Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Vitreoretin Dis ; 5(3): 199-207, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34632255

RESUMO

PURPOSE: To compare dilated smartphone-based imaging with a nonmydriatic, tabletop fundus camera as a teleophthalmology screening tool for diabetic retinopathy (DR). METHODS: This was a single-institutional, cross-sectional, comparative-instrument study. Fifty-six patients at a safety-net hospital underwent teleophthalmology screening for DR using standard, nonmydriatic fundus photography with a tabletop camera (Nidek NM-1000) and dilated fundus photography using a smartphone camera with lens adapter (Paxos Scope, Verana Health). Masked graders performed standardized photo grading. Quantitative comparisons were performed employing descriptive, κ, Bland-Altman, and receiver operating characteristic analyses. RESULTS: Posterior segment photography was of sufficient quality to grade in 89% of mydriatic smartphone-imaged eyes and in 86% of nonmydriatic tabletop camera-imaged eyes (P = .03). Using the tabletop camera as the reference to detect moderate nonproliferative DR or worse (referral-warranted DR), mydriatic smartphone-acquired photographs were found to be 82% sensitive and 96% specific. Dilated smartphone imaging detected referral-warranted DR in 3 eyes whose tabletop camera imaging did not demonstrate referral-warranted DR. Secondary masked review of medical records for the discordances in referral-warranted status from the two imaging modalities was performed, and it revealed revised sensitivity and specificity values of 95% and 98%, respectively. Overall, there was good agreement between tabletop camera and smartphone-acquired photo grades (κ = 0.91 ± 0.1, P < .001; area under the receiver operating characteristic curve = 0.99, 95% CI, 0.98-1.00). CONCLUSIONS: Mydriatic smartphone-based imaging resulted in fewer ungradable photos compared to nonmydriatic table-top camera imaging and detected more patients with referral-warranted DR. Our study supports the use of mydriatic smartphone teleophthalmology as an alternative method to screen for DR.

3.
Prostate Int ; 9(1): 31-41, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33912512

RESUMO

BACKGROUND: This study investigated the inhibition of tumor growth in castrate-resistant prostate cancer (CRPC)-bearing mice by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-overexpressing adipose-derived stem cells (ADSCs) (hTERT-ADSC.sTRAIL), which was enhanced by combined treatment with CPT-11. MATERIALS AND METHODS: An hTERT-ADSC.sTRAIL cell line was established by transfection with a lentiviral vector (CLV-Ubic) encoding the human sTRAIL gene. Quantitative polymerase chain reaction and Western blots were performed to confirm gene overexpression. An invasion study for the selective migration ability toward PC3 cells was performed. In the in vivo study, the tumor volume in mice treated with ADSC. sTRAIL and CPT-11 was measured. RESULTS: Carboxylesterase was generated from hTERT-ADSCs. The gene expression of sTRAIL from hTERT-ADSC.sTRAIL was shown. The directional migration of ADSC.sTRAIL cells toward PC3 cells was significantly stimulated by PC3 cells in vitro (P < 0.05). In the in vitro study, the viability of PC3 cells significantly decreased in the presence of ADSC.sTRAIL (62.7 ± 2.0%) and CPT-11 compared with that of CPT-11 alone (83.0 ± 1.0%) at a cell ratio as low as 0.05 (PC3: ADSC.sTRAIL) (P < 0.05). The proportion of apoptotic PC3 cells significantly increased in the presence of ADSC.sTRAIL (37.2 ± 2.1%) and CPT-11 compared with that of CPT-11 alone (16.5 ± 1.0%) (P < 0.05). In the in vivo study, the inhibition of tumor growth in CRPC-bearing mice by TRAIL-overexpressing adipose stem cells was enhanced by combined treatment with the chemotherapeutic agent CPT-11 compared with that in the treatment with cpt-11 alone. Immunohistochemical staining of the removed tumors showed anti-TRAIL-positive cells and apoptotic bodies after hTERT-ADSC.sTRAIL treatment or combined treatment with hTERT-ADSC.sTRAIL and CPT-11. CONCLUSIONS: Therapeutic stem cells expressing sTRAIL genes combined with CPT-11 can provide a new strategy for treating CRPC in clinical trials using the patients' own ADSCs.

4.
Int J Med Sci ; 17(9): 1235-1245, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32547319

RESUMO

Diabetes-mediated hyperglycemia is a major risk factor for renal fibrosis, resulting in the development of chronic kidney diseases. To address this issue, the effect of melatonin, which has an antioxidative potential, on renal fibrosis in human renal proximal tubule epithelial cells under high glucose conditions was investigated. Under high glucose conditions, the generation of reactive oxygen species was drastically increased in human renal proximal tubule epithelial cells, which lead to the inhibition of cell proliferation, enlargement of cell size, reduction of cell survival, and suppression of antioxidant enzyme activities. High glucose also increased the expression of transforming growth factor-ß, leading to an increase in Smad2 phosphorylation. These fibrotic phenotype changes increased the expression of fibrosis-mediated extracellular matrix proteins, such as fibronectin, collagen I, and α-smooth muscle actin. In addition, the level of cellular prion protein (PrPC), which is associated with several biological processes, was decreased by exposure to high glucose conditions. Melatonin recovered the expression levels of PrPC under high glucose conditions via phosphorylation of Akt, resulting in the prevention of high glucose-induced fibrosis. In particular, overexpression of PrPC blocked the high glucose-mediated fibrotic phenotype change. These findings indicate that melatonin could be a powerful agent for treating hyperglycemia-induced renal fibrosis.


Assuntos
Catalase/metabolismo , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Melatonina/uso terapêutico , Fator de Crescimento Transformador beta/metabolismo , Western Blotting , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Fibronectinas/metabolismo , Glucose/farmacologia , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , L-Lactato Desidrogenase/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Priônicas/metabolismo , Ensaio de Radioimunoprecipitação , Transdução de Sinais/efeitos dos fármacos , Células Th1
5.
Anticancer Res ; 40(3): 1419-1426, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32132038

RESUMO

BACKGROUND/AIM: Anti-cancer drug resistance restricts the efficacy of chemotherapy in malignant tumors. Casein kinase 2α (CK2α) is highly expressed in 5-fluorouracil (5FU)-resistant colorectal cancer (CRC) cells. We hypothesized that inhibition of CK2α might reduce CRC resistance to 5FU. MATERIALS AND METHODS: To investigate the role of CK2α in 5FU-resistant CRC cells, we assessed cell viability, apoptosis, cyclin-dependent kinase 4 (CDK4) activity, cell-cycle progression, invasion, and sphere formation in 5FU-resistant CRC cells. RESULTS: CK2α levels were significantly increased in 5FU-resistant CRC cells compared to those in wild-type CRC cells. During exposure to 5FU, viability, CDK4 activity, cell-cycle progression, invasion, and sphere formation were enhanced, while apoptosis was decreased in 5FU-resistant CRC cells. These effects were mediated by the inhibiting effects of CK2α on endoplasmic reticulum (ER) stress. Combination of CK2α knockdown with 5FU treatment promoted apoptosis of 5FU-resistant CRC cells by inducing ER stress. CONCLUSION: 5FU treatment in combination with a CK2α inhibitor may exert a synergistic effect against drug-resistant cancer cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Caseína Quinase II/antagonistas & inibidores , Neoplasias Colorretais/tratamento farmacológico , Fluoruracila/farmacologia , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Quinase 4 Dependente de Ciclina/metabolismo , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fluoruracila/administração & dosagem , Humanos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia
6.
Biochem Biophys Res Commun ; 525(2): 354-359, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32089265

RESUMO

Ischemic injury is a major risk factor for fibrosis. However, the precise mechanisms by which fibrosis is regulated and induced under ischemic oxidative stress conditions are unknown. To address this, we investigated the effect of melatonin on ischemia-induced fibrosis. In a hindlimb ischemia mouse model, ischemia induced fibrosis by increasing inflammation and the expression of extracellular matrix (ECM) proteins. Melatonin prevented ischemia-induced fibrosis in the injured tissues. In particular, melatonin suppressed the fibrosis-mediated inflammatory reaction in myoblasts through the microRNA-149 (miR-149)/indoleamine 2,3-dioxygenase-1 (IDO-1) signaling pathway. The melatonin-induced increase in miR-149 inhibited the expression of the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) and ECM components, such as collagen I and fibronectin. In addition, melatonin increased antioxidative activity and mitochondrial function in myoblasts via the miR-149/peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) signaling axis, and the anti-fibrotic effects of melatonin were blocked by inhibition of miR-149. These findings indicate that melatonin is a key target molecule in fibrosis related to ischemic diseases and that miR-149 might be a novel target for the treatment of ischemia-induced fibrosis.


Assuntos
Fibrose/etiologia , Isquemia/patologia , Melatonina/farmacologia , MicroRNAs/efeitos dos fármacos , Animais , Fibrose/prevenção & controle , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , MicroRNAs/metabolismo , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores
7.
Biomol Ther (Seoul) ; 28(3): 240-249, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31739381

RESUMO

Despite the therapeutic effect of mesenchymal stem cells (MSCs) in ischemic diseases, pathophysiological conditions, including hypoxia, limited nutrient availability, and oxidative stress restrict their potential. To address this issue, we investigated the effect of melatonin on the bioactivities of MSCs. Treatment of MSCs with melatonin increased the expression of peroxisome proliferatoractivated receptor gamma coactivator-1 alpha (PGC-1α). Melatonin treatment enhanced mitochondrial oxidative phosphorylation in MSCs in a PGC-1α-dependent manner. Melatonin-mediated PGC-1α expression enhanced the proliferative potential of MSCs through regulation of cell cycle-associated protein activity. In addition, melatonin promoted the angiogenic ability of MSCs, including migration and invasion abilities and secretion of angiogenic cytokines by increasing PGC-1α expression. In a murine hindlimb ischemia model, the survival of transplanted melatonin-treated MSCs was significantly increased in the ischemic tissues, resulting in improvement of functional recovery, such as blood perfusion, limb salvage, neovascularization, and protection against necrosis and fibrosis. These findings indicate that the therapeutic effect of melatonin-treated MSCs in ischemic diseases is mediated via regulation of PGC-1α level. This study suggests that melatonin-induced PGC-1α might serve as a novel target for MSC-based therapy of ischemic diseases, and melatonin-treated MSCs could be used as an effective cell-based therapeutic option for patients with ischemic diseases.

8.
Mar Drugs ; 17(9)2019 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-31480724

RESUMO

Mitochondria are considered to be the powerhouses of cells. They are the most commonly damaged organelles within dopaminergic neurons in patients with Parkinson's disease (PD). Despite the importance of protecting neuronal mitochondria in PD patients, the detailed mechanisms underlying mitochondrial dysfunction during pathogenesis and pathophysiological progression of PD have not yet been elucidated. We investigated the protective action of fucoidan against the detrimental action of 1-methyl-4-phenyl-pyridinium (MPP+), a neurotoxin used to model PD, in the mitochondria of SH-SY5Y neural cells. Fucoidan increased the expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and protected the cells from MPP+-induced apoptosis by upregulating the 5' adenosine monophosphate-activated protein kinase (AMPK)-PGC-1α axis. These effects were blocked by the silencing of the PGC-1α axis. These results indicated that fucoidan protects SH-SY5Y cells from mitochondrial dysfunction and cell death caused by MPP+ treatment, via the AMPK-PGC-1α axis. These findings also suggest that fucoidan could potentially be used as a therapeutic agent for PD.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Morte Celular/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Doenças Mitocondriais/tratamento farmacológico , Neurônios/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Polissacarídeos/farmacologia , Adenilato Quinase/metabolismo , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/efeitos dos fármacos , Humanos , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Neurônios/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo
9.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-30959809

RESUMO

Anti-cancer drug resistance is a serious issue for patients with colorectal cancer (CRC). Although recent studies have shown the mechanism by which CRC cells become drug resistant, novel strategies for overcoming this drug resistance have not yet been developed. To address this problem, we characterized 5-fluorouracil (5FU)-resistant CRC cells after treatment with 5FU, and focused on the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in these cells. In 5FU-resistant CRC cells, the 5FU did not considerably decrease the mitochondrial biogenesis or mitochondrial complex I and IV activities, and only partially decreased the antioxidant enzymatic activity, oxygen consumption ratio, and cell survival. The expression of PGC-1α was remarkably increased in the 5FU-resistant CRC cells compared with the 5FU-sensitive CRC cells. The 5FU-resistant CRC cells displayed enhanced mitochondrial biogenesis, oxidative phosphorylation, and antioxidant enzyme activities against 5FU-induced reactive oxygen species, because of the increased expression of PGC-1α. PGC-1α inhibited 5FU-induced endoplasmic reticulum (ER) stress in the 5FU-resistant CRC cells, resulting in the suppression of apoptosis. These findings reveal that PGC-1α plays an important role in drug resistance in 5FU-resistant CRC cells. Moreover, PGC-1α could serve as a novel target in patients with 5FU-resistant CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Estresse do Retículo Endoplasmático , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fluoruracila/farmacologia , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo
10.
Redox Biol ; 22: 101144, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30785084

RESUMO

Although autologous human mesenchymal stem cells (hMSCs) are a promising source for regenerative stem cell therapy in chronic kidney disease (CKD), the barriers associated with pathophysiological conditions limit therapeutic applicability to patients. We confirmed that level of cellular prion protein (PrPC) in serum was decreased and mitochondria function of CKD-derived hMSCs (CKD-hMSCs) was impaired in patients with CKD. We proved that treatment of CKD-hMSCs with tauroursodeoxycholic acid (TUDCA), a bile acid, enhanced the mitochondrial function of these cells through regulation of PINK1-PrPC-dependent pathway. In a murine hindlimb ischemia model with CKD, tail vein injection of TUDCA-treated CKD-hMSCs improved the functional recovery, including kidney recovery, limb salvage, blood perfusion ratio, and vessel formation along with restored expression of PrPC in the blood serum of the mice. These data suggest that TUDCA-treated CKD-hMSCs are a promising new autologous stem cell therapeutic intervention that dually treats cardiovascular problems and CKD in patients.


Assuntos
Isquemia/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas PrPC/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Animais , Biomarcadores , Proliferação de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Isquemia/patologia , Isquemia/terapia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
11.
J Pineal Res ; 66(1): e12535, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30372554

RESUMO

Although mesenchymal stem cell (MSC)-based therapy is a treatment strategy for ischemic diseases associated with chronic kidney disease (CKD), MSCs of CKD patients undergo accelerated senescence, with decreased viability and proliferation upon uremic toxin exposure, inhibiting their utility as a potent stem cell source for transplantation therapy. We investigated the effects of melatonin administration in protecting against cell senescence and decreased viability induced by pathophysiological conditions near the engraftment site. MSCs harvested from CKD mouse models were treated with H2 O2 to induce oxidative stress. CKD-derived MSCs exhibited greater oxidative stress-induced senescence than normal-mMSCs, while melatonin protected CKD-mMSCs from H2 O2 and associated excessive senescence. The latter was mediated by PrPC -dependent mitochondrial functional enhancement; melatonin upregulated PrPC , which bound PINK1, thus promoting mitochondrial dynamics and metabolism. In vivo, melatonin-treated CKD-mMSCs survived longer, with increased secretion of angiogenic cytokines in ischemic disease engraftment sites. CKD-mMSCs are more susceptible to H2 O2 -induced senescence than normal-mMSCs, and melatonin administration protects CKD-mMSCs from excessive senescence by upregulating PrPC and enhancing mitochondrial function. Melatonin showed favorable therapeutic effects by successfully protecting CKD-mMSCs from related ischemic conditions, thereby enhancing angiogenesis and survival. These results elucidate the mechanism underlying senescence inhibition by melatonin in stem cell-based therapies using mouse-derived CKD-mMSCs.


Assuntos
Senescência Celular/efeitos dos fármacos , Melatonina/uso terapêutico , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Insuficiência Renal Crônica/tratamento farmacológico , Animais , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Cicatrização/efeitos dos fármacos
12.
Cell Prolif ; 52(2): e12545, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30430685

RESUMO

OBJECT: The purpose of this study was to explore whether melatonin could protect mesenchymal stem cells (MSCs) against ischaemic injury, by inhibiting endoplasmic reticulum (ER) stress and autophagy both in vivo and in vitro. MATERIALS AND METHODS: To confirm the protective effect of melatonin against ER stress in MSCs, markers of cell viability, apoptosis and autophagy were analysed. To further investigate the regenerative effect of melatonin-treated MSCs in ischaemic tissues, a murine hindlimb ischaemic model was established. RESULTS: Under oxidative stress conditions, treatment with melatonin suppressed the activation of ER stress-associated proteins and autophagy-associated proteins acting through upregulation of cellular prion protein (PrPC ) expression. Consequently, inhibition of apoptotic cell death occurred. Melatonin also promoted the activation of MnSOD and catalase activities in MSCs. In a murine hindlimb ischaemia model, melatonin-treated MSCs also enhanced the functional limb recovery as well as neovascularization. These beneficial effects of melatonin were all blocked by knock-down of PrPC expression. CONCLUSION: Melatonin protects against ER stress/autophagy-induced apoptotic cell death by augmenting PrPC expression. Thus, melatonin-treated MSCs could be a potential cell-based therapeutic agent for ER stress-induced ischaemic diseases, and melatonin-induced PrPC might be a key molecule in ameliorating ER stress and autophagy.


Assuntos
Antioxidantes/uso terapêutico , Autofagia/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Isquemia/tratamento farmacológico , Melatonina/uso terapêutico , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteínas Priônicas/metabolismo , Animais , Antioxidantes/farmacologia , Células Cultivadas , Membro Posterior/irrigação sanguínea , Membro Posterior/efeitos dos fármacos , Membro Posterior/metabolismo , Membro Posterior/patologia , Isquemia/metabolismo , Isquemia/patologia , Masculino , Melatonina/farmacologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Estresse Oxidativo/efeitos dos fármacos , Proteínas Priônicas/análise
13.
Int J Mol Sci ; 19(10)2018 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-30250007

RESUMO

Mesenchymal stem cells (MSC) could be a candidate for cell-based therapy in chronic kidney disease (CKD); however, the uremic toxin in patients with CKD restricts the therapeutic efficacy of MSCs. To address this problem, we explored the effect of pioglitazone as a measure against exposure to the uremic toxin P-cresol (PC) in MSCs. Under PC exposure conditions, apoptosis of MSCs was induced, as well as PC-induced dysfunction of mitochondria by augmentation of mitofusion, reduction of mitophagy, and inactivation of mitochondrial complexes I and IV. Treatment of MSCs with pioglitazone significantly inhibited PC-induced apoptosis. Pioglitazone also prevented PC-induced mitofusion and increased mitophagy against PC exposure through up-regulation of phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK-1). Furthermore, pioglitazone protected against PC-induced mitochondrial dysfunction by increasing the cytochrome c oxidase subunit 4 (COX4) level and activating complexes I and IV, resulting in enhancement of proliferation. In particular, activation of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) regulated the pioglitazone-mediated up-regulation of PINK-1. These results indicate that pioglitazone protects MSCs against PC-induced accumulated mitochondrial dysfunction via the NF-κB⁻PINK-1 axis under P-cresol exposure conditions. Our study suggests that pioglitazone-treated MSCs could be a candidate for MSC-based therapy in patients with CKD.


Assuntos
Cresóis/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , NF-kappa B/metabolismo , Pioglitazona/farmacologia , Proteínas Quinases/metabolismo , Apoptose , Proliferação de Células , Células Cultivadas , Humanos , Hipoglicemiantes/farmacologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Transdução de Sinais , Regulação para Cima
14.
Tissue Cell ; 53: 93-103, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30060833

RESUMO

Mesenchymal stem cells (MSCs) are widely used in transplantation therapy due to their multilineage differentiation potential, abundance, and immuno-modulating ability. However, the risk of allograft rejection limits their application. Here, we proposed a novel method to facilitate MSC transplantation with enhanced applicability and efficacy. We cultured human adipose-derived MSCs in a 3D culture under in vitro expansion conditions and under conventional 2D adherent culture conditions. MSC spheroids promoted extracellular matrix molecules that stimulate MSC proliferation, and produced more angiogenic cytokines such as vascular endothelial growth factor, hepatocyte growth factor, and fibroblast growth factor than 2D-cultured MSCs. Further, MSC spheroids showed increased IDO expression, increased proportion of M2 macrophages, and decreased macrophage proliferation, compared to 2D-cultured MSCs. Next, we proposed the wrapping of autologous cell sheets from the recipient around in-vitro-grown MSC spheroids to prevent allogenic immune rejection during transplantation. Myoblasts from C57BL/6 mice were used to prepare a stem cell composite sheet containing human-derived MSC spheres. The transplantation of MSC spheroids increased the survival rate and decreased the inflammatory response of the immunocompetent C57BL/6 ischemic mice. Thus, combining 3D-cultured MSC spheroid technology with immune evasion stem cell composite sheet improved the outcome and strengthened the protection against allogenic immune rejection.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Mioblastos Esqueléticos/transplante , Esferoides Celulares/metabolismo , Esferoides Celulares/transplante , Aloenxertos , Animais , Autoenxertos , Técnicas de Cocultura , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Esferoides Celulares/citologia
15.
J Pineal Res ; 65(4): e12519, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30091203

RESUMO

Melatonin suppresses tumor development. However, the exact relationship between melatonin and cancer stem cells (CSCs) is poorly understood. This study found that melatonin inhibits colon CSCs by regulating the PrPC -Oct4 axis. In specimens from patients with colorectal cancer, the expressions of cellular prion protein (PrPC ) and Oct4 were significantly correlated with metastasis and tumor stages. Co-treatment with 5-fluorouracil (5-FU) and melatonin inhibited the stem cell markers Oct4, Nanog, Sox2, and ALDH1A1 by downregulating PrPC . In this way, tumor growth, proliferation, and tumor-mediated angiogenesis were suppressed. In colorectal CSCs, PRNP overexpression protects Oct4 against inhibition by 5-FU and melatonin. In contrast, Nanog, Sox2, and ALDH1A1 have no such protection. These results indicate that PrPC directly regulates Oct4, whereas it indirectly regulates Nanog, Sox2, and ALDH1A1. Taken together, our findings suggest that co-treatment with anticancer drug and melatonin is a potential therapy for colorectal cancer. Furthermore, PrPC maintains cancer stemness during tumor progression. Therefore, targeting the PrPC -Oct4 axis may prove instrumental in colorectal cancer therapy.


Assuntos
Fluoruracila/farmacologia , Melatonina/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Priônicas/metabolismo , Idoso , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Autofagia/efeitos dos fármacos , Autofagia/genética , Colo/efeitos dos fármacos , Colo/metabolismo , Neoplasias do Colo/metabolismo , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Príons/metabolismo , RNA Interferente Pequeno/genética , Retinal Desidrogenase , Fatores de Transcrição SOXB1/metabolismo
16.
Int J Mol Sci ; 19(5)2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29734669

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) are a promising source for regenerative medicine. However, their therapeutic potential in patients with chronic kidney disease (CKD) is restricted by the presence of uremic toxins. To address this limitation, we explored the protective effect of melatonin and pioglitazone on MSCs undergoing senescence induced by the uremic toxin, indoxyl sulfate (IS). METHODS: MSC senescence was induced by IS, and the therapeutic effects of melatonin and pioglitazone were identified. The expression of cellular prion protein (PrPC) was suppressed by transfection of MSCs with prion protein gene (PRNP) siRNA. Subsequently, these cells were used to study the protective effects of melatonin and pioglitazone against IS-induced senescence; Results: The IS-induced senescence of MSCs was significantly reduced by co-treatment with melatonin and pioglitazone compared to treatment with melatonin or pioglitazone alone. In the presence of IS, the reduced MSC proliferation was rescued by co-treatment with melatonin and pioglitazone. Melatonin and pioglitazone enhanced the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) in MSCs, which resulted in the augmentation of PrPC level. The inhibitory effect of the co-treatment with melatonin and pioglitazone on IS-induced senescence in MSCs was blocked by the knockdown of PRNP. In addition, the restorative effect of the co-treatment on the reduced MSC proliferation induced by IS was also blocked by the knockdown of PRNP. These findings indicate that co-treatment with melatonin and pioglitazone protected MSCs from uremic toxin-induced senescence through the regulation of the PPAR-γ-PrPC axis. CONCLUSIONS: Our study suggests that co-treatment of MSCs with melatonin and pioglitazone may represent a novel strategy for the development of MSC-based therapies for patients with CKD.


Assuntos
Senescência Celular/genética , Melatonina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Tecido Adiposo/citologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Indicã/toxicidade , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , PPAR gama/genética , Pioglitazona , Proteínas Priônicas/genética , Transdução de Sinais/efeitos dos fármacos
17.
Cell Physiol Biochem ; 46(5): 1835-1848, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29705776

RESUMO

BACKGROUND/AIMS: Stem cell transplantation has emerged as a promising therapeutic strategy, but the exact mechanisms by which stem cells exposed to hypoxic conditions increase the survival rate and rescue ischemic injury at the graft site are not well known. In this study, we aimed to determine if c-Met-activated mesenchymal stem cells (MSCs) pre-exposed to hypoxia promote therapeutic efficacy when transplanted to ischemic models, and whether c-Met interacts with cellular prion protein (PrPC) present in the ischemic tissue. METHODS: Western blot analysis was performed to determine the expression levels of PrPC, C-caspase-3, and C-PARP-1, as well as the phosphorylation of Akt, p38, JNK, and BAX. A co-immunoprecipitation assay was performed to show that PrPC binds with c-Met in vitro. An adhesion assay was performed to explore the alterations in MSCs attached to myoblasts (in vitro), and an invasion assay was performed to determine the effect on MSC invasion capacity upon interaction with myoblast-induced c-Met and PrPC. CD31-positive capillaries and αSMA-positive arterioles in in vivo hindlimb ischemic tissue were quantified by immunofluorescence staining. The level of apoptosis in the tissue of each group was assessed by quantifying the number of C-caspase-3-positive cells. Finally, laser Doppler technology was utilized to detect the enhanced angiogenic effects in vivo. RESULTS: We showed that hypoxic conditions increased PrPC levels in vivo (hindlimb ischemic tissue) and in vitro (myoblasts) and increased c-Met levels in MSCs. To identify the relationship between c-Met from MSCs and PrPC from myoblasts, we used a co-culturing system with myoblasts and MSCs pre-exposed to hypoxia. Hypoxia increased the phosphorylation of mitogen-activated protein kinases. Transplantation of hypoxia-pre-exposed MSCs to the ischemic site increased anti-apoptosis and enhanced the survival and proliferation of transplanted MSCs in a murine hindlimb model, resulting in improved functional recovery of the ischemic tissue. All the aforementioned effects were inhibited by the pretreatment of MSCs with the c-Met-neutralizing antibody Conclusion: c-Met-activated MSCs pre-exposed to hypoxia interact with PrPC at the site of ischemic injury to increase the efficiency of MSC transplantation. Hence, our study demonstrated that c-Met is a potential target for MSC-based therapies.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/metabolismo , Isquemia/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Proteínas PrPC/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Membro Posterior/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mioblastos/citologia , Mioblastos/metabolismo , Mapas de Interação de Proteínas
18.
Anticancer Res ; 38(4): 1993-2000, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29599315

RESUMO

BACKGROUND/AIM: Drug resistance restricts the efficacy of chemotherapy in colorectal cancer. However, the detailed molecular mechanism of drug resistance in colorectal cancer cells remains unclear. MATERIALS AND METHODS: The level of cellular prion protein (PrPC) in oxaliplatin-resistant colorectal cancer (SNU-C5/Oxal-R) cells was assessed. RESULTS: PrPC level in SNU-C5/Oxal-R cells was significantly increased compared to that in wild-type (SNU-C5) cells. Superoxide dismutase and catalase activities were higher in SNU-C5/Oxal-R cells than in SNU-C5 cells. Treatment of SNU-C5/Oxal-R cells with oxaliplatin and melatonin reduced PrPC expression, while suppressing antioxidant enzyme activity and increasing superoxide anion generation. In SNU-C5/Oxal-R cells, endoplasmic reticulum stress and apoptosis were significantly increased following co-treatment with oxaliplatin and melatonin compared to treatment with oxaliplatin alone. CONCLUSION: Co-treatment with oxaliplatin and melatonin increased endoplasmic reticulum stress in and apoptosis of SNU-C5/Oxal-R cells through inhibition of PrPC, suggesting that PrPC could be a key molecule in oxaliplatin resistance of colorectal cancer cells.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Melatonina/farmacologia , Compostos Organoplatínicos/farmacologia , Proteínas PrPC/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Melatonina/administração & dosagem , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Proteínas PrPC/metabolismo , Superóxidos/metabolismo
19.
Int J Mol Sci ; 19(2)2018 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-29370069

RESUMO

Mesenchymal stem cells (MSCs) could be a promising solution in the treatment of various diseases including chronic kidney disease (CKD). However, endoplasmic reticulum (ER) stress induced by ischemia in the area of application limits the integration and survival of MSCs in patients. In our study, we generated ER stress-induced conditions in MSCs using P-cresol. As P-cresol is a toxic compound accumulated in the body of CKD patients and induces apoptosis and inflammation through reactive oxygen species (ROS), we observed ER stress-induced MSC apoptosis activated by oxidative stress, which in turn resulted from ROS generation. To overcome stress-induced apoptosis, we investigated the protective effects of tauroursodeoxycholic acid (TUDCA), a bile acid, on ER stress in MSCs. In ER stress, TUDCA treatment of MSCs reduced ER stress-associated protein activation, including GRP78, PERK, eIF2α, ATF4, IRE1α, and CHOP. Next, to explore the protective mechanism adopted by TUDCA, TUDCA-mediated cellular prion protein (PrPC) activation was assessed. We confirmed that PrPC expression significantly increased ROS, which was eliminated by superoxide dismutase and catalase in MSCs. These findings suggest that TUDCA protects from inflammation and apoptosis in ER stress via PrPC expression. Our study demonstrates that TUDCA protects MSCs against inflammation and apoptosis in ER stress by PrPC expression in response to P-cresol exposure.


Assuntos
Antioxidantes/farmacologia , Células-Tronco Mesenquimais/metabolismo , Proteínas PrPC/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Tecido Adiposo/citologia , Apoptose , Células Cultivadas , Cresóis/toxicidade , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Estresse Oxidativo , Proteínas PrPC/genética
20.
Biomol Ther (Seoul) ; 26(4): 389-398, 2018 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28655071

RESUMO

p-Cresol, found at high concentrations in the serum of chronic kidney failure patients, is known to cause cell senescence and other complications in different parts of the body. p-Cresol is thought to mediate cytotoxic effects through the induction of autophagy response. However, toxic effects of p-cresol on mesenchymal stem cells have not been elucidated. Thus, we aimed to investigate whether p-cresol induces senescence of mesenchymal stem cells, and whether melatonin can ameliorate abnormal autophagy response caused by p-cresol. We found that p-cresol concentration-dependently reduced proliferation of mesenchymal stem cells. Pretreatment with melatonin prevented pro-senescence effects of p-cresol on mesenchymal stem cells. We found that by inducing phosphorylation of Akt and activating the Akt signaling pathway, melatonin enhanced catalase activity and thereby inhibited the accumulation of reactive oxygen species induced by p-cresol in mesenchymal stem cells, ultimately preventing abnormal activation of autophagy. Furthermore, preincubation with melatonin counteracted other pro-senescence changes caused by p-cresol, such as the increase in total 5'-AMP-activated protein kinase expression and decrease in the level of phosphorylated mechanistic target of rapamycin. Ultimately, we discovered that melatonin restored the expression of senescence marker protein 30, which is normally suppressed because of the induction of the autophagy pathway in chronic kidney failure patients by p-cresol. Our findings suggest that stem cell senescence in patients with chronic kidney failure could be potentially rescued by the administration of melatonin, which grants this hormone a novel therapeutic role.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA