Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Publication year range
1.
Asian J Androl ; 23(6): 640-647, 2021.
Article in English | MEDLINE | ID: mdl-34135173

ABSTRACT

To evaluate outcomes between extraperitoneal robotic single-port radical prostatectomy (epR-spRP) and extraperitoneal robotic multiport radical prostatectomy (epR-mpRP) performed with the da Vinci Si Surgical System, comparison was performed between 30 single-port (SP group) and 26 multiport (MP group) cases. Comparisons included operative time, estimated blood loss (EBL), hospital stay, peritoneal violation, pain scores, scar satisfaction, continence, and erectile function. The median operation time and EBL were not different between the two groups. In the SP group, the median operation time of the first 10 patients was obviously longer than that of the latter 20 patients (P < 0.001). The median postoperative hospital stay in the SP group was shorter than that in the MP group (P < 0.001). The rate of peritoneal damage in the SP group was less than that in the MP group (P = 0.017). The pain score and overall need for pain medications in the SP group were lower than those in the MP group (P < 0.001 and P = 0.015, respectively). Patients in the SP group were more satisfied with their scars than those in the MP group 3 months postoperatively (P = 0.007). At 3 months, the cancer control, recovery of erectile function, and urinary continence rates were similar between the two groups. It is safe and feasible to perform epR-spRP using the da Vinci Si surgical system. Therefore, epR-spRP can be a treatment option for localized prostate cancer. Although epR-spRP still has a learning curve, it has advantages for postoperative pain and self-assessed cosmesis. In the absence of the single-port robotic surgery platform, we can still provide minimally invasive surgery for patients.


Subject(s)
Outcome Assessment, Health Care/statistics & numerical data , Perioperative Medicine/instrumentation , Prostatectomy/instrumentation , Robotic Surgical Procedures/standards , Aged , Blood Loss, Surgical/statistics & numerical data , Humans , Male , Middle Aged , Outcome Assessment, Health Care/methods , Perioperative Medicine/standards , Perioperative Medicine/statistics & numerical data , Prostatectomy/methods , Prostatic Neoplasms/surgery , Quality Assurance, Health Care/statistics & numerical data , Robotic Surgical Procedures/methods , Robotic Surgical Procedures/statistics & numerical data
2.
Kidney Blood Press Res ; 45(1): 95-108, 2020.
Article in English | MEDLINE | ID: mdl-31927554

ABSTRACT

BACKGROUND/AIMS: Acute kidney injury (AKI) is a common clinical condition that can lead to chronic kidney failure. Although mesenchymal stem cell-derived extracellular vesicles (MSC EVs) are regarded as a potent AKI treatment, the mechanisms underlying their beneficial effects remain unclear. Oct-4 may play an important role in tissue injury repair. We thus hypothesized that oct-4 overexpression might enhance the therapeutic effects of MSC EVs in AKI treatment. METHODS: Renal tubular epithelial cells were cultured in a low oxygen environment, then cocultured with MSC EVs or control medium for 48 h. BrdU and transferase-mediated dUTP nick-end labeling (TUNEL) staining were used to assess cell proliferation and apoptosis. Mice subjected to ischemia reperfusion were randomly divided into 4 groups, then injected with either phosphate-buffered saline (vehicle), EVs, EVs overexpressing oct-4 (EVs+Oct-4), and EVs not expressing Oct-4 (EVs-Oct-4). Blood creatinine (CREA) and urine nitrone levels were assessed 48 h and 2 weeks after injection. After ischemia reperfusion, renal tissues from each group were stained with TUNEL and proliferating cell nuclear antigen (PCNA) to determine the degree of apoptosis and proliferation. Masson trichrome staining was used to evaluate renal fibrosis progression. Snail gene expression was assessed using polymerase chain reaction (PCR). RESULTS: At 48 h after hypoxic treatment, TUNEL and BrdU staining indicated that the EVs+Oct-4 group had the least apoptosis and the most proliferation, respectively. Treatment with EVs overexpressing Oct-4 significantly decreased serum Crea and blood urea nitrogen levels and rescued kidney fibrosis, as indicated by the low proportion of Masson staining, high number of PCNA-positive cells, and low number of TUNEL-positive cells. PCR analysis indicated that Snail was most upregulated in the vehicle group and least upregulated in the EVs+Oct-4 group. CONCLUSIONS: MSC EVs had a pronounced therapeutic effect on ischemic reperfusion injury-related AKI, and Oct-4 overexpression enhanced these therapeutic effects. Our results may inspire a new direction for AKI treatment with MSC EVs.


Subject(s)
Acute Kidney Injury/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Acute Kidney Injury/pathology , Animals , Disease Models, Animal , Extracellular Vesicles/metabolism , Humans , Immunohistochemistry , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/biosynthesis , Random Allocation
3.
Zhonghua Nan Ke Xue ; 24(1): 14-18, 2018.
Article in Chinese | MEDLINE | ID: mdl-30157354

ABSTRACT

OBJECTIVE: To investigate the mechanisms of stem cell-derived extracellular vesicles (EVs) alleviating acute oxidative stress injury to Leydig cells. METHODS: The model of testicular ischemia-reperfusion was established by unilateral testicular resection in 10 SD rats followed by injection of EVs into the tail vein in 5 of the animals (the experimental group) and that of isotonic PBS in the other 5(the control group).At 2 hours after reperfusion, the infiltration of neutrophilsin the testicular tissue was detected by HE staining, the apoptosis of Leydig cells measured by TUNEL and Caspase-3 immunohistochemistry, and the proliferation of Leydig cellsdetermined by Ki-67 immunohistochemical staining. RESULTS: Compared with the control, the experimental group showed significantlymilderinfiltration of neutrophils in the interstitial tissueof the testis and lower apoptosis and higher proliferation of Leydig cells. CONCLUSIONS: Stem cell EVs can reduce the adhesion of neutrophils in the acute phase and alleviate acute oxidative stress injury to Leydig cells by decreasing the production of reactive oxygen free radicals.


Subject(s)
Extracellular Vesicles/physiology , Leydig Cells/cytology , Neutrophils , Reperfusion Injury/prevention & control , Testis/blood supply , Testis/cytology , Acute Disease , Animals , Apoptosis , Caspase 3/analysis , Cell Movement , Cell Proliferation , Extracellular Vesicles/transplantation , In Situ Nick-End Labeling , Ki-67 Antigen/analysis , Leydig Cells/physiology , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley , Stem Cells/ultrastructure
4.
Asian J Androl ; 19(5): 543-547, 2017.
Article in English | MEDLINE | ID: mdl-27586027

ABSTRACT

Androgen deficiency is a physical disorder that not only affects adults but can also jeopardize children's health. Because there are many disadvantages to using traditional androgen replacement therapy, we have herein attempted to explore the use of human umbilical cord mesenchymal stem cells for the treatment of androgen deficiency. We transplanted CM-Dil-labeled human umbilical cord mesenchymal stem cells into the testes of an ethane dimethanesulfonate (EDS)-induced male rat hypogonadism model. Twenty-one days after transplantation, we found that blood testosterone levels in the therapy group were higher than that of the control group (P = 0.037), and using immunohistochemistry and flow cytometry, we observed that some of the CM-Dil-labeled cells expressed Leydig cell markers for cytochrome P450, family 11, subfamily A, polypeptide 1, and 3-ß-hydroxysteroid dehydrogenase. We then recovered these cells and observed that they were still able to proliferate in vitro. The present study shows that mesenchymal stem cells from human umbilical cord may constitute a promising therapeutic modality for the treatment of male hypogonadism patients.


Subject(s)
Eunuchism/complications , Eunuchism/therapy , Mesenchymal Stem Cell Transplantation/methods , Testicular Diseases/etiology , Testicular Diseases/therapy , Umbilical Cord/cytology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Eunuchism/chemically induced , Female , Humans , Leydig Cells/metabolism , Male , Mesylates , Rats , Rats, Sprague-Dawley , Testicular Diseases/chemically induced , Testosterone/blood
5.
Zhonghua Nan Ke Xue ; 22(8): 680-685, 2016 Aug.
Article in Chinese | MEDLINE | ID: mdl-29019222

ABSTRACT

OBJECTIVE: To explore the feasibility of inducing human umbilical cord mesenchymal stem cells (HUMSCs) to differentiate into Leydig cells in the interstitial tissue of the rat testis. METHODS: HUMSCs were obtained by tissue blocks culture attachment and their purity and multi-lineage differentiation ability were verified by flow cytometry and chondrogenic/adipogenic/osteogenic differentiation. Then the HUMSCs were marked by CM-Dil and transplanted into the interstitial tissue of the rat testis. At 4 and 8 weeks after transplantation, the survival and differentiation status of the HUMSCs were observed by immunofluorescence staining and flow cytometry. The suspension of the rat Leydig cells was obtained at 8 weeks for determining the expression of the Leydig cell marker 3ß-HSD in the HUMSCs, the cells labeled with CM-Dil were sorted and cultured, and the medium collected after 3 days of culture for measurement of the testosterone level. RESULTS: The expression of the Leydig cell marker CYPllal was not observed in the HUMSCs at 4 weeks but found at 8 weeks after transplantation and the differentiation rate of 3ß-HSD was about 14.5% at 8 weeks. CM-Dil labeled cells survived after sorting and testosterone was detected in the medium. CONCLUSIONS: HUMSCs are likely to differentiate into Leydig cells in the interstitium of the rat testis.


Subject(s)
Cell Differentiation , Leydig Cells/cytology , Mesenchymal Stem Cells/cytology , Testis/cytology , Umbilical Cord/cytology , Animals , Biomarkers/metabolism , Carbocyanines , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Feasibility Studies , Humans , Leydig Cells/metabolism , Male , Rats , Time Factors
6.
PLoS One ; 10(3): e0121534, 2015.
Article in English | MEDLINE | ID: mdl-25793303

ABSTRACT

During acute kidney injury (AKI), tubular cell dedifferentiation initiates cell regeneration; hepatocyte growth factor (HGF) is involved in modulating cell dedifferentiation. Mesenchymal stem cell (MSC)-derived microvesicles (MVs) deliver RNA into injured tubular cells and alter their gene expression, thus regenerating these cells. We boldly speculated that MVs might induce HGF synthesis via RNA transfer, thereby facilitating tubular cell dedifferentiation and regeneration. In a rat model of unilateral AKI, the administration of MVs promoted kidney recovery. One of the mechanisms of action is the acceleration of tubular cell dedifferentiation and growth. Both in vivo and in vitro, rat HGF expression in damaged rat tubular cells was greatly enhanced by MV treatment. In addition, human HGF mRNA present in MVs was delivered into rat tubular cells and translated into the HGF protein as another mechanism of HGF induction. RNase treatment abrogated all MV effects. In the in vitro experimental setting, the conditioned medium of MV-treated injured tubular cells, which contains a higher concentration of HGF, strongly stimulated cell dedifferentiation and growth, as well as Erk1/2 signaling activation. Intriguingly, these effects were completely abrogated by either c-Met inhibitor or MEK inhibitor, suggesting that HGF induction is a crucial contributor to the acceleration of cell dedifferentiation and growth. All these findings indicate that MV-induced HGF synthesis in damaged tubular cells via RNA transfer facilitates cell dedifferentiation and growth, which are important regenerative mechanisms.


Subject(s)
Cell Dedifferentiation , Cell-Derived Microparticles/metabolism , Epithelial Cells/cytology , Hepatocyte Growth Factor/metabolism , Kidney Tubules/cytology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Cell Dedifferentiation/drug effects , Cell Proliferation/drug effects , Cell-Derived Microparticles/drug effects , Culture Media, Conditioned/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hepatocyte Growth Factor/genetics , Humans , Hypoxia/pathology , Ischemia/pathology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxygen/metabolism , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
7.
PLoS One ; 8(4): e61366, 2013.
Article in English | MEDLINE | ID: mdl-23593475

ABSTRACT

Several studies suggest that mesenchymal stem cells (MSCs) possess antitumor properties; however, the exact mechanisms remain unclear. Recently, microvesicles (MVs) are considered as a novel avenue intercellular communication, which may be a mediator in MSCs-related antitumor effect. In the present study, we evaluated whether MVs derived from human umbilical cord Wharton's jelly mesenchymal stem cells (hWJMSCs) may inhibit bladder tumor T24 cells growth using cell culture and the BALB/c nu/nu mice xenograft model. CCK-8 assay and Ki-67 immunostaining were performed to estimate cell proliferation in vitro and in vivo. Flow cytometry and TUNEL assay were used to assess cell cycle and apoptosis. To study the conceivable mechanism by which hWJMSC-MVs attenuate bladder tumor T24 cells, we estimated the expression of Akt/p-Akt, p-p53, p21 and cleaved Caspase 3 by Western blot technique after exposing T24 cells to hWJMSC-MVs for 24, 48 and 72h. Our data indicated that hWJMSC-MVs can inhibit T24 cells proliferative viability via cell cycle arrest and induce apoptosis in T24 cells in vitro and in vivo. This study showed that hWJMSC-MVs down-regulated phosphorylation of Akt protein kinase and up-regulated cleaved Caspase 3 during the process of anti-proliferation and pro-apoptosis in T24 cells. These results demonstrate that hWJMSC-MVs play a vital role in hWJMSC-induced antitumor effect and may be a novel tool for cancer therapy as a new mechanism of cell-to-cell communication.


Subject(s)
Cytoplasmic Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/therapy , Wharton Jelly/cytology , Animals , Apoptosis , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Separation , Cell Shape , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoplasmic Vesicles/ultrastructure , Enzyme Activation , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Mice , Mice, Inbred BALB C , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...