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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Pediatr ; 24(1): 279, 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678251

RESUMO

BACKGROUND: Wilms' tumor (WT) is the most common renal tumor in childhood. Pyroptosis, a type of inflammation-characterized and immune-related programmed cell death, has been extensively studied in multiple tumors. In the current study, we aim to construct a pyroptosis-related gene signature for predicting the prognosis of Wilms' tumor. METHODS: We acquired RNA-seq data from TARGET kidney tumor projects for constructing a gene signature, and snRNA-seq data from GEO database for validating signature-constructing genes. Pyroptosis-related genes (PRGs) were collected from three online databases. We constructed the gene signature by Lasso Cox regression and then established a nomogram. Underlying mechanisms by which gene signature is related to overall survival states of patients were explored by immune cell infiltration analysis, differential expression analysis, and functional enrichment analysis. RESULTS: A pyroptosis-related gene signature was constructed with 14 PRGs, which has a moderate to high predicting capacity with 1-, 3-, and 5-year area under the curve (AUC) values of 0.78, 0.80, and 0.83, respectively. A prognosis-predicting nomogram was established by gender, stage, and risk score. Tumor-infiltrating immune cells were quantified by seven algorithms, and the expression of CD8( +) T cells, B cells, Th2 cells, dendritic cells, and type 2 macrophages are positively or negatively correlated with risk score. Two single nuclear RNA-seq samples of different histology were harnessed for validation. The distribution of signature genes was identified in various cell types. CONCLUSIONS: We have established a pyroptosis-related 14-gene signature in WT. Moreover, the inherent roles of immune cells (CD8( +) T cells, B cells, Th2 cells, dendritic cells, and type 2 macrophages), functions of differentially expressed genes (tissue/organ development and intercellular communication), and status of signaling pathways (proteoglycans in cancer, signaling pathways regulating pluripotent of stem cells, and Wnt signaling pathway) have been elucidated, which might be employed as therapeutic targets in the future.


Assuntos
Neoplasias Renais , Piroptose , Tumor de Wilms , Humanos , Piroptose/genética , Tumor de Wilms/genética , Tumor de Wilms/imunologia , Neoplasias Renais/genética , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Prognóstico , Nomogramas , Linfócitos do Interstício Tumoral/imunologia , Transcriptoma , Feminino , Masculino
2.
J Cell Physiol ; 234(10): 17382-17391, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30807656

RESUMO

Heterotopic ossification is common in tendon healing after trauma, but the detailed mechanisms remain unknown. Tendon-derived stem cells (TDSCs) are a type of progenitor cell found in the tendon niche, and their incorrect differentiation after trauma may lead to tendon calcification. The expression of hepatocyte growth factor (HGF) presents drastic fluctuations in serum/tissue after trauma and was found to activate quiescent stellate cells and contribute to wound healing; however, its potential role in TDSCs remains elusive. In this study, TDSCs isolated from rats were cultured in media containing HGF with or without a signaling inhibitor, and the proliferation, migration, and differentiation ability of TDSCs were measured to determine the role and mechanism of HGF in TDSCs. We showed that HGF promotes TDSC proliferation and migration but inhibits TDSC osteogenic differentiation ability. HGF activated-HGF/c-Met, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling, which was positively correlated with TDSCs proliferation and migration but negatively related to TDSC osteogenic differentiation ability. The phosphorylation of Smad1/5/8 was also negatively related to HGF/c-Met, MAPK/ERK1/2, and PI3K/AKT signaling, which demonstrated that the inhibition of osteogenic differentiation was dependent on BMP/Smad1/5/8 signaling. Overall, we showed that HGF could promote TDSCs proliferation and migration and inhibit osteogenic differentiation in vitro, suggesting a potential role for HGF as a cytokine treatment of tendon trauma.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Osteogênese/efeitos dos fármacos , Animais , Células Cultivadas , Fator de Crescimento de Hepatócito/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
3.
Biotechnol Lett ; 39(5): 711-719, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28155178

RESUMO

OBJECTIVES: To investigate the effects of tumor necrosis factor-α (TNF-α) and transforming growth factor-ß1 (TGF-ß1) on the proliferation and differentiation of tendon-derived stem cells (TDSC). RESULTS: TNF-α inhibits the proliferation and tenogenic/osteogenic differentiation of TDSC but, after simultaneous or sequential treatment with TGF-ß1 and TNF-α, the expression of tenogenic/osteogenic-related marker and proliferation of TDSC was significantly increased. During these processes, Smad2/3 and Smad1/5/8 were highly phosphorylated, meaning that the TGF-ß and BMP signaling pathways were highly activated. Further study revealed that the expression of Inhibitor-Smad appeared to be negatively correlated to the proliferation and differentiation of TDSC. CONCLUSIONS: Combining the use of TNF-α and TGF-ß1 could improve the proliferation and differentiation of TDSC in vitro, and the expression of I-Smad is negatively correlated with TDSC proliferation and differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco , Tendões/citologia , Fator de Crescimento Transformador beta1/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad Inibidoras , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
4.
Zhonghua Wai Ke Za Zhi ; 54(4): 317-20, 2016 Apr 01.
Artigo em Zh | MEDLINE | ID: mdl-27029208

RESUMO

The research of bone tissue engineering bases on three basic directions of seed cells, scaffold materials and growth information. Stem cells have been widely studied as seed cells. Human urine-derived stem cell (hUSC) is extracted from urine and described to be adhesion growth, cloning, expression of the majority of mesenchymal stem cell markers and peripheral cell markers, multi-potential and no tumor but stable karyotype with passaging many times. Some researches proposed that hUSC might be a new source of seed cells in tissue engineering because of their invasive and convenient obtention, stable culture and multiple differentiation potential.


Assuntos
Osso e Ossos , Células-Tronco/citologia , Engenharia Tecidual , Urina/citologia , Diferenciação Celular , Humanos , Células-Tronco Mesenquimais
5.
Cytotherapy ; 16(5): 662-73, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24095259

RESUMO

BACKGROUND AIMS: After injury, tendons often heal with poor tissue quality and inferior mechanical properties. Tissue engineering using tendon stem cells (TSCs) is a promising approach in the repair of injured tendon. Tenogenic differentiation of TSCs needs an appropriate environment. More recently, the acellular extracellular matrix (ECM) generated from fibroblasts has been used to construct various engineering tissues. In this study, we successfully developed an engineered tendon tissue formed by seeding TSCs in de-cellularized fibroblast-derived matrix (dFM). METHODS: Patellar TSCs and dermal fibroblast were isolated and cultured. Using the method of osmotic shock, dFM was obtained from dermal fibroblast. ECM proteins in dFM were examined. TSCs at passage 3 were seeded in dFM for 1 week. Proliferative capacity and characterization of TSCs cultured in dFM were determined by population doubling time, immunofluorescence staining and quantitative reverse transcriptase polymerase chain reaction. Engineered tendon tissue was prepared with dFM and TSCs. Its potentials for neo-tendon formation and promoting tendon healing were investigated. RESULTS: dFM is suitable for growth and tenogenic differentiation of TSCs in vitro. Neo-tendon tissue was formed with tendon-specific protein expression when TSCs were implanted together with dFM. In a patellar tendon injury model, implantation of engineered tendon tissue significantly improved the histologic and mechanical properties of injured tendon. CONCLUSIONS: The findings obtained from our study provide a basis for potential use of engineered tendon tissue containing dFM and TSCs in tendon repair and regeneration.


Assuntos
Fibroblastos/citologia , Células-Tronco/citologia , Tendões/citologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Matriz Extracelular/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Neurourol Urodyn ; 33(5): 538-43, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23784934

RESUMO

AIMS: To investigate the protective effects of exogenous hydrogen sulfide (H2 S) against unilateral ureteric obstruction (UUO) induced renal injury and explore the potential mechanisms involved. METHODS: Rats were randomly divided into four groups: sham group, UUO group, UUO + saline group, and UUO + NaHS group. Rats were sacrificed on Day 10 after UUO. The tubulointerstitial injury, interstitial fibrosis, and the infiltrating of macrophages in the interstitium were assessed. Expression of TNF-α in kidney tissue was also measured. Oxidative stress was evaluated by detecting the level of MDA and SOD in renal tissues. RESULTS: Interstitial fibrosis and renal injury score were markedly increased on Day 10 after UUO. In contrast, administration of NaHS significantly reduced the injury score. In addition, the kidneys that were treated with NaHS exhibited minimal interstitial fibrosis. The number of ED1 positive cells in the interstitium and the level of TNF-α were significantly higher in UUO kidneys compared with that in sham group. NaHS treatment significantly attenuated infiltration of macrophages and the expression of TNF-α. Our findings showed a significant increase in MDA level and decrease in the SOD activity in the UUO group compared to the sham group. However, significant decrease in MDA level and increase in SOD activity were observed after treatment of NaHS. CONCLUSIONS: Our study demonstrates that NaHS protects against UUO-induced renal damage via attenuating fibrosis, oxidative stress, and inflammation. Therefore, H2 S may be useful as a potential candidate in the treatment of renal damage induced by obstruction. Neurourol. Urodynam. 33:538-543, 2014. © 2013 Wiley Periodicals, Inc.


Assuntos
Injúria Renal Aguda/prevenção & controle , Gasotransmissores/farmacologia , Sulfeto de Hidrogênio/farmacologia , Rim/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Obstrução Ureteral/complicações , Injúria Renal Aguda/etiologia , Animais , Modelos Animais de Doenças , Fibrose/etiologia , Fibrose/prevenção & controle , Inflamação/metabolismo , Rim/metabolismo , Rim/patologia , Malondialdeído/metabolismo , Ratos , Ratos Wistar , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Med Sci Monit ; 20: 2478-83, 2014 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-25433272

RESUMO

BACKGROUND: Local anesthetics are commonly used for the treatment of a variety of tendinopathies in combination with corticosteroids injection. The goal of this study was to evaluate the effects of lidocaine and triamcinolone acetonide (TA) on cultured rat tenocytes and to determine whether there is a synergistic effect. MATERIAL/METHODS: Rat patellar tendon-derived tenocytes were cultured with or without TA and lidocaine, and the culture without any additive served as the control. Cell morphology and cell viability were evaluated. Expressions of tenocyte-related genes were measured by qRT-PCR. RESULTS: TA, when exposed to tenocytes in vitro, significantly decreased cell viability. The cells cultured with TA had a flattened shape. Moreover, the expressions of tenocyte-related genes in tenocytes were markedly decreased in the TA-treated group. We found that 1% lidocaine synergistically increased the deleterious effects of TA. CONCLUSIONS: Our data provide evidence of the detrimental effects of these drugs on tendon tissues. Injection of TA in combination with 1% lidocaine should be used with caution.


Assuntos
Lidocaína/toxicidade , Tendões/patologia , Triancinolona Acetonida/toxicidade , Animais , Contagem de Células , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Ratos Sprague-Dawley , Tendões/efeitos dos fármacos , Tendões/metabolismo
8.
Biomater Res ; 28: 0013, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617751

RESUMO

Background: Fibrosis and inflammation due to ureteropelvic junction obstruction substantially contributes to poor renal function. Urine-derived stem-cell-derived exosomes (USC-Exos) have therapeutic effects through paracrine. Methods: In vitro, the effects of USC-Exos on the biological functions of HK-2 and human umbilical vein endothelial cells were tested. Cell inflammation and fibrosis were induced by transforming growth factor-ß1 and interleukin-1ß, and their anti-inflammatory and antifibrotic effects were observed after exogenous addition of USC-Exos. Through high-throughput sequencing of microRNA in USC-Exos, the pathways and key microRNAs were selected. Then, the antifibrotic and anti-inflammatory effects of exosomal miR-122-5p and target genes were verified. The role of the miR-122-5p/SOX2 axis in anti-inflammatory and antifibrotic effects was verified. In vivo, a rabbit model of partial unilateral ureteral obstruction (PUUO) was established. Magnetic resonance imaging recorded the volume of the renal pelvis after modeling, and renal tissue was pathologically analyzed. Results: We examined the role of USC-Exos and their miR-122-5p content in obstructive kidney injury. These Exos exhibit antifibrotic and anti-inflammatory activities. SOX2 is the hub gene in PUUO and negatively related to renal function. We confirmed the binding relationship between miR-122-5p and SOX2. The anti-inflammatory and antifibrotic effects of miR-122-5p were inhibited, indicating that miR-122-5p has anti-inflammatory and antifibrotic effects by inhibiting SOX2 expression. In vivo, the PUUO group showed typical obstructive kidney injury after modeling. After USC-Exo treatment, the shape of the renal pelvis shown a remarkable improvement, and inflammation and fibrosis decreased. Conclusions: We confirmed that miR-122-5p from USC-Exos targeting SOX2 is a new molecular target for postoperative recovery treatment of obstructive kidney injury.

9.
J Egypt Natl Canc Inst ; 35(1): 41, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38105365

RESUMO

BACKGROUND: Neuroblastoma, one of the most prevalent childhood cancers, is often treated with surgery, radiation, and chemotherapy. However, prognosis and survival are still dismal for children with neuroblastoma at high risk. Consequently, it is vital to identify new and effective treatment targets. As a component of the meiotic cohesion complex, REC8 is involved in a wide range of malignancies. The current work assessed the impact of REC8 knockdown on SH-SY5Y and SK-N-AS neuroblastoma cells and delved into the molecular mechanism behind this effect. METHODS: Knockdown of REC8 using the small interfering (si) RNA technology, and the results were verified by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot. The Cell Counting Kit-8 (CCK-8) was used to examine cell proliferation, while flow cytometry was used to examine cell cycle progression and apoptosis. Analyses of angiogenesis included tube formation experiments. Transwell tests were used to examine cell migration and invasion. RESULTS: The data showed that downregulation of the REC8 led to a substantial decrease in cell proliferation by stopping the cell cycle in the G1 phase. REC8 knockdown significantly reduced neuroblastoma cell proliferation, migration, invasion, angiogenesis, induced cell cycle arrest, and enhanced apoptosis. We also discovered that repressing REC8 expression in neuroblastoma cell lines SH-SY5Y and SK-N-AS reduced their ability to activate the STAT3/VEGF signaling pathway. CONCLUSIONS: Neuroblastoma therapy may benefit from targeting REC8 and its downstream targets.


Assuntos
Proteínas de Ciclo Celular , Neuroblastoma , Humanos , Angiogênese , Apoptose , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neuroblastoma/genética , Transdução de Sinais , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
10.
Acta Biomater ; 172: 280-296, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37806377

RESUMO

The therapeutic role of tendon stem cells (TSCs) in tendon-related injuries has been well documented. Small extracellular vesicles (sEVs) are being increasingly used as new biotherapeutic agents for various diseases. Therefore, the potential function of TSC-sEVs in tendon injury repair warrants further investigation. In this study, we explored the effects of TSC-sEVs on TSC proliferation, migration, and differentiation in vitro in an autocrine manner. We further used a novel exosomal topical treatment with TSC-sEVs loaded with gelatin methacryloyl (GelMA) hydrogel in vivo; we mixed sufficient amounts of TSC-sEVs with GelMA hydrogel to cover the damaged molded Achilles tendon tissue and then exposed them to UV irradiation for coagulation. GelMA loading ensured that TSC-sEVs were slowly released at the injury site over a long period, thereby achieving their full local therapeutic effects. Treatment with TSC-sEVs loaded with GelMA significantly improved the histological score of the regenerated tendon by increasing the tendon expression while inhibiting the formation of excessive ossification and improving the mechanical properties of the tissue. Moreover, miRNA sequencing in TSC-sEVs, TSCs, and TSCs receiving sEVs revealed that TSC-sEVs altered the miRNA expression profile of TSCs, with increased expression of miR-145-3p. In conclusion, our study demonstrates that TSC-sEVs can play a key role in treating tendon injuries and that loading them with GelMA can enhance their effect in vivo. Moreover, miR-145-3p has a major functional role in the effect of TSC-sEVs. This study offers new therapeutic ideas for the local treatment of Achilles tendon injuries using sEVs. STATEMENT OF SIGNIFICANCE: In this study, we demonstrated that TSC-sEVs play a key role in treating tendon injuries and that loading them with GelMA hydrogel can act as a fixation and slow release in vivo. Moreover, it identifies the major functional role of miR-145-3p in the effect of TSCs that were identified and validated by miRNA sequencing. Our study provides a basis for further research on GelMA slow-release assays that have potential clinical applications. It offers new therapeutic ideas for the local treatment of Achilles tendon injuries using TSC-sEVs.


Assuntos
Tendão do Calcâneo , Vesículas Extracelulares , MicroRNAs , Traumatismos dos Tendões , Humanos , Traumatismos dos Tendões/patologia , MicroRNAs/farmacologia , Células-Tronco , Hidrogéis/farmacologia , Hidrogéis/metabolismo
11.
J Urol ; 187(6): 2249-53, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22503049

RESUMO

PURPOSE: We examined the effectiveness of hydrogen rich saline solution on the prevention of testicular damage induced by ischemia/reperfusion in rats. MATERIALS AND METHODS: Male Sprague-Dawley® rats were divided randomly into 4 groups, including group 1-sham operated, group 2-torsion-detorsion, group 3-torsion-detorsion plus saline and group 4-torsion-detorsion plus hydrogen rich saline solution. Testicular torsion was performed by rotating the left testis 720 degrees clockwise for 4 hours. Reperfusion was allowed for 4 hours. Hydrogen rich saline solution (5 ml/kg) was injected intraperitoneally in rats in group 4 15 minutes before the start of detorsion. Rats were sacrificed after 4-hour initiation of detorsion. Left orchiectomy was done for histopathological examination and biochemical assay. RESULTS: The testicular injury score in groups 2 and 3 was significantly lower than in sham operated group 1 but higher in group 4 with hydrogen rich saline than in group 2 with torsion-detorsion. The apoptosis index was significantly increased in groups 2 and 3. Hydrogen rich saline solution treatment significantly decreased the apoptosis index. A significant increase in malondialdehyde and a decrease in superoxide dismutase activity were observed in groups 2 and 3. In group 4 malondialdehyde was significantly lowered and superoxide dismutase activity was significantly improved compared with groups 2 and 3. CONCLUSIONS: Results provide a biochemical and histopathological basis for the action of hydrogen rich saline solution as a therapeutic agent for testicular damage induced by ischemia/reperfusion injury.


Assuntos
Antioxidantes/uso terapêutico , Hidrogênio/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Cloreto de Sódio/uso terapêutico , Torção do Cordão Espermático/patologia , Testículo/patologia , Animais , Apoptose , Masculino , Substâncias Protetoras , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Torção do Cordão Espermático/fisiopatologia , Testículo/fisiopatologia
12.
Comput Math Methods Med ; 2022: 8504441, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35529267

RESUMO

Clear cell renal carcinoma (ccRCC) is one of the most common renal carcinomas worldwide, which has worse prognosis compared with other subtypes of tumors. We propose a potential RNA regulatory mechanism associated with ccRCC progression. Accordingly, we screened out clinical factors and the expression of RNAs and miRNAs of ccRCC from the TCGA database. 9 lncRNAs (FGF12-AS2, WT1-AS, TRIM36-IT1, AC009093.1, LINC00443, TCL6, COL18A1-AS1, AC110619.1, HOTTIP), 2 miRNAs (mir-155 and mir-21), and 3 mRNAs (COL4A4, ERMP1, PRELID2) were selected from differential expression RNAs and built predictive survival models. The survival models performed very well in predicting prognosis and were found to be highly correlated with tumor stage. In addition, the survival-related lncRNA-miRNA-mRNA (ceRNA) network was constructed by 18 RNAs including 12 mRNAs, 2 miRNAs, and 4 lncRNAs. It is found that the "ECM-receptor interaction," "Pathways in cancer," and "Chemokine signaling pathway" as the main pathways in KEGG pathway analysis. Overall, we established predictive survival model and ceRNA network based on multivariate Cox regression analysis. It may open a new approach and potential biomarkers for clinical prognosis and treatment of ccRCC patients.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , MicroRNAs , RNA Longo não Codificante , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Neoplasias Renais/genética , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
Stem Cells Int ; 2022: 1940761, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35578661

RESUMO

Peritoneal regeneration and repair can alleviate postoperative intraperitoneal adhesions, and mesenchymal stem cells (MSCs) have demonstrated the potential for peritoneal repair and regeneration. However, extracellular vesicles (EVs) are the main carriers for the MSC activity. Thus far, the roles of MSC-derived EVs on peritoneal repair are not well understood. To investigate the therapeutic effect of adipose-derived mesenchymal stem cell-derived EVs (ADSC-EVs) in peritoneal injuries, ADSC-EVs were injected in vivo via the tail vein of rats. The antiadhesion effects were evaluated following abdominal surgery. In addition, the levels of the peritoneal fibrinolysis system were determined via enzyme-linked immunosorbent assay. Expression differences in inflammatory and apoptotic markers were detected using immunofluorescence. The expression of extracellular matrix-related indexes and peritoneal healing were observed using immunohistochemistry. In vitro, rat peritoneal mesothelial cell proliferation was assessed via a 5-ethynyl-2-deoxyuridine assay. Cell migration was determined using scratch wound and transwell assays. Related signaling networks were estimated based on sequencing and bioinformatics analyses. The roles of the MAPK-ERK1/2 and PI3K-Akt signaling networks were analyzed using immunoblotting. This is the first report of the effectiveness of ADSC-EVs in the treatment of postoperative adhesions. ADSC-EVs were incorporated in vitro and induced rat peritoneal mesothelial cell proliferation and migration. This was mediated by stimulation of the MAPK-ERK1/2 and PI3K-Akt axes. ADSC-EVs promote the healing of the injured peritoneum, suggesting a promising therapeutic approach for peritoneal adhesions.

14.
Oxid Med Cell Longev ; 2022: 8065771, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757503

RESUMO

Adipose mesenchymal stromal cell-derived exosomes (ADSC-Exos) have shown great potential in the treatment of oxidative stress induced by ischemia-reperfusion injury. However, alleviation of testicular torsion injury by ADSC-Exos has not been reported. Therefore, we investigated the protective effect of ADSC-Exos against testicular torsion-detorsion injury. ADSC-Exos were isolated by ultracentrifugation and injected into torsion-detorsion-affected testes of rats. H&E staining and sperm quality were used to evaluate the therapeutic effects of ADSC-Exos, and tissue oxidative stress was measured by determining MDA and SOD levels. In addition, TUNEL staining and immunohistological analysis (Ki67, Cleaved Caspase-3, IL-6, IL-10, CCR7, and CD163) were used to clarify the effects of ADSC-Exos on spermatogenic cell proliferation, apoptosis, and the inflammatory microenvironment in vivo. Possible signaling pathways were predicted using sequencing technology and bioinformatics analysis. The predicted signaling pathways were validated in vitro by assessing the proliferation (EdU assay), migration (transwell assay and scratch test), and apoptosis (flow cytometry, TUNEL staining, and western blotting) of spermatogenic cells. The results showed that ADSC-Exos alleviated testicular torsion-detorsion injury by attenuating oxidative stress and the inflammatory response. In addition, ADSC-Exos promoted the proliferation and migration of spermatogenic cells and inhibited their apoptosis by activating the PI3K/AKT and MAPK/ERK1/2 signaling pathways.


Assuntos
Exossomos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Torção do Cordão Espermático , Tecido Adiposo/citologia , Animais , Exossomos/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Sêmen/metabolismo , Torção do Cordão Espermático/metabolismo , Torção do Cordão Espermático/prevenção & controle , Testículo/metabolismo
15.
Eur J Appl Physiol ; 111(7): 1457-63, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21165643

RESUMO

Both myofibroblast differentiation and extracellular matrix (ECM) deposition are essential components of scar formation in tendons, and hepatocyte growth factor (HGF) is reported to prevent fibrogenic responses in tendons. Matrix metalloproteinases-2(MMP-2) is also involved in the healing process in tendons. Whether HGF protects healed Achilles tendons from injury-induced scar formation and the mechanisms are unknown. Daily for 2 weeks after wounding, except for the non-surgical control group, the Achilles tendons in rats were locally injected with HGF (100 ng 50 µl(-1) per mouse) or phosphate-buffered saline (PBS). Histological examination showed HGF ameliorated disorganized collagen fibers caused by surgical incisions in rats. After transforming growth factor beta-1 (TGF-ß1) induced fibrogenic responses in primary Achilles tendon fibroblasts in rats, HGF treatment for 24 h reduced α-smooth muscle actin (α-SMA) (0.60 ± 0.07-fold, P < 0.05) and type III collagen expression (0.39 ± 0.07-fold, P < 0.05). Moreover, HGF elevated MMP-2 expression (1.23 ± 0.11-fold, P < 0.05). The MMP-2 inhibitor, tissue inhibitors of metalloproteinase-1 (TIMP-1), partially blocked the inhibitory effects of HGF on α-SMA expression (from 0.60 ± 0.07-fold to 0.83 ± 0.07-fold, P < 0.05) and type III collagen expression (from 0.39 ± 0.06-fold to 0.86 ± 0.08-fold, P < 0.05). These results indicate HGF attenuates TGF-ß1-induced fibrogenic responses in Achilles tendon, which was mediated by MMP-2. These results will aid in developing effective therapeutic approaches for the dysfunctional repair in Achilles tendons.


Assuntos
Tendão do Calcâneo/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Metaloproteinase 2 da Matriz/fisiologia , Miofibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Tendão do Calcâneo/citologia , Tendão do Calcâneo/metabolismo , Tendão do Calcâneo/fisiologia , Animais , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Colágenos Fibrilares/metabolismo , Injeções , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Miofibroblastos/fisiologia , Ratos , Ratos Sprague-Dawley , Regulação para Cima/efeitos dos fármacos
16.
Front Cell Dev Biol ; 9: 659389, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34222233

RESUMO

The therapeutic impact of stem cells is potentially largely attributable to secretion of exosomes and soluble factors. The present study evaluates the impact of hepatocyte growth factor (HGF)-expressing tendon stem cells (TSCs) on tendon healing in a rat model. Patellar tendon TSCs were isolated and underwent transfection with lentiviral vectors containing HGF or green fluorescent protein (GFP) genes. In vivo, immunohistochemistry of tendons sampled 1 week postsurgery demonstrated that all stem cell-treated groups exhibited higher numbers of CD163+ M2 monocytes and IL-10+ cells (anti-inflammatory), and lower numbers of CCR7+ M1 monocytes and IL-6+ as well as COX-2+ cells (pro-inflammatory). Effects were most pronounced in the HGF-expressing TSCs (TSCs + HGF) treated group. Histology ± immunohistochemistry of tendons sampled 4 and 8 weeks postsurgery demonstrated that all stem cell-treated groups exhibited more ordered collagen fiber arrangement and lower levels of COLIII, α-SMA, TGF-ß1, and fibronectin (proteins relevant to fibroscarring). Effects were most pronounced in the TSCs + HGF-treated group. For the in vitro study, isolated tendon fibroblasts pretreated with TGF-ß1 to mimic the in vivo microenvironment of tendon injury were indirectly cocultured with TSCs, TSCs + GFP, or TSCs + HGF using a transwell system. Western blotting demonstrated that all stem cell types decreased TGF-ß1-induced increases in fibroblast levels of COX-2, COLIII, and α-SMA, concomitant with decreased activation of major TGF-ß1 signaling pathways (p38 MAPK, ERK1/2, but not Smad2/3). This effect was most pronounced for TSCs + HGF, which also decreased the TGF-ß1-induced increase in activation of the Smad2/3 signaling pathway. The presence of specific inhibitors of these pathways during fibroblast TGF-ß1 stimulation also attenuated increases in levels of COX-2, COLIII, and α-SMA. In conclusion, TSCs + HGF, which exhibit HGF overexpression, may promoting tendon healing via decreasing inflammation and fibrosis, perhaps partly via inhibiting TGF-ß1-induced signaling. These findings identify a novel potential therapeutic strategy for tendon injuries, warranting additional research.

17.
Transl Pediatr ; 10(10): 2418-2431, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34765465

RESUMO

BACKGROUND: Wilms tumor (WT) is a widespread urologic tumor in children. Ferroptosis, on the other hand, is a novel form of cell death associated with tumor development. In this study, we aim to explore the predictability of ferroptosis-related biomarkers in estimating prognosis in WT patients. METHODS: To determine a link between ferroptosis-related gene expression and WT prognosis, we first collected RNA sequencing data and clinical information, involving 124 WT and 6 healthy tissue samples, from the TARGET database. Next, we screened the collected information for ferroptosis-related long non-coding RNA using Cox regression analysis, and constructed a signature model, as well as a nomogram, related to prognosis. Finally, we explored a potential link between ferroptosis-related lncRNA and tumor immunity and screened for possible immune checkpoints. RESULTS: We constructed a WT prognosis prediction signature containing 12 ferroptosis-related lncRNAs. The area under the curves values, from the ROC curves, predicting overall survival rates at the 1, 3-, and 5-year timepoints were 0.775, 0.867, and 0.891 respectively. Moreover, we generated a nomogram, using clinical features and risk scores, carrying a C-index value of 0.836, which suggested a high predictive value. We also demonstrated significant differences in tumor immunity between low- and high-risk WT patients, particularly in the presence of B cells, NK cells, Th1 cells, Treg cells, inflammation promoting, and type I and II IFN responses. In addition, we showed that immune checkpoints like SIRPA, ICOSLG, LAG3, PVRIG, NECTIN1, and SIRPB2 can serve as potential therapeutic targets for WT. CONCLUSIONS: Based on our analyses, we generated a ferroptosis-related lncRNA signature that can both estimate prognosis of WT patients and may provide basis for future WT therapy.

18.
Stem Cell Res Ther ; 12(1): 338, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112236

RESUMO

BACKGROUND: The use of adipose-derived mesenchymal stromal cell-derived exosomes (ADSC-Exos) may become a new therapeutic method in biomedicine owing to their important role in regenerative medicine. However, the role of ADSC-Exos in tendon repair has not yet been evaluated. Therefore, we aimed to clarify the healing effects of ADSC-Exos on tendon injury. METHODS: The adipose-derived mesenchymal stromal cells (ADSCs) and tendon stem cells (TSCs) were isolated from the subcutaneous fat and tendon tissues of Sprague-Dawley rats, respectively, and exosomes were isolated from ADSCs. The proliferation and migration of TSCs induced by ADSC-Exos were analyzed by EdU, cell scratch, and transwell assays. We used western blot to analyze the tenogenic differentiation of TSCs and the role of the SMAD signaling pathways. Then, we explored a new treatment method for tendon injury, combining exosome therapy with local targeting using a biohydrogel. Immunofluorescence and immunohistochemistry were used to detect the expression of inflammatory and tenogenic differentiation after tendon injury, respectively. The quality of tendon healing was evaluated by hematoxylin-eosin (H&E) staining and biomechanical testing. RESULTS: ADSC-Exos could be absorbed by TSCs and promoted the proliferation, migration, and tenogenic differentiation of these cells. This effect may have depended on the activation of the SMAD2/3 and SMAD1/5/9 pathways. Furthermore, ADSC-Exos inhibited the early inflammatory reaction and promoted tendon healing in vivo. CONCLUSIONS: Overall, we demonstrated that ADSC-Exos contributed to tendon regeneration and provided proof of concept of a new approach for treating tendon injuries.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Proteínas Smad , Traumatismos dos Tendões/terapia , Tendões/fisiologia , Tecido Adiposo/citologia , Animais , Células Cultivadas , Ratos , Ratos Sprague-Dawley , Proteína Smad1/genética , Tendões/citologia
19.
Front Cell Dev Biol ; 9: 654084, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33898452

RESUMO

Tendon repair is a medical challenge. Our present study investigated the effectiveness of acellular therapy consisting of conditioned medium (CM) of tendon stem cells (TSCs) induced with hepatocyte growth factor (HGF) in promoting the healing of injured Achilles tendon in a rat model. Proteomic analysis of soluble substances in the CM was performed using an array chip, and bioinformatic analysis was carried out to evaluate interactions among the factors. The effects of CM on viability and migratory capacity of tendon fibroblasts derived from rats with ruptured Achilles tendon were evaluated with the Cell Counting Kit 8 and wound healing assay, respectively. The expression of extracellular matrix (ECM)-related protein was assessed by western blotting. Rats with Achilles tendon injury were treated with CM by local injection for 2 weeks, and the organization of tendon fibers at the lesion site was evaluated by hematoxylin and eosin and Masson's trichrome staining of tissue samples. The deposition and degradation of ECM proteins and the expression of inflammatory factors at the lesion site were evaluated by immunohistochemistry and immunofluorescence. Biomechanical testing was carried out on the injured tendons to assess functional recovery. There were 12 bioactive molecules in the CM, with HGF as the hub of the protein-protein interaction network. CM treatment enhanced the viability and migration of tendon fibroblasts, altered the expression of ECM proteins, promoted the organization of tendon fibers, suppressed inflammation and improved the biomechanics of the injured Achilles tendon. These results suggest that HGF stimulates the secretion of soluble secretory products by TSCs and CM promotes the repair and functional recovery of ruptured Achilles tendon. Thus, HGF-induced TSC CM has therapeutic potential for the treatment of tendinopathy.

20.
Front Oncol ; 11: 608433, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718161

RESUMO

Wilms tumor (WT) commonly occurs in infants and children. We evaluated clinical factors and the expression of multiple RNAs in WT samples in the TARGET database. Eight long non-coding RNAs (lncRNAs; AC079310.1, MYCNOS, LINC00271, AL445228.3, Z84485.1, AC091180.5, AP002518.2, and AC007879.3), two microRNAs (miRNAs; hsa-mir-152 andhsa-mir-181a), and nine messenger RNAs (mRNAs; TCTEX1D4, RNF133, VRK1, CCNE1, HEY1, C10orf71, SPRY1, SPAG11A, and MAGEB18) were screened from differentially expressed RNAs and used to construct predictive survival models. These models showed good prognostic ability and were highly correlated with tumor stage and histological classification. Additionally, survival-related ceRNA network was constructed using 35 RNAs (15 lncRNAs, eight miRNAs, and 12 mRNAs). KEGG pathway analysis suggested the "Wnt signaling pathway" and "Cellular senescence" as the main pathways. In conclusion, we established a multinomial predictive survival model and a survival-related ceRNA network, which provide new potential biomarkers that may improve the prognosis and treatment of WT patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA