals (73.62%). Further, it showed remarkable anti-inflammatory (24.09-62.16%) and anti-bacterial activity (zone of inhibition, ZOI: 13.00 mm, minimum inhibitory concentration, MIC: 6.25 mg/mL and minimum bactericidal concentration, MBC: 12.51 mg/mL) and also controlled the growth rate of methicillin resistant Staphylococcus aureus (MRSA) within 30 min of treatment. The angiogenic potential of C. guianensis was proved in chick chorioallantoic membrane (CAM) model and it does not exhibit any toxicity in peripheral blood monocyte cells (PBMC) model.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Extratos Vegetais , Humanos , Extratos Vegetais/química , Antibacterianos/química , Metanol/química , Profissionais de Medicina Tradicional , Antioxidantes/farmacologia , Frutas/química , Leucócitos Mononucleares , Cicatrização , Testes de Sensibilidade Microbiana , Medicina Tradicional
4.
Int J Mol Sci ; 24(19)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37834069

RESUMO

Three-dimensional (3D) culture systems have been widely used to promote the viability and metabolic activity of mesenchymal stem cells (MSCs). The aim of this study was to explore the synergistic benefits of using dual 3D MSC culture systems to promote vascular regeneration and enhance therapeutic potential. We used various experimental assays, including dual 3D cultures of human adipose MSCs (hASCs), quantitative reverse transcription polymerase chain reaction (qRT-PCR), in vitro cell migration, Matrigel tube network formation, Matrigel plug assay, therapeutic assays using an ischemic hind limb mouse model, and immunohistochemical analysis. Our qRT-PCR results revealed that fibroblast growth factor 2 (FGF-2), granulocyte chemotactic protein-2 (GCP-2), and vascular endothelial growth factor-A (VEGF-A) were highly upregulated in conventional 3D-cultured hASCs (ASC-3D) than in two-dimensional (2D)-cultured hASCs. Hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), and stromal-cell-derived factor-1 (SDF-1) showed higher expression levels in cytokine-cocktail-based, 3D-cultured hASCs (ASC-3Dc). A conditioned medium (CM) mixture of dual 3D ASCs (D-3D; ASC-3D + ASC-3Dc) resulted in higher migration and Matrigel tube formation than the CM of single 3D ASCs (S-3D; ASC-3D). Matrigel plugs containing D-3D contained more red blood cells than those containing S-3D. D-3D transplantation into ischemic mouse hind limbs prevented limb loss and augmented blood perfusion when compared to S-3D transplantation. Transplanted D-3D also revealed a high capillary density and angiogenic cytokine levels and transdifferentiated into endothelial-like cells in the hind limb muscle. These findings highlight the benefits of using the dual 3D culture system to optimize stem-cell-based therapeutic strategies, thereby advancing the therapeutic strategy for ischemic vascular disease and tissue regeneration.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Neovascularização Fisiológica/fisiologia , Células-Tronco Mesenquimais/metabolismo , Isquemia/terapia , Isquemia/metabolismo , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Tecido Adiposo/metabolismo
5.
Stem Cells Transl Med ; 12(6): 379-390, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37263619

RESUMO

Human multipotent mesenchymal stromal/stem cells (MSCs) have been utilized in cell therapy for various diseases and their clinical applications are expected to increase in the future. However, the variation in MSC-based product quality due to the MSC heterogeneity has resulted in significant constraints in the clinical utility of MSCs. Therefore, we hypothesized that it might be important to identify and ensure/enrich suitable cell subpopulations for therapies using MSC-based products. In this study, we aimed to identify functional cell subpopulations to predict the efficacy of angiogenic therapy using bone marrow-derived MSCs (BM-MSCs). To assess its angiogenic potency, we observed various levels of vascular endothelial growth factor (VEGF) secretion among 11 donor-derived BM-MSC lines under in vitro ischemic culture conditions. Next, by clarifying the heterogeneity of BM-MSCs using single-cell RNA-sequencing analysis, we identified a functional cell subpopulation that contributed to the overall VEGF production in BM-MSC lines under ischemic conditions. We also found that leucine-rich repeat-containing 75A (LRRC75A) was more highly expressed in this cell subpopulation than in the others. Importantly, knockdown of LRRC75A using small interfering RNA resulted in significant inhibition of VEGF secretion in ischemic BM-MSCs, indicating that LRRC75A regulates VEGF secretion under ischemic conditions. Therefore, LRRC75A may be a useful biomarker to identify cell subpopulations that contribute to the angiogenic effects of BM-MSCs. Our work provides evidence that a strategy based on single-cell transcriptome profiles is effective for identifying functional cell subpopulations in heterogeneous MSC-based products.


Assuntos
Células-Tronco Mesenquimais , Fator A de Crescimento do Endotélio Vascular , Humanos , Células da Medula Óssea , Diferenciação Celular , Proliferação de Células , Isquemia/genética , Isquemia/terapia , Isquemia/metabolismo , Análise da Expressão Gênica de Célula Única , Células-Tronco , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Fatores de Crescimento do Endotélio Vascular/farmacologia
6.
Toxics ; 11(4)2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37112570

RESUMO

A review of recent literature suggests that bismuth oxide (Bi2O3, referred to as B in this article) nanoparticles (NPs) elicit an appreciable response only after a concentration above 40-50 µg/mL in different cells all having an epithelial origin, to the best of our knowledge. Here, we report the toxicological profile of Bi2O3 NPs (or BNPs) (71 ± 20 nm) in a human endothelial cell (HUVE cell line) in which BNPs exerted much steeper cytotoxicity. In contrast to a high concentration of BNPs (40-50 µg/mL) required to stimulate an appreciable toxicity in epithelial cells, BNPs induced 50% cytotoxicity in HUVE cells at a very low concentration (6.7 µg/mL) when treated for 24 h. BNPs induced reactive oxygen species (ROS), lipid peroxidation (LPO), and depletion of the intracellular antioxidant glutathione (GSH). BNPs also induced nitric oxide (NO,) which can result in the formation of more harmful species in a fast reaction that occurs with superoxide (O2•-). Exogenously applied antioxidants revealed that NAC (intracellular GSH precursor) was more effective than Tiron (a preferential scavenger of mitochondrial O2•-) in preventing the toxicity, indicating ROS production is extra-mitochondrial. Mitochondrial membrane potential (MMP) loss mediated by BNPs was significantly less than that of exogenously applied oxidant H2O2, and MMP loss was not as intensely reduced by either of the antioxidants (NAC and Tiron), again suggesting BNP-mediated toxicity in HUVE cells is extra-mitochondrial. When we compared the inhibitory capacities of the two antioxidants on different parameters of this study, ROS, LPO, and GSH were among the strongly inhibited biomarkers, whereas MMP and NO were the least inhibited group. This study warrants further research regarding BNPs, which may have promising potential in cancer therapy, especially via angiogenesis modulation.

7.
Pharmaceutics ; 15(3)2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36986815

RESUMO

The short shelf life of platelet concentrates (PC) of up to 5-7 days leads to higher wastage due to expiry. To address this massive financial burden on the healthcare system, alternative applications for expired PC have emerged in recent years. Engineered nanocarriers functionalized with platelet membranes have shown excellent targeting abilities for tumor cells owing to their platelet membrane proteins. Nevertheless, synthetic drug delivery strategies have significant drawbacks that platelet-derived extracellular vesicles (pEV) can overcome. We investigated, for the first time, the use of pEV as a carrier of the anti-breast cancer drug paclitaxel, considering it as an appealing alternative to improve the therapeutic potential of expired PC. The pEV released during PC storage showed a typical EV size distribution profile (100-300 nm) with a cup-shaped morphology. Paclitaxel-loaded pEV showed significant anti-cancer effects in vitro, as demonstrated by their anti-migratory (>30%), anti-angiogenic (>30%), and anti-invasive (>70%) properties in distinct cells found in the breast tumor microenvironment. We provide evidence for a novel application for expired PC by suggesting that the field of tumor treatment research may be broadened by the use of natural carriers.

8.
Cell Mol Life Sci ; 79(5): 259, 2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35474498

RESUMO

Delayed wound healing and chronic skin lesions represent a major health problem. Over the past years, growth factors mediated by platelet-rich plasma (PRP) and cell-based therapies were developed as effective and affordable treatment able to improve wound healing capacity. We have advanced existing concepts to develop a highly efficient high-throughput protocol with proven application for the isolation of PRP and pro-angiogenic cells (AngioPRP). This protocol outlines the effectiveness of AngioPRP in promoting the critical healing process including wound closure, re-epithelialization, granulation tissue growth, and blood vessel regeneration. We coupled this effect with normalization of mechanical properties of rescued mouse wounds, which is sustained by a correct arrangement of elastin and collagen fibers. Proteomic analysis of treated wounds demonstrated a fingerprint of AngioPRP based on the up-regulation of detoxification pathway of glutathione metabolism, correlated to a decrease in inflammatory response. Overall, these results have enabled us to provide a framework for how AngioPRP supports wound healing, opening avenues for further clinical advances.


Assuntos
Plaquetas , Plasma Rico em Plaquetas , Animais , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Plasma Rico em Plaquetas/metabolismo , Proteômica , Cicatrização/fisiologia
9.
J Gene Med ; 23(7): e3342, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33870576

RESUMO

BACKGROUND: Mesenchymal stromal cells (MSC) have been exploited for the treatment of ischemic diseases given their angiogenic potential. Despite bone marrow (BM) being the most studied tissue source, cells with similar intrinsic properties can be isolated from adipose tissue (AT) and umbilical cord matrix (UCM). The present study aims to compare the angiogenic potential of MSC obtained from BM, AT and UCM that were genetically modified with vascular endothelial growth factor (VEGF)-encoding minicircle (MC) vectors. The overexpression of VEGF combined with the intrinsic properties of MSC could represent a promising strategy towards angiogenic therapies. METHODS: We established a microporation-based protocol to transfect human MSC using VEGF-encoding MC (MC-VEGF). VEGF production levels were measured by an enzyme-linked immunosorbent assay and a quantitative polymerase chain reaction. The in vitro angiogenic potential of transfected cells was quantified using cell tube formation and migration functional studies. RESULTS: MSC isolated from BM, AT or UCM showed similar levels of VEGF secretion after transfection with MC-VEGF. Those values were significantly higher when compared to non-transfected cells, indicating an effective enhancement of VEGF production. Transfected cells displayed higher in vitro angiogenic potential than non-transfected controls, as demonstrated by functional in vitro assays. No significant differences were observed among cells from different sources. CONCLUSIONS: Minicircles can be successfully used to transiently overexpress VEGF in human MSC, regardless of the cell tissue source, representing an important advantage in a clinical context (i.e., angiogenic therapy) because a standard protocol might be applied to MSC of different tissue sources, which can be differentially selected according to the application (e.g., autologous versus allogeneic settings).


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Tecido Adiposo/metabolismo , Medula Óssea/metabolismo , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Expressão Gênica , Humanos , Neovascularização Fisiológica , Transfecção/métodos , Cordão Umbilical/metabolismo
10.
J Cell Physiol ; 235(10): 7224-7238, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32037550

RESUMO

Mesenchymal stromal cells (MSCs) have been widely exploited for the treatment of several conditions due to their intrinsic regenerative and immunomodulatory properties. MSC have demonstrated to be particularly relevant for the treatment of ischemic diseases, where MSC-based therapies can stimulate angiogenesis and induce tissue regeneration. Regardless of the condition targeted, recent analyses of MSC-based clinical trials have demonstrated limited benefits indicating a need to improve the efficacy of this cell product. Preconditioning MSC ex vivo through microenvironment modulation was found to improve MSC survival rate and thus prolong their therapeutic effect. This workstudy aims at enhancing the in vitro angiogenic capacity of a potential MSC-based medicinal product by comparing different sources of MSC and culture conditions. MSC from three different sources (bone marrow [BM], adipose tissue [AT], and umbilical cord matrix [UCM]) were cultured with xenogeneic-/serum-free culture medium under static conditions and their angiogenic potential was studied. Results indicated a higher in vitro angiogenic capacity of UCM MSC, compared with cells derived from BM and AT. Physicochemical preconditioning of UCM MSC through a microcarrier-based culture platform and low oxygen concentration (2% O2 , compared with atmospheric air) increased the in vitro angiogenic potential of the cultured cells. Envisaging the clinical manufacturing of an allogeneic, off-the-shelf MSC-based product, preconditioned UCM MSC maintain the angiogenic gene expression profile upon cryopreservation and delivery processes in the conditions of our study. These results are expected to contribute to the development of MSC-based therapies in the context of angiogenesis.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Microambiente Celular/fisiologia , Criopreservação , Meios de Cultura Livres de Soro , Humanos , Imunofenotipagem , Técnicas In Vitro , Neovascularização Fisiológica/genética , Oxigênio , Transcriptoma , Cordão Umbilical/citologia
11.
Int J Biol Macromol ; 143: 763-774, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31626822

RESUMO

However, labelling of stem cells using nanoparticles (NPs) for tracking purpose has been intensively investigated, the biosafety of these materials needs more clarification. Herein, different forms of iron oxide Fe2O3, Fe3O4, and CoxNi1-x Fe2O4 NPs either uncoated or starch-coated (ST-coated) were prepared. We successfully labelled adipose-derived stem cells (ASCs) using these NPs with the aid of lipofectamine as a transfection agent (TA). We then evaluated the effect of these NPs on stem cell proliferation, viability, migration and angiogenesis. Results showed that ASCs labelled with Fe2O3, Fe3O4, ST-Fe2O3 and ST-Fe3O4 did not show any significant difference in proliferation compared to that of TA-treated cells. Moreover, they have shown a protective effect against apoptosis. Conversely, CoxNi1-x Fe2O4 NPs caused a significant decrease in cell proliferation. Compared to that of the TA-treated cells, the migration capacity of cells labelled with Fe2O3, Fe3O4 and CoxNi1-xFe2O4 was significantly compromised. Interestingly, the ST-coated composites reversed this effect. Among the groups treated with different NPs, the angiogenic potential of the ASCs was most robust in the ST-Fe2O3-treated group. In conclusion, labelling ASCs with ST-Fe2O3 NPs enhanced cell migration and angiogenic potential and conferred higher resistance to apoptosis than labelling the cells with the other tested NPs.


Assuntos
Rastreamento de Células , Nanopartículas de Magnetita/química , Amido/farmacologia , Células-Tronco/citologia , Apoptose/efeitos dos fármacos , Capilares/efeitos dos fármacos , Capilares/crescimento & desenvolvimento , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/ultraestrutura , Neovascularização Fisiológica/efeitos dos fármacos , Espectroscopia de Infravermelho com Transformada de Fourier , Células-Tronco/efeitos dos fármacos , Difração de Raios X
12.
Adv Med Sci ; 64(2): 216-223, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30818220

RESUMO

PURPOSE: The biggest problem with the occurrence of breast cancer is late diagnosis, which is associated with high mortality rates. The aim of the study was to appraise the number of circulating endothelial precursors and the concentration of vascular endothelial growth factor A (VEGF-A) and the soluble forms of its receptors, sVEGFR1 and sVEGFR2, in breast cancer patients with respect to clinicopathological features. MATERIAL AND METHODS: The study involved 85 women of Caucasian ethnicity aged 45-66 with primary breast cancer without distant metastases (M0). Inclusion criteria were as follows: histopathological examination confirming the diagnosis of primary breast cancer, without previous radiotherapy and chemotherapy. Immunohistochemistry evaluation of oestrogen and progesterone receptors, human epidermal growth factor receptor 2, Ki67 expression was made in all cases. In the EDTA-plasma, the concentrations of VEGF-A and its soluble receptors, sVEGFR1 and sVEGFR2, were measured applying immunoassay techniques. Circulating endothelial progenitor cells (EPCs) were identified with the immune-phenotype CD45-, CD34+, CD133+, CD31+ using flow cytometry. RESULTS: Older women with breast cancer had significantly higher concentrations of VEGF-A as well as sVEGFR2 compared with their younger counterparts. A significantly higher concentration of the soluble form of VEGF receptor type 1 in patients with T1 breast cancer in relation to T2 cases was noted. Also, negative correlations between circulating EPCs and histological grading as well as a soluble form of VEGFR2 with histological grading of breast cancer according to the Elston-Ellis classification were observed. CONCLUSIONS: Anti-angiogenic potential is divergent in relation to the clinicopathological determinants.


Assuntos
Neoplasias da Mama/patologia , Idoso , Pressão Sanguínea/fisiologia , Neoplasias da Mama/metabolismo , Células Progenitoras Endoteliais/citologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
13.
Int J Mol Sci ; 19(11)2018 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-30380605

RESUMO

Adult human multipotent neural cell (ahMNC) is a candidate for regeneration therapy for neurodegenerative diseases. Here, we developed a primary clump culture method for ahMNCs to increase the efficiency of isolation and in vitro expansion. The same amount of human temporal lobe (1 g) was partially digested and then filtered through strainers with various pore sizes, resulting in four types of clumps: Clump I > 100 µm, 70 µm < Clump II < 100 µm, 40 µm < Clump III < 70 µm, and Clump IV < 40 µm. At 3 and 6 days after culture, Clump II showed significantly higher number of colonies than the other Clumps. Moreover, ahMNCs derived from Clump II (ahMNCs-Clump II) showed stable proliferation, and shortened the time to first passage from 19 to 15 days, and the time to 1 × 108 cells from 42 to 34 days compared with the previous single-cell method. ahMNCs-Clump II had neural differentiation and pro-angiogenic potentials, which are the characteristics of ahMNCs. In conclusion, the novel clump culture method for ahMNCs has significantly higher efficiency than previous techniques. Considering the small amount of available human brain tissue, the clump culture method would promote further clinical applications of ahMNCs.


Assuntos
Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Multipotentes/citologia , Células-Tronco Neurais/citologia , Adulto , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Fisiológica
14.
Biomed Pharmacother ; 108: 1328-1337, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30372835

RESUMO

Metformin is commonly prescribed as a hypoglycemic agent following the onset of type 2 diabetes mellitus. This study aimed to investigate pro- and/or anti-angiogenic effects of Metformin on human bone marrow mesenchymal stem cells. Cells were incubated with different doses of Metformin including 0.5, 1, 10, 50, 100, 200 and 500 µM for 14 days. Cell viability and total fatty acids profile were examined by MTT and gas chromatography methods. Differentiation of cells to endothelial lineage was studied by monitoring the expression of VEGFR-2 and Tie-2 receptors and VE-cadherin via real-time PCR and western blotting. Angiogenic potential and migration of cells were assessed by tubulogenesis and Transwell migration assays. PCR array was performed to analyze mTOR signaling. CD133+ and VEGFR-2+ cells were detected in blood samples of non-diabetic control, diabetic subjects and diabetics received Metformin. Metformin dose-dependently reduced cell survival. Decreased content of palmitate and oleate coincided increased level of stearate, palmitoleate, and linoleate (p < 0.05). Metformin decreased the angiogenic potential of cells by decreasing VEGFR-2 and Tie-2 expression (p < 0.05). The protein level of VE-cadherin decreased in cells received Metformin. Compared to the control, Metformin blunted the expression of VEGF subtypes and directed cells to energy status by induction of PRKAA1, PRKAB2, and PRKAG1 genes (p < 0.05). Non-significant differences were observed regarding the number of CD133 and VEGFR-2 cells in blood samples (p > 0.05). These data support a notion that Metformin could blunt the angiogenic behavior of human mesenchymal stem cells by modulating mTOR signaling pathway.


Assuntos
Células-Tronco Mesenquimais/efeitos dos fármacos , Metformina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Antígeno AC133/sangue , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácidos Graxos/análise , Humanos , Imunofenotipagem , Células-Tronco Mesenquimais/citologia , Serina-Treonina Quinases TOR/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue
15.
Neurourol Urodyn ; 37(2): 716-725, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29439287

RESUMO

AIMS: Pelvic organ prolapse and stress urinary incontinence affect 40-50% of postmenopausal women worldwide. Polypropylene meshes have been extensively used for the surgical intervention of these disorders; however, these meshes can lead to severe complications in some patients. The need for synthetic materials more suited for use in pelvic floor repair is widely accepted. This study aims to develop an electrospun 17-ß-estradiol releasing polyurethane (PU) scaffold that not only provides the appropriate mechanical support but can also stimulate new extracellular matrix (ECM) production and angiogenesis. METHODS: PU scaffolds with and without 17-ß-estradiol (25 and 50 mg/g) were prepared by blend electrospinning. Mechanical properties of scaffolds were assessed by uniaxial cyclic and non-cyclic testing. The viability and ECM production of human adipose derived mesenchymal stem cells (hADMSCs) cultured on 17-ß-estradiol releasing PU scaffolds was evaluated. Angiogenic potential of estradiol releasing scaffolds was demonstrated by using an ex ovo chick chorioallantoic membrane (CAM) assay. RESULTS: The inclusion of estradiol in PU scaffolds did not change the ultrastructure but it significantly increased the ultimate tensile strength of scaffolds. hADMSCs on estradiol-releasing PU scaffolds showed more ECM production. The CAM assay revealed a significantly higher angiogenic potential of estradiol-releasing PU scaffolds with an additive effect seen when hADMSCs cultured on estradiol scaffolds. Histological examination of CAM tissue sections showed extensive cellular infiltration and a good tissue integration for all constructed scaffolds. CONCLUSIONS: This study shows the angiogenic potential of estradiol-releasing PU scaffolds with appropriate strength and elasticity desirable to support the pelvic floor.


Assuntos
Matriz Extracelular/fisiologia , Neovascularização Fisiológica/fisiologia , Diafragma da Pelve/cirurgia , Prolapso de Órgão Pélvico/cirurgia , Alicerces Teciduais , Incontinência Urinária por Estresse/cirurgia , Estradiol/uso terapêutico , Feminino , Humanos , Teste de Materiais , Células-Tronco Mesenquimais , Poliuretanos , Engenharia Tecidual , Resultado do Tratamento
16.
Colloids Surf B Biointerfaces ; 159: 493-500, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28841499

RESUMO

Recent evidence demonstrates that vessel involvement is crucial in various bone remodeling situations, indicating that blood vessel formation within or surrounding the implant is essential for establishment of rigid implant fixation. In this work, the ability of the silicon-doped porous TiO2 coatings fabricated via plasma electrolyte oxidation method (PEO) to enhance the angiogenic potential of human umbilical vein endothelial cells (HUVECs) were investigated. The cellular responses of HUVECs on the silicon-doped porous TiO2 coatings were studied through cell proliferation, vascular endothelial growth factor (VEGF) secretion, and angiogenic-associated gene (VEGF, HIF-1α and HGF) expression analysis. The results show that small amount of silicon significantly enhanced angiogenic activity of HUVECs, while larger amount of silicon appears excessive. Hence, the silicon-doped TiO2 coating offers a potential solution to improve bone vascularization to achieve efficient osseointegration and restoration of function after implantation.


Assuntos
Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Titânio/química , Titânio/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Porosidade , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Cell Tissue Bank ; 18(1): 27-43, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28004288

RESUMO

INTRODUCTION: Transplantation of a cell-seeded graft may improve wound healing after radiotherapy. However, the survival of the seeded cells depends on a rapid vascularization of the graft. Co-culturing of adult stem cells may be a promising strategy to accelerate the vessel formation inside the graft. Thus, we compared the in vivo angiogenic potency of mesenchymal stem cells (MSC) and endothelial progenitor cells (EPC) using dorsal skinfold chambers and intravital microscopy. MATERIALS AND METHODS: Cells were isolated from rat bone marrow and adipose tissue and characterized by immunostaining and flow cytometry. Forty-eight rats received a dorsal skinfold chamber and were divided into 2 main groups, irradiated and non-irradiated. Each of these 2 groups were further subdivided into 4 groups: unseeded matrices, matrices + fibroblasts + pericytes, matrices + fibroblasts + pericytes + MSCs and matrices + fibroblasts + pericytes + EPCs. Vessel densities were quantified semi-automatically using FIJI. RESULTS: Fibroblasts + pericytes - seeded matrices showed a significantly higher vascular density in all groups with an exception of non-irradiated rats at day 12 compared to unseeded matrices. Co-seeding of MSCs increased vessel densities in both, irradiated and non-irradiated groups. Co-seeding with EPCs did not result in an increase of vascularization in none of the groups. DISCUSSION: We demonstrated that the pre-radiation treatment led to a significant decreased vascularization of the implanted grafts. The augmentation of the matrices with fibroblasts and pericytes in co-culture increased the vascularization compared to the non-seeded matrices. A further significant enhancement of vessel ingrowth into the matrices could be achieved by the co-seeding with MSCs in both, irradiated and non-irradiated groups.


Assuntos
Derme Acelular , Células Progenitoras Endoteliais/citologia , Fibroblastos/citologia , Microscopia Intravital , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Pericitos/citologia , Derme Acelular/metabolismo , Derme Acelular/efeitos da radiação , Animais , Células Cultivadas , Técnicas de Cocultura , Células Progenitoras Endoteliais/efeitos da radiação , Fibroblastos/efeitos da radiação , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos da radiação , Pericitos/efeitos da radiação , Ratos , Ratos Endogâmicos F344 , Engenharia Tecidual , Cicatrização/efeitos da radiação
18.
J Endod ; 43(2): 297-305, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28027822

RESUMO

INTRODUCTION: To investigate the roles of semaphorin 4D (Sema4D)/plexin-B1 signaling on the angiogenic potential and osteo-/odontogenic differentiation of human dental pulp stem cells (DPSCs) and to uncover the corresponding molecular mechanisms. METHODS: DPSCs were treated with Sema4D (10 µg/mL) for different time durations. Osteo-/odontogenic differentiation was assessed by quantifying alkaline phosphatase activity, mineralized nodule formation, and osteo-/odontogenic gene (ALP, Col1A1, BSP, RUNX2, and DSPP) and protein (Col1A1 and DSPP) expression. Involvement of the Sema4D/plexin-B1 signaling pathway was analyzed by Western blot analysis. Additionally, angiogenic gene and protein expression was assessed by reverse-transcription polymerase chain reaction and enzyme-linked immunosorbent assay. In vitro endothelial tube formation assay on Matrigel (BD Biosciences, San Jose, CA) was performed to evaluate the angiogenic inductive potential of the Sema4D-treated DPSCs conditioned medium. Results were analyzed using 1-way analysis of variance and the Student t test. RESULTS: Sema4D significantly inhibited ALP activity and mineralized nodule formation of DPSCs. Furthermore, Sema4D-treated DPSCs displayed marked down-regulation in the expression of osteo-/odontogenic genes (ALP, Col1A1, BSP, RUNX2, and DSPP) as well as proteins (Col1A1 and DSPP). Elevated levels of plexin-B1 and downstream RhoA protein expression together with phosphorylated plexin-B1 confirmed the involvement of Sema4D/plexin-B1 signaling. Protein expression of ErbB2 was up-regulated, and Met was slightly down-regulated. Furthermore, Sema4D-treated DPSCs exhibited enhanced expression of vascular endothelial growth factor at both the messenger RNA and protein level. Accordingly, the conditioned medium of Sema4D-treated DPSCs promoted the formation of vessel-like structures as shown by the Matrigel assay. CONCLUSIONS: Sema4D markedly enhances the angiogenic potential but suppresses osteo-/odontogenic differentiation of DPSCs. Sema4D/plexin-B signaling was activated via the RhoA-mediated pathway.


Assuntos
Antígenos CD/farmacologia , Polpa Dentária/efeitos dos fármacos , Semaforinas/farmacologia , Células-Tronco/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Polpa Dentária/citologia , Polpa Dentária/crescimento & desenvolvimento , Ensaio de Imunoadsorção Enzimática , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Células-Tronco/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Stem Cells Transl Med ; 4(10): 1109-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26273064

RESUMO

UNLABELLED: Mesenchymal stem cells (MSCs) of placental origin have become increasingly translational owing to their abundance and accessibility. MSCs of different origin share several features but also present biological differences that might point to distinct clinical properties. Hence, mixing fetal and maternal cells from the same placenta can lead to contradicting results. We analyzed the biological characteristics of haploidentical MSCs isolated from fetal sources, including the umbilical cord (UC-MSCs) and chorion (Ch-MSCs), compared with maternal decidua MSCs (Dc-MSCs). All MSCs were analyzed for general stem cell properties. In addition, immunosuppressive capacity was assessed by the inhibition of T-cell proliferation, and angiogenic potential was evaluated in a Matrigel transplantation assay. The comparison between haploidentical MSCs displayed several distinct features, including (a) marked differences in the expression of CD56, (b) a higher proliferative capacity for Dc-MSCs and UC-MSCs than for Ch-MSCs, (c) a diversity of mesodermal differentiation potential in favor of fetal MSCs, (d) a higher capacity for Ch-MSCs to inhibit T-cell proliferation, and (e) superior angiogenic potential of Ch-MSCs evidenced by a higher capability to form tubular vessel-like structures and an enhanced release of hepatocyte growth factor and vascular endothelial growth factor under hypoxic conditions. Our results suggest that assessing the prevalence of fetomaternal contamination within placental MSCs is necessary to increase robustness and limit side effects in their clinical use. Finally, our work presents evidence positioning fetoplacental cells and notably Ch-MSCs in the forefront of the quest for cell types that are superior for applications in regenerative medicine. SIGNIFICANCE: This study analyzed the biological characteristics of mesenchymal stem cells (MSCs) isolated from fetal and maternal placental origins. The findings can be summarized as follows: (a) important differences were found in the expression of CD56, (b) a different mesodermal differentiation potential was found in favor of fetal MSCs, (c) a higher immunosuppressive capacity for chorion MSCs was noted, and (d) superior angiogenic potential of Ch-MSCs was observed. These results suggest that assessing the prevalence of fetomaternal contamination within placental MSCs is necessary to increase robustness and limit side effects in their clinical use. The evidence should allow clinicians to view fetoplacental cells, notably Ch-MSCs, favorably as candidates for use in regenerative medicine.


Assuntos
Córion/citologia , Decídua/citologia , Células-Tronco Mesenquimais/citologia , Antígeno CD56/biossíntese , Antígeno CD56/genética , Diferenciação Celular , Células Cultivadas , Feminino , Sangue Fetal/citologia , Feto/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Terapia de Imunossupressão , Recém-Nascido , Masculino , Neovascularização Fisiológica , Especificidade de Órgãos , Medicina Regenerativa , Linfócitos T/imunologia
20.
Growth Factors ; 32(3-4): 83-116, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24712317

RESUMO

Growth factors and cytokines released at sites of injury and inflammation play an important role in stimulating endothelial progenitor cell (EPC) migration to these sites. A comparative analysis of the literature shows under neutral in vitro conditions (pH 7.4), several growth factors and cytokines influenced favorably indices of EPC angiogenic function. They included SDF-1, VEGF, PlGF, FGF-2, NGF and IL-1ß. Others, e.g. TNF-α, have an unfavorable influence. SDF-1 and VEGF in combination increased chemotactic cell migration and reduced apoptosis caused by serum starvation. Under acidic conditions (pH 6.5), the biological activity of certain growth factors may be impaired, although TPO, SCF and IL-3 were each able to rescue EPCs from acidic exposure apoptosis, a combination of these three factors stimulated cell proliferation and prevented apoptosis. Possible combinations of growth factors and cytokines together with EPC transplantation may provide for a greater extent of vessel repair and new vessel formation.


Assuntos
Citocinas/metabolismo , Células Progenitoras Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Fisiológica , Movimento Celular , Proliferação de Células , Citocinas/genética , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/fisiologia , Humanos , Técnicas In Vitro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA