Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Cancer ; 24(1): 909, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075471

RESUMO

BACKGROUND: Advancements in the diagnosis, treatment, and surveillance of castration-resistant prostate cancer (CRPC) have progressed considerably, but a new biomarker that combines existing clinical and pathological data could be useful for a more precise diagnosis and prognosis. Some investigations have found that extracellular vesicle (EV)-derived miRNAs play crucial roles in various types of malignant tumors. The objective of this study was to explore EV miRNA and identify its biologic function as a biomarker for the diagnosis and prognosis of CRPC. METHODS: Plasma samples were collected from five healthy donors (Control, CT) and 17 CRPC patients, categorizing into two groups based on their endocrine treatment response: partial response (PR; n = 10) and progressive disease (PD; n = 7). Candidate extracellular vesicle (EV) miRNAs were identified using miRNA microarray and RT-qPCR. The biological functions of the selected miRNAs were evaluated using the MTT assay, wound healing assay, trans-well assay, and RNA sequencing in CRPC cells after transient miRNA expression. RESULTS: Microarray analysis revealed a significant downregulation of EV-miR-6880-5p in the PD samples compared to both CT and PR samples (p < 0.01). The expression of EV-miR-6880-5p in CRPC patients was decreased compared with that CT group (p = 0.0336) using RT-qPCR. In the PR group, EV-miR-6880-5p was increased at follow-up compared with the baseline (p = 0.2803), while in the PD group, it decreased at follow-up compared with the baseline samples (p = 0.4356). Furthermore, overexpression of miR-6880-5p hampered cell proliferation, migration, and invasion, downregulated pathways associated with tumor progression, and simultaneously upregulated pathways associated with cell growth and apoptosis in CRPC cells. CONCLUSIONS: EV-miR-6880-5p shows promise as a prognostic biomarker in patients with CRPC. Further, prospective validations are necessary to evaluate the potential of these candidate miRNAs.


Assuntos
Biomarcadores Tumorais , Vesículas Extracelulares , Regulação Neoplásica da Expressão Gênica , MicroRNAs , Neoplasias de Próstata Resistentes à Castração , Humanos , Masculino , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/sangue , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , MicroRNAs/sangue , MicroRNAs/genética , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Prognóstico , Idoso , Pessoa de Meia-Idade , Linhagem Celular Tumoral , Proliferação de Células , Perfilação da Expressão Gênica/métodos , Movimento Celular/genética
2.
Mol Med ; 26(1): 15, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-32005100

RESUMO

BACKGROUND: Sca-1+ cardiac stem cells and their limited proliferative potential were major limiting factors for use in various studies. METHODS: Therefore, the effects of sphere genetically engineered cardiac stem cells (S-GECS) inserted with telomerase reverse transcriptase (TERT) were investigated to examine cardiomyocyte survival under hypoxic conditions. GECS was obtained from hTERT-immortalized Sca-1+ cardiac stem cell (CSC) lines, and S-GECS were generated using poly-HEMA. RESULTS: The optimal conditions for S-GECS was determined to be 1052 GECS cells/mm2 and a 48 h culture period to produce spheroids. Compared to adherent-GECS (A-GECS) and S-GECS showed significantly higher mRNA expression of SDF-1α and CXCR4. S-GECS conditioned medium (CM) significantly reduced the proportion of early and late apoptotic cardiomyoblasts during CoCl2-induced hypoxic injury; however, gene silencing via CXCR4 siRNA deteriorated the protective effects of S-GECS against hypoxic injury. As downstream pathways of SDF-1α/CXCR4, the Erk and Akt signaling pathways were stimulated in the presence of S-GECS CM. S-GECS transplantation into a rat acute myocardial infarction model improved cardiac function and reduced the fibrotic area. These cardioprotective effects were confirmed to be related with the SDF-1α/CXCR4 pathway. CONCLUSIONS: Our findings suggest that paracrine factors secreted from transplanted cells may protect host cardiomyoblasts in the infarcted myocardium, contributing to beneficial left ventricle (LV) remodeling after acute myocardial infarction (AMI).


Assuntos
Ataxina-1/metabolismo , Miócitos Cardíacos/citologia , Esferoides Celulares/citologia , Células-Tronco/citologia , Telomerase/genética , Animais , Ataxina-1/genética , Adesão Celular , Técnicas de Cultura de Células , Hipóxia Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Quimiocina CXCL12/genética , Cobalto/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Engenharia Genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Comunicação Parácrina , Regiões Promotoras Genéticas , Ratos , Receptores CXCR4/genética , Esferoides Celulares/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
3.
Soft Matter ; 16(2): 428-434, 2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31799582

RESUMO

Maturation and synchronisation of heart cells, including cardiomyocytes and fibroblasts, are essential to develop functional biomimetic cardiac tissues for regenerative medicine and drug discovery. Synchronisation of cells in the biomimetic cardiac tissue requires the structural integrity and functional maturation of cardiomyocytes with other cell types. However, it is challenging to synchronise the beating of macroscale cardiac tissues and induce maturation of cardiomyocytes derived from stem cells. Here, we developed a simple assembly technology to modulate cell-cell interactions by combining layer-by-layer (LBL) deposition and centrifugation of cells with collagen type I to control cell-cell interactions for the preparation of cardiac macro tissues (CMTs). We found that maturation of cardiomyocytes in CMTs was largely enhanced by growth factors FGF-4 and ascorbic acid, but synchronisation of cardiac beating required LBL deposition of cardiomyocytes and cardiac fibroblasts in addition to the growth factors during the maturation process. Our findings have important implications because incorporation of cardiac fibroblasts into the cardiomyocyte layer is a prerequisite for synchronised beating of macroscale cardiac tissues in addition to growth factors to facilitate maturation of stem cell-derived cardiomyocytes.


Assuntos
Fibroblastos/citologia , Miócitos Cardíacos/citologia , Materiais Biomiméticos/química , Materiais Biomiméticos/metabolismo , Biomimética , Bioimpressão , Comunicação Celular , Colágeno Tipo I/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator 4 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Humanos , Miócitos Cardíacos/metabolismo
4.
Int J Mol Sci ; 21(6)2020 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-32245208

RESUMO

Thymosin ß4 (Tß4) is a G-actin sequestering protein that contributes to diverse cellular activities, such as migration and angiogenesis. In this study, the beneficial effects of combined cell therapy with Tß4 and human adipose-derived stem cells (hASCs) in a mouse ischemic hindlimb model were investigated. We observed that exogenous treatment with Tß4 enhanced endogenous TMSB4X mRNA expression and promoted morphological changes (increased cell length) in hASCs. Interestingly, Tß4 induced the active state of hASCs by up-regulating intracellular signaling pathways including the PI3K/AKT/mTOR and MAPK/ERK pathways. Treatment with Tß4 significantly increased cell migration and sprouting from microbeads. Moreover, additional treatment with Tß4 promoted the endothelial differentiation potential of hASCs by up-regulating various angiogenic genes. To evaluate the in vivo effects of the Tß4-hASCs combination on vessel recruitment, dorsal window chambers were transplanted, and the co-treated mice were found to have a significantly increased number of microvessel branches. Transplantation of hASCs in combination with Tß4 was found to improve blood flow and attenuate limb or foot loss post-ischemia compared to transplantation with hASCs alone. Taken together, the therapeutic application of hASCs combined with Tß4 could be effective in enhancing endothelial differentiation and vascularization for treating hindlimb ischemia.


Assuntos
Membro Posterior/metabolismo , Isquemia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Timosina/metabolismo , Timosina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Transplante de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/genética , Isquemia/terapia , Sistema de Sinalização das MAP Quinases/genética , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Neovascularização Fisiológica/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Timosina/genética , Timosina/uso terapêutico , Cicatrização/genética
5.
Mol Med ; 25(1): 33, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31307370

RESUMO

BACKGROUND: The beneficial effects of angiotensin II type 1 receptor blockers (ARBs) on atherosclerosis have been demonstrated in numerous studies. We investigated the effects of fimasartan on reducing neointimal formation and systemic inflammation after carotid artery (CA) injury in Apolipoprotein E knockout (ApoE KO) mice. METHODS: ApoE KO mice were randomly allocated to Group I (without CA injury), Group II (without CA injury + Fimasartan), Group III (CA injury), and Group IV (CA injury + Fimasartan). Fimasartan was orally administered everyday starting 3 days before iatrogenic left CA injury. RESULTS: At 28 days, neointimal hyperplasia and the inflammatory cytokines including TNFα, IL-6, ICAM, and MMP-9 in the peripheral blood were significantly reduced in Groups II and IV compared to Groups I and III, respectively. All fimasartan-administered groups revealed significant increases of CD4+CD25+Foxp3+ regulatory T (Treg) cells with increased plasma levels of IL-10 and TGFß. In addition, increased CD8+ T cells by fimasartan were correlated with reduced smooth muscle cell (SMC) proliferation in the neointima in Groups II and IV. Furthermore, the populations of Treg and CD8+ T cells in total splenocytes were increased in Groups II and IV compared to Groups I and III, respectively. The enlargement of spleens due to CA injury in the Group III was attenuated by fimasartan, as shown in the Group IV. These data indicate that fimasartan significantly reduced SMC proliferation in neointima and increased Treg cells in ApoE KO CA injury mice. CONCLUSIONS: This study suggests fimasartan could be an efficient strategy for reduction of atherosclerotic progression, with a decrease in immune response and systemic inflammation.


Assuntos
Compostos de Bifenilo/farmacocinética , Compostos de Bifenilo/uso terapêutico , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/tratamento farmacológico , Inflamação/sangue , Inflamação/tratamento farmacológico , Neointima/sangue , Neointima/tratamento farmacológico , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Tetrazóis/farmacocinética , Tetrazóis/uso terapêutico , Antagonistas de Receptores de Angiotensina/uso terapêutico , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Interleucina-6/sangue , Masculino , Metaloproteinase 9 da Matriz/sangue , Camundongos , Camundongos Knockout , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue
6.
Int J Mol Sci ; 18(1)2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-28117756

RESUMO

Talin is a focal adhesion protein that activates integrins and recruits other focal adhesion proteins. Talin regulates the interactions between integrins and the extracellular matrix, which are critical for endothelial cells during angiogenesis. In this study, we successfully synthesized a novel talin modulator, N-((2-(1H-indol-3-yl)ethyl)carbamoyl)-2-(benzo[d][1,3]dioxol-5-yloxy)acetamide, referred to as KCH-1521. KCH-1521 was determined to bind talin and modulate downstream signaling molecules of talin. After 24 h of treatment, KCH-1521 changed the cell morphology of human umbilical vein endothelial cells (HUVECs) and reduced focal adhesion protein expression including vinculin and paxillin. Talin downstream signaling is regulated via focal adhesion kinase (FAK), kinase B (AKT), and extracellular signal-regulated kinase (ERK) pathways, however, treatment with KCH-1521 decreased phosphorylation of FAK, AKT, and ERK, leading to reduction of cell proliferation, survival, and angiogenesis. Interestingly, the expression of various angiogenic genes was significantly decreased after treatment with KCH-1521. Also, in vitro tube forming assay revealed that KCH-1521 reduced angiogenic networks in a time-dependent manner. To investigate the reversibility of its effects, KCH-1521 was removed after treatment. HUVECs recovered their morphology through rearrangement of the cytoskeleton and the expression of angiogenic genes was also recovered. By further optimization and in vivo studies of KCH-1521, a novel drug of talin modulation could be used to achieve therapeutic anti-angiogenesis for vascular diseases and cancers.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Talina/metabolismo , Ureia/farmacologia , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos , Ureia/química
7.
Biol Chem ; 397(3): 249-55, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26733157

RESUMO

Key regulatory genes in pluripotent stem cells are of interest not only as reprogramming factors but also as regulators driving tumorigenesis. Nanog is a transcription factor involved in the maintenance of embryonic stem cells and is one of the reprogramming factors along with Oct4, Sox2, and Lin28. Nanog expression has been detected in different types of tumors, and its expression is a poor prognosis for cancer patients. However, there is no clear evidence that Nanog is functionally involved in tumorigenesis. In this study, we induced overexpression of Nanog in mouse embryonic fibroblast cells and subsequently assessed their morphological changes, proliferation rate, and tumor formation ability. We found that Nanog overexpression induced immortalization of mouse embryonic fibroblast cells (MEFs) and increased their proliferation rate in vitro. We also found that formation of tumors after subcutaneous injection of retroviral-Nanog infected MEFs (N-MEFs) into athymic mouse. Cancer-related genes such as Bmi1 were expressed at high levels in N-MEFs. Hence, our results demonstrate that Nanog is able to transform normal somatic cells into tumor cells.


Assuntos
Carcinogênese/genética , Transformação Celular Neoplásica/genética , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Regulação para Cima , Animais , Carcinogênese/patologia , Transformação Celular Neoplásica/patologia , Células Cultivadas , Fibroblastos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Proteína Homeobox Nanog
8.
Int J Mol Sci ; 17(6)2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27231894

RESUMO

Cardiac stem cells (CSCs) were known to secrete diverse paracrine factors leading to functional improvement and beneficial left ventricular remodeling via activation of the endogenous pro-survival signaling pathway. However, little is known about the paracrine factors secreted by CSCs and their roles in cardiomyocyte survival during hypoxic condition mimicking the post-myocardial infarction environment. We established Sca-1+/CD31- human telomerase reverse transcriptase-immortalized CSCs (Sca-1+/CD31- CSCs(hTERT)), evaluated their stem cell properties, and paracrine potential in cardiomyocyte survival during hypoxia-induced injury. Sca-1+/CD31- CSCs(hTERT) sustained proliferation ability even after long-term culture exceeding 100 population doublings, and represented multi-differentiation potential into cardiomyogenic, endothelial, adipogenic, and osteogenic lineages. Dominant factors secreted from Sca-1+/CD31- CSCs(hTERT) were EGF, TGF-ß1, IGF-1, IGF-2, MCP-1, HGF R, and IL-6. Among these, MCP-1 was the most predominant factor in Sca-1+/CD31- CSCs(hTERT) conditioned medium (CM). Sca-1+/CD31- CSCs(hTERT) CM increased survival and reduced apoptosis of HL-1 cardiomyocytes during hypoxic injury. MCP-1 silencing in Sca-1+/CD31- CSCs(hTERT) CM resulted in a significant reduction in cardiomyocyte apoptosis. We demonstrated that Sca-1+/CD31- CSCs(hTERT) exhibited long-term proliferation capacity and multi-differentiation potential. Sca-1+/CD31- CSCs(hTERT) CM protected cardiomyocytes from hypoxic injury partly via MCP-1-dependent mechanism. Thus, they are valuable sources for in vitro and in vivo studies in the cardiovascular field.


Assuntos
Quimiocina CCL2/metabolismo , Miócitos Cardíacos/citologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Células-Tronco/metabolismo , Telomerase/genética , Animais , Diferenciação Celular , Hipóxia Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Humanos , Camundongos , Modelos Biológicos , Células-Tronco/citologia
9.
J Cell Physiol ; 230(8): 1807-21, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25521758

RESUMO

Dimethyl sulfoxide (DMSO) is widely used to induce multilineage differentiation of embryonic and adult progenitor cells. To date, little is known about the mechanisms underlying DMSO-induced mesodermal specification. In this study, we investigated the signaling pathways and lineage-determining genes involved in DMSO-induced mesodermal specification in P19 cells. Wnt/ß-catenin and TGF-ß superfamily signaling pathways such as BMP, TGF-ß and GDF1 signaling were significantly activated during DMSO-induced mesodermal specification. In contrast, Nodal/Cripto signaling pathway molecules, required for endoderm specification, were severely downregulated. DMSO significantly upregulated the expression of cardiac mesoderm markers but inhibited the expression of endodermal and hematopoietic lineage markers. Among the DMSO-activated cell lineage markers, the expression of Mixl1 and Flk1 was dramatically upregulated at both the transcript and protein levels, and the populations of Mixl1+, Flk1+ and Mixl1+/Flk1+ cells also increased significantly. DMSO modulated cell cycle molecules and induced cell apoptosis, resulting in significant cell death during EB formation of P19 cells. An inhibitor of Flk1, SU5416 significantly blocked expressions of TGF-ß superfamily members, mesodermal cell lineage markers and cell cycle molecules but it did not affect Wnt molecules. These results demonstrate that Mixl1 and Flk1 play roles as key downstream or interacting effectors of Wnt/TGF-ß signaling pathway during DMSO-induced mesodermal specification in P19 cells.


Assuntos
Proteínas de Homeodomínio/metabolismo , Mesoderma/citologia , Fator de Crescimento Transformador beta/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Wnt/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Dimetil Sulfóxido/farmacologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Mesoderma/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real
10.
Biochem Biophys Res Commun ; 468(1-2): 372-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26498525

RESUMO

A number of researchers have been reporting a wide range of in vitro and in vivo studies of cell engraftment to enhance angiogenesis using stem cells. Despite these efforts, studies involving three-dimensional (3D) culture method that mimics in vivo environment have not reached its peak yet. In this study, we investigated the change and effects on cellular angiogenic growth factors through sphere formation of adipose stem cell (ASC) which is engineered by poly-2-hydroxyethyl methacrylate (Poly-HEMA). First of all, we successfully induced sphere formation of ASC (sph-ASC) on Poly-HEMA coated plates. sph-ASC represented significantly higher expression levels of anti-apoptotic and hypoxic factors compared to monolayer adherent ASC (adh-ASC). Interestingly, sph-ASC showed higher mRNA levels of the following genes; CD31, CD144, vWF, IGF-2, MCP-1, PDGF-A, VEGF-A, VEGF-C, and FGF-2. In addition, mRNA expressions of angiogenic growth factor receptors such as Flk1, FGFR1, FGFR2, and Tie2 were elevated in sph-ASC. In protein level, Cytokine/Chemokines antibody array revealed a significant increase of FGF-2 in sph-ASC (3.17-fold) compared to adh-ASC. To investigate the effects of FGF-2 on sph-ASC, Matrigel angiogenic invasion assay showed significant reduced level of FGF-2 in FGF-2 siRNA transfected sph-ASC (2.27-fold) compared to negative control siRNA transfected sph-ASC. These findings suggest that Poly-HEMA coated plates induce sphere formation of ASC which has significantly higher expression of FGF-2, and plays a critical role as a major regulating growth factor of in vitro angiogenesis.


Assuntos
Tecido Adiposo/citologia , Materiais Revestidos Biocompatíveis/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Neovascularização Fisiológica , Poli-Hidroxietil Metacrilato/metabolismo , Esferoides Celulares/citologia , Células-Tronco/citologia , Animais , Movimento Celular , Células Cultivadas , Humanos , Camundongos Endogâmicos C57BL , Esferoides Celulares/metabolismo , Células-Tronco/metabolismo , Engenharia Tecidual
11.
Stem Cells ; 32(4): 998-1011, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24715692

RESUMO

Redox balance has been suggested as an important determinant of "stemness" in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I(-/-) or Prx II(-/-) ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity.


Assuntos
Células-Tronco Embrionárias/enzimologia , MAP Quinase Quinase 4/metabolismo , Neurogênese/fisiologia , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Células-Tronco Embrionárias/citologia , MAP Quinase Quinase 4/genética , Camundongos , Camundongos Knockout , Peroxirredoxinas/genética
12.
Circ J ; 79(4): 880-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25737017

RESUMO

BACKGROUND: Aberrant expression of microRNAs is associated with neointimal hyperplasia (NIH) in type 2 diabetes. We prospectively compared the effects of pioglitazone on coronary NIH and changes in microRNAs according to NIH status in type 2 diabetic patients during 9-month follow-up. METHODS AND RESULTS: Type 2 diabetic patients were randomly assigned to the pioglitazone (n=36) or control groups (n=36) after coronary stenting. Primary endpoint was the comparison of changes in neointimal volume on OCT and in the level of circulating microRNA-17,-24,-92a,-126 and -145 during 9-month follow-up. Secondary endpoint was the comparison of changes in brachial artery flow-mediated dilation and inflammatory markers such as IL-6, TNF-α, hsCRP, adiponectin, sICAM-1, and sVCAM-1 between the 2 groups. Neointimal volume was significantly lower in the pioglitazone group (25.02±17.78 mm(3)vs. 55.10±30.01 mm(3), P<0.001) with significant increases in circulating microRNA-24 (0.264±0.084 vs. 0.006±0.030, P<0.001) during follow-up. FMD was significantly greater in the pioglitazone than control group at 9 months (0.47±0.14 mm vs. 0.28±0.18 mm, P<0.05, respectively). Decreases in inflammatory markers such as IL-6, TNF-α, and sVCAM-1 were significantly greater in the pioglitazone than the control group during the follow-up. CONCLUSIONS: Pioglitazone significantly decreased NIH with increases in circulating microRNA-24 at 9-month follow-up. The decrease in microRNA-24 could be used as a potential predictor of increases in NIH in type 2 diabetic patients.


Assuntos
Vasos Coronários , Diabetes Mellitus Tipo 2 , Angiopatias Diabéticas , Hipoglicemiantes/administração & dosagem , MicroRNAs/sangue , Tiazolidinedionas/administração & dosagem , Tomografia de Coerência Óptica , Idoso , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Angiopatias Diabéticas/sangue , Angiopatias Diabéticas/tratamento farmacológico , Angiopatias Diabéticas/patologia , Feminino , Humanos , Hiperplasia/sangue , Hiperplasia/tratamento farmacológico , Hiperplasia/patologia , Masculino , Pessoa de Meia-Idade , Neointima/sangue , Neointima/tratamento farmacológico , Neointima/patologia , Pioglitazona
13.
Biochem Biophys Res Commun ; 449(4): 405-11, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24858689

RESUMO

Proangiogenic cell therapy using autologous progenitors is a promising strategy for treating ischemic disease. Considering that neovascularization is a harmonized cellular process that involves both endothelial cells and vascular smooth muscle cells, peripheral blood-originating endothelial colony-forming cells (ECFCs) and smooth muscle progenitor cells (SMPCs), which are similar to mature endothelial cells and vascular smooth muscle cells, could be attractive cellular candidates to achieve therapeutic neovascularization. We successfully induced populations of two different vascular progenitor cells (ECFCs and SMPCs) from adult peripheral blood. Both progenitor cell types expressed endothelial-specific or smooth muscle-specific genes and markers, respectively. In a protein array focused on angiogenic cytokines, SMPCs demonstrated significantly higher expression of bFGF, EGF, TIMP2, ENA78, and TIMP1 compared to ECFCs. Conditioned medium from SMPCs and co-culture with SMPCs revealed that SMPCs promoted cell proliferation, migration, and the in vitro angiogenesis of ECFCs. Finally, co-transplantation of ECFCs and SMPCs induced robust in vivo neovascularization, as well as improved blood perfusion and tissue repair, in a mouse ischemic hindlimb model. Taken together, we have provided the first evidence of a cell therapy strategy for therapeutic neovascularization using two different types of autologous progenitors (ECFCs and SMPCs) derived from adult peripheral blood.


Assuntos
Células Endoteliais/citologia , Miócitos de Músculo Liso/citologia , Neovascularização Fisiológica , Células-Tronco/citologia , Adulto , Animais , Sobrevivência Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/terapia , Camundongos , Modelos Animais , Miócitos de Músculo Liso/metabolismo , Células-Tronco/metabolismo
14.
Cells ; 12(16)2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37626839

RESUMO

The generation of mature and vascularized human pluripotent stem cell-derived cardiac organoids (hPSC-COs) is necessary to ensure the validity of drug screening and disease modeling. This study investigates the effects of cellular aggregate (CA) stemness and self-organization on the generation of mature and vascularized hPSC-COs and elucidates the mechanisms underlying cardiac organoid (CO) maturation and vascularization. COs derived from 2-day-old CAs with high stemness (H-COs) and COs derived from 5-day-old CAs with low stemness (L-COs) were generated in a self-organized microenvironment via Wnt signaling induction. This study finds that H-COs exhibit ventricular, structural, metabolic, and functional cardiomyocyte maturation and vessel networks consisting of endothelial cells, smooth muscle cells, pericytes, and basement membranes compared to L-COs. Transcriptional profiling shows the upregulation of genes associated with cardiac maturation and vessel formation in H-COs compared with the genes in L-COs. Through experiments with LIMK inhibitors, the activation of ROCK-LIMK-pCofilin via ECM-integrin interactions leads to cardiomyocyte maturation and vessel formation in H-COs. Furthermore, the LIMK/Cofilin signaling pathway induces TGFß/NODAL and PDGF pathway activation for the maturation and vascularization of H-COs. The study demonstrates for the first time that LIMK/Cofilin axis activation plays an important role in the generation of mature and vascularized COs.


Assuntos
Células Endoteliais , Organoides , Humanos , Miócitos Cardíacos , Via de Sinalização Wnt , Fatores de Despolimerização de Actina , Matriz Extracelular , Neovascularização Patológica , Integrinas
15.
J Cell Physiol ; 227(11): 3678-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22378194

RESUMO

To identify potential downstream targets of Nanog, a key transcription factor in the maintenance of pluripotency of embryonic stem (ES) and embryonal carcinoma (EC) cells, global gene expression profiles in Nanog small interfering RNA (siRNA)-transfected P19 EC stem cells were performed using cDNA, 60-mer, and 30-mer microarray platforms. The putative Nanog target genes identified by Nanog silencing were verified using reverse transcription-polymerase chain reaction after Nanog overexpression. Downregulation of Nanog in P19 cells resulted in reduction of pluripotency markers, such as Fgf4, Klf2, Mtf2, Oct-4, Rex1, Sox1, Yes, and Zfp143, whereas overexpression of Nanog in P19 cells reversely upregulated their expression. However, expressions of pluripotency markers Cripto, germ cell nuclear factor, Sox2, and Zfp57 as well as leukemia inhibitory factor (LIF)/Stat3 pathway molecules LIF, IL6st, and Stat3 were not affected after 48 h transfection with Nanog siRNA or construct. Nanog silencing also downregulated expression of molecules involved in the p53- and cell cycle-signaling pathway (Atf3, Jdp2, Cul3, Hist1hic, and Bcl6), whereas expression of E2f1, Tob1, Lyn, and Smarcc1 was upregulated by Nanog silencing. Expressions of cyclins D1, D2, D3, and E1 as well as cyclin-dependent kinase (Cdk) 1 and Cdk6 were downregulated by Nanog silencing in P19 cells, whereas Nanog overexpression reversely increased their expressions. Taken together, examination of global transcriptional changes after Nanog silencing followed by verification by Nanog overexpression has revealed new molecules involved in the maintenance of self-renewal and in the regulation of the p53- and cell cycle-pathway of P19 cells.


Assuntos
Proteínas de Ciclo Celular , Células-Tronco de Carcinoma Embrionário , Células-Tronco Embrionárias , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Homeodomínio/genética , Humanos , Proteína Homeobox Nanog , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , RNA Interferente Pequeno , Transdução de Sinais , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo
16.
Circ J ; 76(5): 1213-21, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22354195

RESUMO

BACKGROUND: The correlations between circulating angiogenic cell mobilizations and improvement of microvascular integrity were investigated in patients (n=110) with acute myocardial infarction (AMI) during an 8-month follow up. METHODS AND RESULTS: Coronary flow reserve (CFR) was measured at baseline and at 8 months by using an intracoronary Doppler wire. Serial changes in the absolute numbers of circulating angiogenic cells such as CD34+, CXCR4+, CD117+, CD133+ and C-met+ were measured at baseline, day 1, day 5 and at 8 months. The absolute numbers of circulating angiogenic cells at day 1 were significantly higher than those at baseline. A positive correlation was found between the numbers of circulating angiogenic cells of CD34+, CXCR4+, CD117+ and CD133+ cells at day 1 and the CFR changes from baseline. The cut-off value of CFR changes at 8 months by a receiver operating characteristic curve between a circulating CD34+ cell at day 1 and changes of CFR at 8 months was 0. Late-loss showed the positive correlation with the absolute number of C-met+ cells and the negative correlation with the absolute number of CXCR4+ cells after AMI. The negative correlation was found between changes in high-sensitive C-reactive protein and soluble intercellular adhesion molecule-1 and changes in CFR at 8 months. CONCLUSIONS: The recovery of microvascular integrity after acute ischemic injury was expedited by the increases in circulating angiogenic cell mobilization together with the greater decreases in inflammatory cytokines. The improvement in CFR could be predicted by the measurement of circulating angiogenic cells after AMI.


Assuntos
Circulação Coronária , Mobilização de Células-Tronco Hematopoéticas , Infarto do Miocárdio/sangue , Infarto do Miocárdio/fisiopatologia , Células-Tronco/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/sangue , Citocinas/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Estudos Prospectivos , Receptores CXCR4/sangue , Células-Tronco/patologia , Fatores de Tempo
17.
Biosens Bioelectron ; 198: 113828, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-34847362

RESUMO

An exosome species containing CD63 as a marker of melanoma was isolated from bulk exosome population and used as a sample for detecting malignant melanoma. A calcium binding protein (CBP) was produced and then used to raise monoclonal antibody. The antibody was sensitive to a conformational change of CBP caused by Ca2+ binding. Immuno-magnetic beads were prepared by immobilizing the conformation-sensitive binder and subsequent binding of CBP conjugated with the capture antibody specific to CD63. These immuno-beads were used to isolate CD63-positive exosome from a bulk exosome sample (normal or melanoma) based on the 'calcium switch-on/off' mechanism through magnetic separation. After recovery, the subpopulation sample was analyzed by immunoassays for cavelion1 (Cav1), CD81, and CD9 as sub-subpopulation markers. Normalized signals of Cav1 and/or CD81 over CD9 were higher in melanoma samples than in normal samples, depending on clinical stages (I, II, and IV) of patients. This was in contrast to assay results for the bulk exosome population that showed a completely mixed state of melanoma and normal samples. These results showed that an exosome subpopulation sample prepared using a 'Ca2+-dependent switch' technology might be useful for diagnosing malignant melanoma at an early stage to increase 5-year survival rates.


Assuntos
Técnicas Biossensoriais , Exossomos , Melanoma , Biomarcadores Tumorais , Cálcio , Humanos , Melanoma/diagnóstico
18.
Phytother Res ; 25(2): 239-49, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20641058

RESUMO

The effects of red ginseng extract on circulating angiogenic cell mobilization and improvement of microvascular integrity were evaluated in ST-elevation acute myocardial infarction (AMI) patients during 8-month follow-up. AMI patients (n = 50) were randomly assigned to the red ginseng group (3 g/day, n = 25) or the placebo group (n = 25) after coronary stenting. Coronary flow reserve (CFR) was measured at baseline and at 8 months with an intracoronary Doppler wire. Serial changes in the absolute numbers of circulating angiogenic cells such as CD34(+) , CXCR4(+) , CD117(+) , CD133(+) and C-met(+) were measured at baseline, 1 day, 5 days and at 8 months. CFR were similar between the two groups at baseline, and CFR was significantly higher in the red ginseng group than in the placebo group (2.80 ± 0.91 and 2.56 ± 0.77, p < 0.05, respectively) after 8 months of red ginseng administration. The absolute numbers of circulating CD34(+) , CXCR4(+) and CD117(+) cells were significantly higher in the red ginseng group at 1 and 5 days after stenting. Significant positive correlations were found between the numbers of circulating angiogenic cells at day 1 and the changes from baseline in CFR for CD34(+) , CXCR4(+) , CD117(+) and C-met(+) cells. Red ginseng extract increased CD34(+) , CXCR4(+) and CD117(+) circulating angiogenic cell mobilization and decreased inflammation in AMI patients, thereby improving CFR during the 8-month follow-up.


Assuntos
Coração/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Panax/química , Fitoterapia , Extratos Vegetais/uso terapêutico , Idoso , Velocidade do Fluxo Sanguíneo , Ecocardiografia Doppler , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Fisiológica/efeitos dos fármacos , Extratos Vegetais/farmacologia , Estudos Prospectivos , Método Simples-Cego
19.
Biofabrication ; 13(4)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34404035

RESUMO

A novel tissue engineering strategy using 3D bio-print technology has become a promising therapeutic method for acute myocardial infarction (AMI) in an animal model. However, the application of 3D bio-printed tissue remains limited due to poor graft survival. Therefore, it is a scientific priority to enhance graft survival by precisely adjusting the 3D environment of encapsulated cells. In this study, novel transplantable 3D cardiac mesh (cMesh) tissue with a porous mesh structure was presented using human cardiomyocytes, human cardiac fibroblasts, and gelatin-methacryloyl-collagen hydrogel. Cardiomyocytes and cardiac fibroblasts were well spreaded. The cardiomyocytes were connected with a gap junction channel in bio-printed cMesh and a 3D cardiac patch with an aggregated structure. Porous cMesh demonstrated structural advantages by increased phosphorylation of mTOR, AKT, and ERK signals associated with cell survival. Transplanted cMesh in rats with AMI improved long-term graft survival, vessel formation, and stabilization, reduced fibrosis, increased left ventricle thickness, and enhanced cardiac function. Our results suggest that porous cMesh provides structural advantages and a positive therapeutic effect in an AMI animal model.


Assuntos
Infarto do Miocárdio , Telas Cirúrgicas , Animais , Gelatina , Hidrogéis , Infarto do Miocárdio/terapia , Miócitos Cardíacos , Impressão Tridimensional , Ratos , Engenharia Tecidual
20.
Biomaterials ; 278: 121133, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34571434

RESUMO

The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.


Assuntos
Proteínas de Homeodomínio , Células-Tronco Pluripotentes Induzidas , Fatores de Determinação Direita-Esquerda , Miócitos Cardíacos , Células-Tronco Pluripotentes , Fatores de Transcrição , Diferenciação Celular , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Determinação Direita-Esquerda/metabolismo , Mesoderma , Organoides , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteína Homeobox PITX2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA