Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Cytotherapy ; 25(12): 1317-1330, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37804283

RESUMO

BACKGROUND AIMS: Cell failure and angiogenesis are the key to bladder wall regeneration. Three-dimensional (3D) culture using porous gelatin microspheres (GMs) as a vehicle promotes stem cell proliferation and improves the paracrine capacity of cells. This study aimed to evaluate the therapeutic potential of GMs constructed from adipose-derived mesenchymal stromal cells (ADSCs) (ADSC-GMs) combined with bladder acellular matrix (BAM) in tissue-engineered bladders. METHODS: Isolation of ADSCs, flow cytometry, scanning electron microscopy and cell counting kit-8, ß-galactosidase and enzyme-linked immunosorbent assays were performed in vitro to compare two-dimensional (2D) and 3D cultures. In the in vivo study, male Sprague-Dawley rats were randomly divided into three groups: the BAM replacement alone (BAM) group, ADSCs grown on BAM in replacement (ADSC) group and ADSC-GMs combined with BAM followed by replacement (ADSC-GM) group. Bladder function assessed by urodynamics after 12 weeks of bladder replacement, and the rats were sacrificed at 4 and 12 weeks for further experiments. RESULTS: The in vitro results showed that GM culture promoted ADSC proliferation, inhibited apoptosis and delayed senescence compared with those in the 2D culture. In addition, ADSC-GMs increased the secretion of the angiogenic factors vascular endothelial growth factor, platelet-derived growth factor-BB, and basal fibroblast growth factor. In vivo experiments revealed that ADSC-GMs adhered to the BAM for longer than ADSCs. Moreover, ADSC-GMs significantly promoted the regeneration of bladder vessels and smooth muscle, thereby facilitating the recovery of bladder function. The expression of phosphorylated protein kinase B (AKT) and phosphorylated endothelial nitric oxide synthase (eNOS) was significantly greater in the ADSC-GMs group compared with the BAM and ADSCs groups. CONCLUSIONS: ADSC-GMs increased retention of ADSCs on the BAM, thereby promoting the regeneration and functional recovery of the bladder tissue. ADSC-GMs promoted angiogenesis by activating the AKT/eNOS pathway.


Assuntos
Células-Tronco Mesenquimais , Bexiga Urinária , Ratos , Masculino , Animais , Bexiga Urinária/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Gelatina/metabolismo , Tecido Adiposo , Ratos Sprague-Dawley , Microesferas , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Porosidade , Transdução de Sinais
2.
Arch Toxicol ; 97(12): 3209-3226, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37798514

RESUMO

Administration of CHK1-targeted anticancer therapies is associated with an increased cumulative risk of cardiac complications, which is further amplified when combined with gemcitabine. However, the underlying mechanisms remain elusive. In this study, we generated hiPSC-CMs and murine models to elucidate the mechanisms underlying CHK1 inhibition combined with gemcitabine-induced cardiotoxicity and identify potential targets for cardioprotection. Mice were intraperitoneally injected with 25 mg/kg CHK1 inhibitor AZD7762 and 20 mg/kg gemcitabine for 3 weeks. hiPSC-CMs and NMCMs were incubated with 0.5 uM AZD7762 and 0.1 uM gemcitabine for 24 h. Both pharmacological inhibition or genetic deletion of CHK1 and administration of gemcitabine induced mtROS overproduction and pyroptosis in cardiomyocytes by disrupting mitochondrial respiration, ultimately causing heart atrophy and cardiac dysfunction in mice. These toxic effects were further exacerbated with combination administration. Using mitochondria-targeting sequence-directed vectors to overexpress CHK1 in cardiomyocyte (CM) mitochondria, we identified the localization of CHK1 in CM mitochondria and its crucial role in maintaining mitochondrial redox homeostasis for the first time. Mitochondrial CHK1 function loss mediated the cardiotoxicity induced by AZD7762 and CHK1-knockout. Mechanistically, mitochondrial CHK1 directly phosphorylates SIRT3 and promotes its expression within mitochondria. On the contrary, both AZD7762 or CHK1-knockout and gemcitabine decreased mitochondrial SIRT3 abundance, thus resulting in respiration dysfunction. Further hiPSC-CMs and mice experiments demonstrated that SIRT3 overexpression maintained mitochondrial function while alleviating CM pyroptosis, and thereby improving mice cardiac function. In summary, our results suggest that targeting SIRT3 could represent a novel therapeutic approach for clinical prevention and treatment of cardiotoxicity induced by CHK1 inhibition and gemcitabine.


Assuntos
Quinase 1 do Ponto de Checagem , Células-Tronco Pluripotentes Induzidas , Sirtuína 3 , Animais , Camundongos , Cardiotoxicidade/metabolismo , Gencitabina , Homeostase , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos , Oxirredução , Sirtuína 3/genética , Quinase 1 do Ponto de Checagem/metabolismo
3.
J Mol Cell Cardiol ; 166: 91-106, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35235835

RESUMO

Adult mammals have limited potential for cardiac regeneration after injury. In contrast, neonatal mouse heart, up to 7 days post birth, can completely regenerate after injury. Therefore, identifying the key factors promoting the proliferation of endogenous cardiomyocytes (CMs) is a critical step in the development of cardiac regeneration therapies. In our previous study, we predicted that mitogen-activated protein kinase (MAPK) interacting serine/threonine-protein kinase 2 (MNK2) has the potential of promoting regeneration by using phosphoproteomics and iGPS algorithm. Here, we aimed to clarify the role of MNK2 in cardiac regeneration and explore the underlying mechanism. In vitro, MNK2 overexpression promoted, and MNK2 knockdown suppressed cardiomyocyte proliferation. In vivo, inhibition of MNK2 in CMs impaired myocardial regeneration in neonatal mice. In adult myocardial infarcted mice, MNK2 overexpression in CMs in the infarct border zone activated cardiomyocyte proliferation and improved cardiac repair. In CMs, MNK2 binded to eIF4E and regulated its phosphorylation level. Knockdown of eukaryotic translation initiation factor (eIF4E) impaired the proliferation-promoting effect of MNK2 in CMs. MNK2-eIF4E axis stimulated CMs proliferation by activating cyclin D1. Our study demonstrated that MNK2 kinase played a critical role in cardiac regeneration. Over-expression of MNK2 promoted cardiomyocyte proliferation in vitro and in vivo, at least partly, by activating the eIF4E-cyclin D1 axis. This investigation identified a novel target for heart regenerative therapy.


Assuntos
Fator de Iniciação 4E em Eucariotos , Infarto do Miocárdio , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Ciclina D1/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Mamíferos/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fosforilação
4.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32761803

RESUMO

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Assuntos
Células Progenitoras Endoteliais/fisiologia , Tratamento por Ondas de Choque Extracorpóreas , Rim/irrigação sanguínea , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Movimento Celular , Quimiocina CXCL12/biossíntese , Quimiocina CXCL12/genética , Quimiocina CXCL12/fisiologia , Tratamento por Ondas de Choque Extracorpóreas/métodos , Corantes Fluorescentes/farmacocinética , Marcação In Situ das Extremidades Cortadas , Rim/patologia , Rim/fisiologia , Masculino , Microscopia de Fluorescência , Microvasos/patologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores CXCR/antagonistas & inibidores , Receptores CXCR/biossíntese , Receptores CXCR/genética , Receptores CXCR/fisiologia , Regeneração , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Fatores de Tempo
5.
Med Sci Monit ; 26: e919185, 2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32024811

RESUMO

BACKGROUND The present study was conducted to explore the influence of remote ischemic preconditioning (RIPC) on the adjustment of renal fibrosis after ischemia-reperfusion injury (IRI). MATERIAL AND METHODS Male Sprague-Dawley rats were randomly assigned to 3 groups following right-side nephrectomy: the Sham group (without renal artery clamping), the IRI group (45 min left renal artery clamping), and the RIPC group (rats were treated daily with 3 cycles of 5 min of limb ischemia and 5 min of reperfusion on 3 consecutive days before left renal artery occlusion). After 3 months of reperfusion, the renal function and the extent of tubular injury and renal fibrosis were assessed. The expressions of transforming growth factor beta1 (TGF-ß1), p-Smad2, Smad2, p-Smad3, and Smad3 were also evaluated. RESULTS There was no significant difference in renal function and tubular damage among the 3 groups after 45 min of kidney ischemia followed by 3 months of reperfusion. However, an obvious increase of extracellular matrix components and alpha-SMA could be observed in the kidney tissues of the IRI group, and the changes were significantly ameliorated in rats treated with enhanced RIPC. Compared with the IRI group, the expression of TGF-ß1 and the level of p-Smad2 and p-Smad3 were decreased after the intervention of enhanced RIPC. CONCLUSIONS Enhanced RIPC ameliorated renal fibrosis after IRI in rats, which appears to be associated with inhibition of the TGF-ß1/p-Smad2/3 signalling pathway.


Assuntos
Precondicionamento Isquêmico , Rim/irrigação sanguínea , Rim/patologia , Traumatismo por Reperfusão/complicações , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Actinas/metabolismo , Animais , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Fibronectinas/metabolismo , Fibrose , Rim/fisiopatologia , Masculino , Fosforilação , Ratos Sprague-Dawley , Traumatismo por Reperfusão/fisiopatologia , Proteínas Smad/metabolismo
6.
Int Braz J Urol ; 46(5): 691-704, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31961625

RESUMO

BACKGROUND: The diagnostic value and suitability of prostate cancer antigen 3 (PCA3) for the detection of prostate cancer (PCa) have been inconsistent in previous studies. Thus, the aim of the present meta-analysis was performed to systematically evaluate the diagnostic value of PCA3 for PCa. MATERIALS AND METHODS: A meta-analysis was performed to search relevant studies using online databases EMBASE, PubMed and Web of Science published until February 1st, 2019. Ultimately, 65 studies met the inclusion criteria for this meta-analysis with 8.139 cases and 14.116 controls. The sensitivity, specificity, positive likelihood ratios (LR+), negative likelihood ratios (LR-), and other measures of PCA3 were pooled and determined to evaluate the diagnostic rate of PCa by the random-effect model. RESULTS: With PCA3, the pooled overall diagnostic sensitivity, specificity, LR+, LR-, and 95% confidence intervals (CIs) for predicting significant PCa were 0.68 (0.64-0.72), 0.72 (0.68-0.75), 2.41 (2.16-2.69), 0.44 (0.40-0.49), respectively. Besides, the summary diagnostic odds ratio (DOR) and 95% CIs for PCA3 was 5.44 (4.53-6.53). In addition, the area under summary receiver operating characteristic (sROC) curves and 95% CIs was 0.76 (0.72-0.79). The major design deficiencies of included studies were differential verification bias, and a lack of clear inclusion and exclusion criteria. CONCLUSIONS: The results of this meta-analysis suggested that PCA3 was a non-invasive method with the acceptable sensitivity and specificity in the diagnosis of PCa, to distinguish between patients and healthy individuals. To validate the potential applicability of PCA3 in the diagnosis of PCa, more rigorous studies were needed to confirm these conclusions.


Assuntos
Neoplasias da Próstata , Antígenos de Neoplasias , Biomarcadores Tumorais , Humanos , Masculino , Razão de Chances , Neoplasias da Próstata/diagnóstico , Curva ROC , Sensibilidade e Especificidade
7.
J Cell Physiol ; 234(4): 4910-4923, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30317571

RESUMO

MicroRNAs (miRNAs), a group of small noncoding RNAs, are widely involved in the regulation of gene expression via binding to complementary sequences at 3'-untranslated regions (3'-UTRs) of target messenger RNAs. Recently, downregulation of miR-133b has been detected in various human malignancies. Here, the potential biological role of miR-133b in bladder cancer (BC) was investigated. In this study, we found the expression of miR-133b was markedly downregulated in BC tissues and cell lines (5637 and T24), and was correlated with poor overall survival. Notably, transgelin 2 (TAGLN2) was found to be widely upregulated in BC, and overexpression of TAGLN2 also significantly increased risks of advanced TMN stage. We further identified that upregulation of miR-133b inhibited glucose uptake, invasion, angiogenesis, colony formation and enhances gemcitabine chemosensitivity in BC cell lines by targeting TAGLN2. Additionally, we showed that miR-133b promoted the proliferation of BC cells, at least partially through a TAGLN2-mediated cell cycle pathway. Our results suggest a novel miR-133b/TAGLN2/cell cycle pathway axis controlling BC progression; a molecular mechanism which may offer a potential therapeutic target.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , MicroRNAs/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Neovascularização Patológica/genética , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/prevenção & controle , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Humanos , Camundongos , Camundongos Nus , MicroRNAs/biossíntese , Invasividade Neoplásica/genética , Transplante de Neoplasias , Transplante Heterólogo
8.
Cell Physiol Biochem ; 44(3): 1213-1223, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29179219

RESUMO

BACKGROUND/AIMS: Acute rejection (AR) is a major complication post renal transplantation, with no widely-accepted non-invasive biomarker. This study aimed to explore the expression profiles of long non-coding RNAs (lncRNAs) in the peripheral blood (PB) of renal transplant recipients and their potential diagnostic values. METHODS: The genome-wide lncRNA expression profiles were analyzed in 150 PB samples from pediatric and adult renal transplant (PRTx and ARTx) cohorts. The diagnostic performance of differentially expressed lncRNA was determined using receiver operator characteristic curve, with area under the curve (AUC) and 95% confidential interval (CI). Finally, a risk score was constructed with logistical regression model. RESULTS: A total of 162 lncRNAs were found differentially expressed in PRTx cohort, while 163 in ARTx cohort. Among these identified lncRNAs, 23 deregulated accordingly in both cohorts, and could distinguish AR recipients from those without AR. Finally, a risk score with two most significant lncRNAs (AF264622 and AB209021) was generated and exhibited excellent diagnostic performance in both PRTx (AUC:0.829, 95% CI:0.735-0.922) and ARTx cohorts (AUC: 0.889, 95% CI: 0.817-0.960). CONCLUSION: A molecular signature of two lncRNAs in PB could serve as a novel non-invasive biomarker for the diagnosis of AR in both pediatric and adult renal transplant recipients.


Assuntos
Rejeição de Enxerto/patologia , Transplante de Rim , RNA Longo não Codificante/sangue , Doença Aguda , Área Sob a Curva , Biomarcadores/sangue , Estudos de Coortes , Rejeição de Enxerto/genética , Rejeição de Enxerto/metabolismo , Humanos , Curva ROC , Transcriptoma , Transplante Homólogo
9.
Zhonghua Nan Ke Xue ; 22(7): 579-583, 2016 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-28965372

RESUMO

OBJECTIVE: To investigate the feasibility and practicability of establishing an animal model of primary premature ejaculation using the ejaculation distribution theory. METHODS: We induced behavioral estrus in 32 ovariectomized female SD rats by subcutaneous injection of 20 µg estradiol benzoate at 48 hours and 500 µg progesterone at 4 hours before mating them with 49 male rats once a week for six times. During the last three opulations, we observed the male animals for mounting latency (ML), intromission latency (IL), ejaculation latency (EL), postejaculation interval (PEI), mounting frequency (MF), intromission frequency (IF), intromission rate (IR), and ejaculation frequency (EF). RESULTS: Finally, 22 of the male rats were included in this study. The mean EF>33 was deemed rapid ejaculation,EF<1 sluggish ejaculation, and EF 1.5-2.5 normal ejaculation. The EL was significantly shorter in the rapid ejaculation group than in the sluggish and normal ejaculation groups. The IF was the lowest in those with rapid ejaculation. No statistically significant differences were observed in the ML among the three groups of rats. CONCLUSIONS: Based on the mean ejaculation frequency, the male rats with rapid ejaculation were easily screened, and this animal model may play an important role in exploring the mechanisms of primary premature ejaculation.


Assuntos
Modelos Animais de Doenças , Ejaculação Precoce/fisiopatologia , Animais , Ejaculação , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal
10.
Zhonghua Yi Xue Za Zhi ; 95(16): 1248-52, 2015 Apr 28.
Artigo em Chinês | MEDLINE | ID: mdl-26081512

RESUMO

OBJECTIVE: To explore the pneumoperitoneum-mediated renoprotective effects of preconditioning, mobilizing and homing of endothelial progenitor cells (EPCs) in rats. METHODS: A total of 40 rats were randomized by a numerical table into 5 groups of gasless (C), pneumoperitoneum injury (Pp), long-term pneumoperitoneum preconditioning (P-L), mid-term pneumoperitoneum preconditioning (P-M) and short-term pneumoperitoneum preconditioning (P-S). C group had a pneumoperitoneum pressure of 0 mmHg; Pp group 15 mmHg, time 60 min; P-L, P-M, P-S groups were deflated and deflated preconditioning before pneumoperitoneum, then the same as Pp group, P-L group: inflation time was 25 min, gas discharge time 10 min; P-M group: 15 min, 10 min; P-S group: 5 min, 10 min. At 24 h post-operation, the animals were sacrificed by destroying cervical spine. And the specimens of venous blood and kidneys were harvested. Also the extent of renal injury, the homing of EPCs, the proliferation and angiogenesis of renal endothelial cell and the expression of angiogenic growth factor were analyzed. RESULTS: Compared with Pp group, P-L, P-M and P-S groups exhibited significant improvements in renal function, morphology and histological score (1.88 ± 0.35, 1.63 ± 0.52, 1.75 ± 0.46 vs 2.38 ± 0.52, all P < 0.05). The histological scores of P-M and P-S groups improved significantly versus P-L group (both P < 0.05). P-M and P-S groups showed no significant difference in histological score (P > 0.05). The number of EPCs in kidneys increased in P-L, P-M and P-S groups versus Pp group (2.18% ± 0.14%, 2.87% ± 0.29%, 2.90% ± 0.24% vs 1.73% ± 0.19%, all P < 0.05). The EPCs numbers of P-M and P-S groups were more than that of P-L group (both P < 0.05). And no significant difference existed between P-M and P-S groups (P > 0.05). Compared with Pp group, EPCs of P-L, P-M and P-S groups markedly increased in kidneys. No significant difference existed between P-M and P-S groups, but P-L group was the lowest. Also there was an up-regulated expression of stromal cell derived factor 1-α in pretreated kidneys versus Pp group (all P < 0.05). And P-M and P-S groups increased markedly. CONCLUSIONS: Pneumoperitoneum-mediated preconditioning protects against kidney injury by promoting EPC homing and enhancing endothelial cell and vascular proliferations. And short and medium-term preconditioning protocols are more effective for protecting kidneys.


Assuntos
Células Progenitoras Endoteliais , Rim , Pneumoperitônio , Animais , Quimiocina CXCL12 , Ratos , Ratos Sprague-Dawley
11.
Tumour Biol ; 35(9): 9331-41, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24943683

RESUMO

Neovascularization is a key role of renal cell carcinoma (RCC) and the status of neovascularization in RCC is closely correlated with the tumor development and patient prognosis. Endothelial progenitor cells (EPCs) are considered as important building blocks for neovascularization. However, the role of mobilized EPCs in RCC remains unknown. In this study, the orthotopic RCC model was established to investigate the distribution, frequency, and significance of mobilized EPCs. We found that circulating endothelial progenitor cell (CEPC) levels and plasma angiogenic factors (vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) were higher in peripheral blood (PB) of the RCC than those in the normal group and positively correlated with each other. EPC levels in adjacent nonmalignant kidney tissue (AT) were significantly higher than those in tumor tissue (TT) and normal kidney tissue (NT), which were positively correlated with CEPC levels. VEGF, VEGF receptor-2 (Flk), and SDF-1 and its SDF-1 receptor (CXCR4) expression in AT was significantly higher than that in TT and NT. Levels of these angiogenic factors in AT were positively correlated with those in PB. Mean microvessel density (MVD) was higher in AT than in TT, and that in TT was slightly lower than that in NT. Our findings propose that mobilized EPCs play an important role in RCC neovascularization. EPCs in PB and AT can be used as a biomarker for predicting RCC progression.


Assuntos
Carcinoma de Células Renais/metabolismo , Células Progenitoras Endoteliais/metabolismo , Neoplasias Hepáticas/metabolismo , Neovascularização Patológica/metabolismo , Animais , Antígenos CD34/metabolismo , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/sangue , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Rim/irrigação sanguínea , Rim/metabolismo , Rim/patologia , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/genética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Confocal , Neovascularização Patológica/sangue , Neovascularização Patológica/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
J Sex Med ; 11(11): 2801-8, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25130949

RESUMO

INTRODUCTION: The pathologic mechanisms of primary premature ejaculation (PPE) are complex and multifactorial, and hyperactivity of the sympathetic nervous system is one of the mechanisms. AIM: To examine the effects of sertraline on sympathetic nervous system activity and assess the predictive value of the sympathetic skin response located in the penis (PSSR) on the response to sertraline treatment in PPE patients. METHODS: Sixty-one patients with PPE were recruited. Each received 50 mg sertraline daily for 8 weeks. Before and after the experiment, the patients were evaluated for PSSR tests and sexual performance parameters. Additionally, based on the latency of PSSR, we divided the patients into a normal PSSR group and an abnormal PSSR group, and compared the sertraline treatment efficacy between the two groups. MAIN OUTCOME MEASURES: Changes in intravaginal ejaculation latency time (IELT) and the Chinese premature ejaculation index-5 (CIPE-5), and the latencies and amplitudes of PSSR after sertraline treatment. RESULTS: Overall, 58 (95.1%) patients completed the entire study and were analyzed. After the 8-week sertraline treatment, compared with those of pretreatment, IELT and CIPE-5 scores were significantly increased (both P < 0.001), and the amplitudes and latencies of PSSR in the PPE patients were remarkably decreased and prolonged, respectively (both P < 0.001). In addition, the changes of the latencies of PSSR were positively correlated with the increment of IELT (r = 0.375, P = 0.004). The treatment outcome was better in patients with a baseline abnormal PSSR than in those with a baseline normal PSSR (P = 0.021). CONCLUSIONS: These results suggest that clinical improvement in response to sertraline in the PPE patients, at least in part, is mediated through reducing sympathetic nervous system activity indexed by PSSR. Measurement of the PSSR appears to provide useful information for predicting treatment responses in the PPE patients.


Assuntos
Pênis/inervação , Ejaculação Precoce/tratamento farmacológico , Sertralina/uso terapêutico , Pele/inervação , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pênis/fisiopatologia , Ejaculação Precoce/fisiopatologia , Pele/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Resultado do Tratamento
13.
J Surg Res ; 186(1): 63-72, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24095026

RESUMO

BACKGROUND: To compare the effects of various recombinant growth factors on bladder regeneration and angiogenesis for tissue engineering of bladder in patients with neurogenic bladder through in vitro cellular biological methods. MATERIALS AND METHODS: Human bladder smooth muscle cells (HBSMCs) and human bladder urothelial cells (HBUCs) were cultured from patients with neurogenic bladder and used for comparative evaluations of various growth factors. Human umbilical vein endothelial cells (HUVECs) were also used. Eight potential growth factors, platelet-derived growth factor BB (PDGF-BB), platelet-derived growth factor CC (PDGF-CC), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), insulin-like growth factor 1 (IGF-1), hepatocyte growth factor (HGF), and transforming growth factor beta 1 (TGF-ß1), were selected and their effects on the proliferation, migration, and wound healing of HBSMCs, HBUCs, and HUVECs were compared. RESULTS: PDGF-BB, PDGF-CC, bFGF, VEGF, IGF-1, or HGF enhanced the proliferation, migration, and wound healing of HBSMCs, whereas TGF-ß1 inhibited their proliferation. Proliferation, migration, and wound healing of HBUCs and HUVECs were enhanced by bFGF, VEGF, EGF, IGF-1, or HGF, whereas inhibited by TGF-ß1. PDGF-BB failed to enhance cell activity of HUVECs, whereas PDGF-CC could enhance their migration and wound healing. PDGF-BB, EGF, and VEGF were the most potent factors for stimulating the activities of HBSMCs, HBUCs, and HUVECs, respectively. CONCLUSIONS: Our findings suggest the potential use of a combination of PDGF-BB, EGF, and VEGF for bladder regeneration and angiogenesis. The synergetic effects of the three growth factors on cell activities in a three-dimensional scaffold and an animal model with neurogenic bladder need to be further evaluated.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Engenharia Tecidual/métodos , Bexiga Urinaria Neurogênica/terapia , Bexiga Urinária/efeitos dos fármacos , Idoso , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Recombinantes/uso terapêutico , Bexiga Urinária/citologia , Cicatrização/efeitos dos fármacos
14.
Tissue Eng Part C Methods ; 30(1): 15-26, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37756374

RESUMO

Insufficient vascularization is still a challenge that impedes bladder tissue engineering and results in unsatisfied smooth muscle regeneration. Since bladder regeneration is a complex articulated process, the aim of this study is to investigate whether combining multiple pathways by exploiting a combination of biomaterials, cells, and bioactive factors, contributes to the improvements of smooth muscle regeneration and vascularization in tissue-engineered bladder. Autologous endothelial progenitor cells (EPCs) and bladder smooth muscle cells (BSMCs) are cultured and incorporated into our previously prepared porcine bladder acellular matrix (BAM) for bladder augmentation in rabbits. Simultaneously, exogenous vascular endothelial growth factor (VEGF) and platelet-derived growth factor BB (PDGF-BB) mixed with Matrigel were injected around the implanted cells-BAM complex. In the results, compared with control rabbits received bladder augmentation with porcine BAM seeded with BSMCs, the experimental animals showed significantly improved smooth muscle regeneration and vascularization, along with more excellent functional recovery of tissue-engineered bladder, due to the additional combination of autologous EPCs and bioactive factors, including VEGF and PDGF-BB. Furthermore, cell tracking suggested that the seeded EPCs could be directly involved in neovascularization. Therefore, it may be an effective method to combine multiple pathways for tissue-engineering urinary bladder.


Assuntos
Células Progenitoras Endoteliais , Bexiga Urinária , Suínos , Coelhos , Animais , Bexiga Urinária/irrigação sanguínea , Bexiga Urinária/metabolismo , Células Progenitoras Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Becaplermina/farmacologia , Becaplermina/metabolismo , Engenharia Tecidual/métodos , Regeneração
15.
Mater Today Bio ; 25: 101012, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38464495

RESUMO

Urethral stricture (US) is a common disease in urology, lacking effective treatment options. Although injecting a stem cells suspension into the affected area has shown therapeutic benefits, challenges such as low retention rate and limited efficacy hinder the clinical application of stem cells. This study evaluates the therapeutic impact and the mechanism of adipose-derived vascular fraction (SVF) combined with cell sheet engineering technique on urethral fibrosis in a rat model of US. The results showed that SVF-cell sheets exhibit positive expression of α-SMA, CD31, CD34, Stro-1, and eNOS. In vivo study showed less collagen deposition, low urethral fibrosis, and minimal tissue alteration in the group receiving cell sheet transplantation. Furthermore, the formation of a three-dimensional (3D) tissue-like structure by the cell sheets enhances the paracrine effect of SVF, facilitates the infiltration of M2 macrophages, and suppresses the TGF-ß/Smad2 pathway through HGF secretion, thereby exerting antifibrotic effects. Small animal in vivo imaging demonstrates improved retention of SVF cells at the damaged urethra site with cell sheet application. Our results suggest that SVF combined with cell sheet technology more efficiently inhibits the early stages of urethral fibrosis.

16.
Eur J Pharmacol ; 983: 177005, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39299480

RESUMO

Restoration of the expression of factors regulating neonatal heart regeneration in the adult heart can promote myocardial repair. Therefore, investigations of the regulatory factors that play key roles in neonatal heart regeneration are urgently needed for the development of cardiac regenerative therapies. In our previous study, we identified ankyrin repeat domain 1 (Ankrd1) through multiomics analysis in a neonatal mouse model of cardiac regeneration and hypothesized that Ankrd1 plays a regulatory role in neonatal heart regeneration. In the present study, we aimed to determine the role of Ankrd1 in neonatal heart regeneration and adult myocardial repair. Our findings confirmed that Ankrd1 could mediate cardiomyocyte proliferation and that Ankrd1 knockdown in cardiomyocytes inhibited myocardial regeneration after apical resection in neonatal mice. Furthermore, we found that cardiomyocyte-specific Ankrd1 overexpression promoted cardiac repair and cardiac function recovery after adult myocardial infarction (MI). Mechanistically, Ankrd1 could regulate the cell cycle of cardiomyocytes and significantly mediate cardiac regeneration, at least in part, through cyclin D1. Overall, our study demonstrates that Ankrd1 is an effective target for achieving cardiac repair after MI, providing new ideas for the treatment of ischemic heart disease in the future.

17.
J Control Release ; 370: 66-81, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631490

RESUMO

Renal ischemia-reperfusion injury (IRI) is one of the most important causes of acute kidney injury (AKI). Interleukin (IL)-37 has been suggested as a novel anti-inflammatory factor for the treatment of IRI, but its application is still limited by its low stability and delivery efficiency. In this study, we reported a novel engineered method to efficiently and easily prepare neutrophil membrane-derived vesicles (N-MVs), which could be utilized as a promising vehicle to deliver IL-37 and avoid the potential side effects of neutrophil-derived natural extracellular vesicles. N-MVs could enhance the stability of IL-37 and targetedly deliver IL-37 to damaged endothelial cells of IRI kidneys via P-selectin glycoprotein ligand-1 (PSGL-1). In vitro and in vivo evidence revealed that N-MVs encapsulated with IL-37 (N-MV@IL-37) could inhibit endothelial cell apoptosis, promote endothelial cell proliferation and angiogenesis, and decrease inflammatory factor production and leukocyte infiltration, thereby ameliorating renal IRI. Our study establishes a promising delivery vehicle for the treatment of renal IRI and other endothelial damage-related diseases.


Assuntos
Células Endoteliais , Interleucina-1 , Rim , Neutrófilos , Traumatismo por Reperfusão , Animais , Traumatismo por Reperfusão/tratamento farmacológico , Interleucina-1/administração & dosagem , Masculino , Humanos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Rim/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Camundongos Endogâmicos C57BL , Apoptose/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Glicoproteínas de Membrana/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Injúria Renal Aguda , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/química
18.
Research (Wash D C) ; 7: 0451, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39193132

RESUMO

The potential of circular RNAs (circRNAs) as biomarkers and therapeutic targets is becoming increasingly evident, yet their roles in cardiac regeneration and myocardial renewal remain largely unexplored. Here, we investigated the function of circIGF1R and related mechanisms in cardiac regeneration. Through analysis of circRNA sequencing data from neonatal and adult cardiomyocytes, circRNAs associated with regeneration were identified. Our data showed that circIGF1R expression was high in neonatal hearts, decreased with postnatal maturation, and up-regulated after cardiac injury. The elevation was validated in patients diagnosed with acute myocardial infarction (MI) within 1 week. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and myocardial tissue from mice after apical resection and MI, we observed that circIGF1R overexpression enhanced cardiomyocyte proliferation, reduced apoptosis, and mitigated cardiac dysfunction and fibrosis, while circIGF1R knockdown impeded endogenous cardiac renewal. Mechanistically, we identified circIGF1R binding proteins through circRNA precipitation followed by mass spectrometry. RNA pull-down Western blot and RNA immunoprecipitation demonstrated that circIGF1R directly interacted with DDX5 and augmented its protein level by suppressing ubiquitin-dependent degradation. This subsequently triggered the ß-catenin signaling pathway, leading to the transcriptional activation of cyclin D1 and c-Myc. The roles of circIGF1R and DDX5 in cardiac regeneration were further substantiated through site-directed mutagenesis and rescue experiments. In conclusion, our study highlights the pivotal role of circIGF1R in facilitating heart regeneration and repair after ischemic insults. The circIGF1R/DDX5/ß-catenin axis emerges as a novel therapeutic target for enhancing myocardial repair after MI, offering promising avenues for the development of regenerative therapies.

19.
Commun Biol ; 7(1): 795, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38951640

RESUMO

The peroxisome is a versatile organelle that performs diverse metabolic functions. PEX3, a critical regulator of the peroxisome, participates in various biological processes associated with the peroxisome. Whether PEX3 is involved in peroxisome-related redox homeostasis and myocardial regenerative repair remains elusive. We investigate that cardiomyocyte-specific PEX3 knockout (Pex3-KO) results in an imbalance of redox homeostasis and disrupts the endogenous proliferation/development at different times and spatial locations. Using Pex3-KO mice and myocardium-targeted intervention approaches, the effects of PEX3 on myocardial regenerative repair during both physiological and pathological stages are explored. Mechanistically, lipid metabolomics reveals that PEX3 promotes myocardial regenerative repair by affecting plasmalogen metabolism. Further, we find that PEX3-regulated plasmalogen activates the AKT/GSK3ß signaling pathway via the plasma membrane localization of ITGB3. Our study indicates that PEX3 may represent a novel therapeutic target for myocardial regenerative repair following injury.


Assuntos
Membrana Celular , Integrina beta3 , Camundongos Knockout , Regeneração , Animais , Masculino , Camundongos , Membrana Celular/metabolismo , Proliferação de Células , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Traumatismos Cardíacos/genética , Integrina beta3/metabolismo , Integrina beta3/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Plasmalogênios/metabolismo , Transdução de Sinais
20.
J Am Heart Assoc ; 13(13): e034805, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38934866

RESUMO

BACKGROUND: The regenerative capacity of the adult mammalian hearts is limited. Numerous studies have explored mechanisms of adult cardiomyocyte cell-cycle withdrawal. This translational study evaluated the effects and underlying mechanism of rhCHK1 (recombinant human checkpoint kinase 1) on the survival and proliferation of cardiomyocyte and myocardial repair after ischemia/reperfusion injury in swine. METHODS AND RESULTS: Intramyocardial injection of rhCHK1 protein (1 mg/kg) encapsulated in hydrogel stimulated cardiomyocyte proliferation and reduced cardiac inflammation response at 3 days after ischemia/reperfusion injury, improved cardiac function and attenuated ventricular remodeling, and reduced the infarct area at 28 days after ischemia/reperfusion injury. Mechanistically, multiomics sequencing analysis demonstrated enrichment of glycolysis and mTOR (mammalian target of rapamycin) pathways after rhCHK1 treatment. Co-Immunoprecipitation (Co-IP) experiments and protein docking prediction showed that CHK1 (checkpoint kinase 1) directly bound to and activated the Serine 37 (S37) and Tyrosine 105 (Y105) sites of PKM2 (pyruvate kinase isoform M2) to promote metabolic reprogramming. We further constructed plasmids that knocked out different CHK1 and PKM2 amino acid domains and transfected them into Human Embryonic Kidney 293T (HEK293T) cells for CO-IP experiments. Results showed that the 1-265 domain of CHK1 directly binds to the 157-400 amino acids of PKM2. Furthermore, hiPSC-CM (human iPS cell-derived cardiomyocyte) in vitro and in vivo experiments both demonstrated that CHK1 stimulated cardiomyocytes renewal and cardiac repair by activating PKM2 C-domain-mediated cardiac metabolic reprogramming. CONCLUSIONS: This study demonstrates that the 1-265 amino acid domain of CHK1 binds to the 157-400 domain of PKM2 and activates PKM2-mediated metabolic reprogramming to promote cardiomyocyte proliferation and myocardial repair after ischemia/reperfusion injury in adult pigs.


Assuntos
Proliferação de Células , Quinase 1 do Ponto de Checagem , Modelos Animais de Doenças , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Animais , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Quinase 1 do Ponto de Checagem/metabolismo , Quinase 1 do Ponto de Checagem/genética , Humanos , Piruvato Quinase/metabolismo , Piruvato Quinase/genética , Células HEK293 , Suínos , Reprogramação Celular , Proteínas de Ligação a Hormônio da Tireoide , Regeneração , Ligação Proteica , Sus scrofa , Remodelação Ventricular/fisiologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Metabolismo Energético/efeitos dos fármacos , Hormônios Tireóideos/metabolismo , Reprogramação Metabólica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa