Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Diabetes Obes Metab ; 26(1): 32-45, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37722965

RESUMO

AIM: To investigate the therapeutic effects and immunomodulatory mechanisms of human placenta-derived mesenchymal stem cells (PMSCs) in diabetic kidney disease (DKD). METHODS: Streptozotocin-induced DKD rats were administered an equivalent volume of saline or PMSCs (1 × 106 in 2 mL phosphate-buffered saline per rat) for 3 weeks. Eight weeks after treatment, we examined the biochemical parameters in the blood and urine, the ratio of T helper 17 cells (Th17) and regulatory T cells (Treg) in the blood, cytokine levels in the kidney and blood, and renal histopathological changes. In addition, we performed PMSC tracing and renal transcriptomic analyses using RNA-sequencing. Finally, we determined whether PMSCs modulated the Th17/Treg balance by upregulating programmed death 1 (PD-1) in vitro. RESULTS: The PMSCs significantly improved renal function, which was assessed by serum creatinine levels, urea nitrogen, cystatin C levels, urinary albumin-creatinine ratio, and the kidney index. Further, PMSCs alleviated pathological changes, including tubular vacuolar degeneration, mesangial matrix expansion, and glomerular filtration barrier injury. In the DKD rats in our study, PMSCs were mainly recruited to immune organs, rather than to the kidney or pancreas. PMSCs markedly promoted the Th17/Treg balance and reduced the levels of pro-inflammatory cytokines (interleukin [IL]-17A and IL-1ß) in the kidney and blood of DKD rats. In vitro experiments showed that PMSCs significantly reduced the proportion of Th17 cells and increased the proportion of Treg cells by upregulating PD-1 in a cell-cell contact manner and downregulating programmed death-ligand 1 (PD-L1) expression in PMSCs, which reversed the Th17/Treg balance. CONCLUSION: We found that PMSCs improved renal function and pathological damage in DKD rats and modulated Th17/Treg balance through the PD-1/PD-L1 pathway. These findings provide a novel mechanism and basis for the clinical use of PMSCs in the treatment of DKD.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Células-Tronco Mesenquimais , Humanos , Ratos , Animais , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/farmacologia , Nefropatias Diabéticas/terapia , Nefropatias Diabéticas/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Ligantes , Fatores Imunológicos/farmacologia , Citocinas/metabolismo , Citocinas/farmacologia , Células-Tronco Mesenquimais/metabolismo , Diabetes Mellitus/metabolismo
2.
Wound Repair Regen ; 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39022990

RESUMO

There is a lack of effective treatment options for diabetic refractory wounds, which presents a critical clinical issue that needs to be addressed urgently. Our research has demonstrated that human placenta-derived mesenchymal stem cells (plaMSCs) facilitate the migration and proliferation of HaCat cells, thereby enhancing diabetic wound healing primarily via the exosomes derived from plaMSCs (plaMSCs-Ex). Using label-free proteomics, plaMSCs and their exosomes were analysed for proteome taxonomic content in order to explore the underlying effective components mechanism of plaMSCs-Ex in diabetic wound healing. Differentially expressed proteins enriched in plaMSCs-Ex were identified and underwent bioinformatics analysis including GO annotation, KEGG pathway enrichment, gene set enrichment analysis (GSEA) and protein-protein interaction analysis (PPI). Results showed that the proteins enriched in plaMSCs-Ex are significantly involved in extracellular matrix organisation, epithelium morphogenesis, cell growth, adhesion, proliferation and angiogenesis. PPI analysis filtered 2 wound healing-related clusters characterised by hub proteins such as POSTN, FN1, SPARC, TIMP1, SERPINE1, LRP1 and multiple collagens. In brief, the exosomal proteins derived from plaMSCs reveal diverse functions of regeneration and tissue remodelling based on proteomics analysis and potentially play a role in diabetic wound healing.

3.
Int Urogynecol J ; 35(3): 553-559, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38206335

RESUMO

INTRODUCTION AND HYPOTHESIS: Polypropylene meshes (PM) used in pelvic organ prolapse surgery are being withdrawn from the market. Although concerns about the usage of PMs in stress incontinence surgery have been raised, it is still one of the best methods of curing stress urinary incontinence. With advancements in stem cell-based therapies, especially mesenchymal stem cells (MSCs), it is believed that coating the synthetic meshes with MSCs may minimize excessive tissue reactions ultimately leading to clinical problems such as pain, erosion or extrusion of the implanted material. In our study we tried to show the possibility of coating the PM with placenta-derived MSCs. METHODS: Mesenchymal stem cells obtained from six placentas were isolated, cultured, and identified. MSCs were then soaked in either fibronectin or collagen prior to co-culturing with strips of PMs. One group is used as a control, and hence was not pretreated before co-culturing. Specimens were fixed and stained with both Gram and hematoxylin and eosin and marked with Vybran Dil and DAPI. All preparations were examined under a light microscope. The IMAGEJ program was utilized to determine the surface area of meshes coated with MSCs. RESULTS: We clearly showed that PMs can be coated successfully with placenta-derived MSCs. The percentage of the coated area is significantly increased when meshes were pretreated with fibronectin or collagen (p<0.0001). CONCLUSIONS: Placenta-derived MSCs can successfully coat PMs. The immunomodulatory properties of MSCs, which may be of great advantage in preventing the side effects of meshes, should be tested by in vivo and hopefully human studies before clinical applications.


Assuntos
Células-Tronco Mesenquimais , Incontinência Urinária por Estresse , Humanos , Polipropilenos , Projetos Piloto , Fibronectinas , Telas Cirúrgicas/efeitos adversos , Colágeno , Incontinência Urinária por Estresse/cirurgia
4.
Ren Fail ; 46(1): 2303396, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38234193

RESUMO

Diabetic kidney disease (DKD) is a common chronic microvascular complication of diabetes mellitus. Although studies have indicated the therapeutic potential of mesenchymal stem cells (MSCs) for DKD, the underlying molecular mechanisms remain unclear. Herein, we explored the renoprotective effect of placenta-derived MSCs (P-MSCs) and the potential mechanism of SIRT1/FOXO1 pathway-mediated autophagy in DKD. The urine microalbumin/creatinine ratio was determined using ELISA, and renal pathological changes were detected by special staining techniques. Immunofluorescence was used for detecting the renal tissue expression of podocin and nephrin; immunohistochemistry for the renal expression of autophagy-related proteins (LC3, Beclin-1, SIRT1, and FOXO1); and western blotting and PCR for the expression of podocyte autophagy- and pathway-related indicators. We found that P-MSCs ameliorated renal tubular injury and glomerular mesangial matrix deposition and alleviated podocyte damage in DKD rats. PMSCs enhanced autophagy levels and increased SIRT1 and FOXO1 expression in DKD rat renal tissue, whereas the autophagy inhibitor 3-methyladenine significantly attenuated the renoprotective effect of P-MSCs. P-MSCs improved HG-induced Mouse podocyte clone5(MPC5)injury, increased podocyte autophagy, and upregulated SIRT1 and FOXO1 expression. Moreover, downregulation of SIRT1 expression blocked the P-MSC-mediated enhancement of podocyte autophagy and improvement of podocyte injury. Thus, P-MSCs can significantly improve renal damage and reduce podocyte injury in DKD rats by modulating the SIRT1/FOXO1 pathway and enhancing podocyte autophagy.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Células-Tronco Mesenquimais , Podócitos , Ratos , Camundongos , Animais , Nefropatias Diabéticas/tratamento farmacológico , Sirtuína 1/metabolismo , Autofagia , Rim/patologia , Células-Tronco Mesenquimais/metabolismo , Podócitos/patologia
5.
J Cell Biochem ; 124(9): 1249-1258, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37450693

RESUMO

This study aims to investigate the effect of placenta-derived mesenchymal stem cells (PMSCs) administration on tissue repair following acute lung injury (ALI). PMSCs were transplanted intravenously to a mouse model of lipopolysaccharide-induced ALI. The therapeutic effects were determined by evaluating several indicators, including pathology; the wet/dry ratio of the lungs; blood gas analysis; the total protein content, cell numbers, and the activity of myeloperoxidase (MPO) in bronchial alveolar lavage fluid (BALF); and the levels of anti-inflammatory and proinflammatory cytokines in serum and BALF. To investigate the underlying mechanism, PMSC-derived exosomes were used for ALI treatment. Administration of PMSCs improved the degree of lung injury, reduced inflammation, increased the expression levels of anti-inflammatory cytokines, and protected lung function. As expected, the effects of PMSC-derived exosomes in the ALI model were similar to those of PMSCs, both in terms of improved lung function and reduced inflammation. These findings suggest that PMSCs have ameliorating effects on ALI that are potentially mediated via their secreted exosomes.


Assuntos
Lesão Pulmonar Aguda , Células-Tronco Mesenquimais , Camundongos , Animais , Humanos , Células-Tronco Mesenquimais/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/terapia , Lesão Pulmonar Aguda/metabolismo , Pulmão/metabolismo , Citocinas/metabolismo , Lipopolissacarídeos/efeitos adversos , Fatores Imunológicos , Inflamação/metabolismo
6.
Int J Mol Sci ; 24(22)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38003735

RESUMO

The insulin resistance caused by impaired glucose metabolism induces ovarian dysfunction due to the central importance of glucose as a source of energy. However, the research on glucose metabolism in the ovaries is still lacking. The objectives of this study were to analyze the effect of PD-MSCs on glucose metabolism through IGFBP2-AMPK signaling and to investigate the correlation between glucose metabolism and ovarian function. Thioacetamide (TAA) was used to construct a rat injury model. PD-MSCs were transplanted into the tail vein (2 × 106) 8 weeks after the experiment started. The expression of the IGFBP2 gene and glucose metabolism factors (e.g., AMPK, GLUT4) was significantly increased in the PD-MSC group compared to the nontransplantation (NTx) group (* p < 0.05). The levels of follicular development markers and the sex hormones AMH, FSH, and E2 were also higher than those in the TAA group. Using ex vivo cocultivation, the mRNA and protein expression of IGFBP2, AMPK, and GLUT4 were significantly increased in the cocultivation with the PD-MSCs group and the recombinant protein-treated group (* p < 0.05). These findings suggest that the increased IGFBP2 levels by PD-MSCs play an important role in glucose metabolism and ovarian function through the IGFBP2-AMPK signaling pathway.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Animais , Tioacetamida/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Glucose/metabolismo
7.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36902127

RESUMO

The use of mesenchymal stem cells (MSCs) has become a new strategy for treating diabetic kidney disease (DKD). However, the role of placenta derived mesenchymal stem cells (P-MSCs) in DKD remains unclear. This study aims to investigate the therapeutic application and molecular mechanism of P-MSCs on DKD from the perspective of podocyte injury and PINK1/Parkin-mediated mitophagy at the animal, cellular, and molecular levels. Western blotting, reverse transcription polymerase chain reaction, immunofluorescence, and immunohistochemistry were used to detect the expression of podocyte injury-related markers and mitophagy-related markers, SIRT1, PGC-1α, and TFAM. Knockdown, overexpression, and rescue experiments were performed to verify the underlying mechanism of P-MSCs in DKD. Mitochondrial function was detected by flow cytometry. The structure of autophagosomes and mitochondria were observed by electron microscopy. Furthermore, we constructed a streptozotocin-induced DKD rat model and injected P-MSCs into DKD rats. Results showed that as compared with the control group, exposing podocytes to high-glucose conditions aggravated podocyte injury, represented by a decreased expression of Podocin along with increased expression of Desmin, and inhibited PINK1/Parkin-mediated mitophagy, manifested as a decreased expression of Beclin1, the LC3II/LC3I ratio, Parkin, and PINK1 associated with an increased expression of P62. Importantly, these indicators were reversed by P-MSCs. In addition, P-MSCs protected the structure and function of autophagosomes and mitochondria. P-MSCs increased mitochondrial membrane potential and ATP content and decreased the accumulation of reactive oxygen species. Mechanistically, P-MSCs alleviated podocyte injury and mitophagy inhibition by enhancing the expression of the SIRT1-PGC-1α-TFAM pathway. Finally, we injected P-MSCs into streptozotocin-induced DKD rats. The results revealed that the application of P-MSCs largely reversed the markers related to podocyte injury and mitophagy and significantly increased the expression of SIRT1, PGC-1α, and TFAM compared with the DKD group. In conclusion, P-MSCs ameliorated podocyte injury and PINK1/Parkin-mediated mitophagy inhibition in DKD by activating the SIRT1-PGC-1α-TFAM pathway.


Assuntos
Nefropatias Diabéticas , Células-Tronco Mesenquimais , Podócitos , Animais , Feminino , Gravidez , Ratos , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Células-Tronco Mesenquimais/metabolismo , Mitofagia , Placenta/citologia , Placenta/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Proteínas Quinases/metabolismo , Sirtuína 1/metabolismo , Estreptozocina , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
8.
J Cell Physiol ; 236(9): 6678-6690, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33624308

RESUMO

The trophoblast is a critical cell for placental development and embryo implantation in the placenta. We previously reported that placenta-derived mesenchymal stem cells (PD-MSCs) increase trophoblast invasion through several signaling pathways. However, the paracrine effects of PD-MSCs on mitochondrial function in trophoblasts are still unclear. Therefore, the objective of the study was to analyze the mitochondrial function of trophoblasts in response to cocultivation with PD-MSCs. The results showed that PD-MSCs regulate the balance between cell survival and death and protect damaged mitochondria in trophoblasts from oxidative stress. Moreover, PD-MSCs upregulate factors involved in mitochondrial autophagy in trophoblast cells. Finally, PD-MSCs improve trophoblast invasion. Taken together, the data indicate that PD-MSCs can regulate trophoblast invasion through dynamic effects on mitochondrial energy metabolism. These results support the fundamental role of mitochondrial energy mechanism in trophoblast invasion and suggest a new therapeutic strategy for infertility.


Assuntos
Células-Tronco Mesenquimais/citologia , Mitocôndrias/metabolismo , Placenta/citologia , Trofoblastos/citologia , Biomarcadores/metabolismo , Respiração Celular , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica , Glicólise/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mitofagia , Consumo de Oxigênio , Gravidez , Espécies Reativas de Oxigênio/metabolismo , Trofoblastos/metabolismo
9.
J Surg Res ; 265: 100-113, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33895582

RESUMO

BACKGROUND: Stem cell-based regenerative therapy is a novel approach to severe damaged skin. Perinatal tissues such as placenta are considered as promising alternatives. The present study aimed to investigate the effect of insulin-like growth factor-1 (IGF-1)-expressing placenta-derived mesenchymal stem cells (hPMSCs) on healing of burn wounds. MATERIALS AND METHODS: hPMSCs were isolated from human placenta, and IGF-1 was transducted into hPMSCs via lentivirus. Flow cytometry and MTT assay were performed to assess cell apoptosis and viability, respectively. Immunostaining of CK19 and ki67 was for evaluating epithelial differentiation ability and cell proliferation. For in vivo studies, we established a mouse model of scalding and performed local administration of IGF-1-expressing hPMSCs via subcutaneous injection. Wound histology was analyzed with H&E staining. The expression of fibrogenic cytokines was detected by western blot. The production of pro-inflammatory factors was measured by ELISA. RESULTS: Overexpression of IGF-1 promoted cell proliferation and epithelial differentiation of hPMSCs in vitro and in vivo. Mice with burn injury displayed increased wound contraction and healing rates following treatment with IGF-1-expressing hPMSCs. There was less inflammatory infiltration and reduced collagen disposition in the presence of IGF-1 at the wound site. Administration of IGF-1-expressing hPMSCs suppressed inflammation by decreasing the levels of pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1ß, and interleukin-6. Besides, IGF-1 increased VEGF expression, and decreased TGF-ß1, collagen I and collagen III expressions in vivo. CONCLUSIONS: IGF-1-expressing PMSCs promotes cell proliferation and epithelial differentiation, inhibits inflammation and collagen deposition, and thus contributes to wound healing.


Assuntos
Queimaduras/terapia , Fator de Crescimento Insulin-Like I/metabolismo , Transplante de Células-Tronco Mesenquimais , Placenta/citologia , Cicatrização , Animais , Queimaduras/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Humanos , Fator de Crescimento Insulin-Like I/genética , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Gravidez , Distribuição Aleatória
10.
Int J Mol Sci ; 22(22)2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34830410

RESUMO

Human pluripotent stem cell-derived neural progenitor cells (NPCs) have the potential to recover from nerve injury. We previously reported that human placenta-derived mesenchymal stem cells (PSCs) have neuroprotective effects. To evaluate the potential benefit of NPCs, we compared them to PSCs using R28 cells under hypoxic conditions and a rat model of optic nerve injury. NPCs and PSCs (2 × 106 cells) were injected into the subtenon space. After 1, 2, and 4 weeks, we examined changes in target proteins in the retina and optic nerve. NPCs significantly induced vascular endothelial growth factor (Vegf) compared to age-matched shams and PSC groups at 2 weeks; they also induced neurofilaments in the retina compared to the sham group at 4 weeks. In addition, the expression of brain-derived neurotrophic factor (Bdnf) was high in the retina in the NPC group at 2 weeks, while expression in the optic nerve was high in both the NPC and PSC groups. The low expression of ionized calcium-binding adapter molecule 1 (Iba1) in the retina had recovered at 2 weeks after NPC injection and at 4 weeks after PSC injection. The expression of the inflammatory protein NLR family, pyrin domain containing 3 (Nlrp3) was significantly reduced at 1 week, and that of tumor necrosis factor-α (Tnf-α) in the optic nerves of the NPC group was lower at 2 weeks. Regarding retinal ganglion cells, the expressions of Brn3a and Tuj1 in the retina were enhanced in the NPC group compared to sham controls at 4 weeks. NPC injections increased Gap43 expression from 2 weeks and reduced Iba1 expression in the optic nerves during the recovery period. In addition, R28 cells exposed to hypoxic conditions showed increased cell survival when cocultured with NPCs compared to PSCs. Both Wnt/ß-catenin signaling and increased Nf-ĸb could contribute to the rescue of damaged retinal ganglion cells via upregulation of neuroprotective factors, microglial engagement, and anti-inflammatory regulation by NPCs. This study suggests that NPCs could be useful for the cellular treatment of various optic neuropathies, together with cell therapy using mesenchymal stem cells.


Assuntos
Células-Tronco Neurais/transplante , Doenças do Nervo Óptico/terapia , Traumatismos do Nervo Óptico/terapia , Nervo Óptico/crescimento & desenvolvimento , Células-Tronco Pluripotentes/transplante , Animais , Axônios/metabolismo , Axônios/fisiologia , Sobrevivência Celular/genética , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Feminino , Humanos , Regeneração Nervosa/genética , Nervo Óptico/patologia , Nervo Óptico/transplante , Doenças do Nervo Óptico/patologia , Gravidez , Ratos , Células Ganglionares da Retina/transplante
11.
Dent Traumatol ; 37(1): 73-80, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32794607

RESUMO

BACKGROUND/AIM: The viability of periodontal ligament cells on the root surface is a major factor that influences the healing of replanted teeth. A suitable storage medium is necessary to preserve avulsed teeth before replantation. Conditioned medium from placenta-derived mesenchymal stem cells (PMSC-CM) contains a variety of growth factors. The aim of this study was to evaluate the effectiveness of PMSC-CM as a storage medium to maintain the cell viability of avulsed teeth. MATERIAL AND METHODS: Extracted premolars from healthy humans were randomly stored in Hank's balanced salt solution (HBSS) and PMSC-CM for 6, 12 and 24 hours, respectively, at room temperature, and then the ratio of apoptosis of the periodontal ligament (PDL) cells was identified by flow cytometry. Human periodontal ligament stem cells (PDLSCs) were incubated with HBSS and PMSC-CM, respectively, for 6, 12, 24 and 48 hours in 5% CO2 at 37°C. Then, the cell viability of PDLSCs was determined using the cell counting kit-8 (CCK-8) and a cell cycle assay was performed. RESULTS: The apoptosis rate of PDL cells in PMSC-CM was significantly lower than that in HBSS at 24 hours (P < .001), while the two groups showed similar cell apoptosis rates at 6 and 12 hours (P > .05). The cell proliferation of PDLSCs treated with PMSC-CM significantly increased compared with the HBSS group (P < .05). The cell cycle assay revealed that the PDLSCs treated with HBSS were arrested at the G1 phase, while there was no difference between the PMSC-CM group and the control group (P > .05). CONCLUSIONS: Compared with HBSS, PMSC-CM showed better inhibition of apoptosis of PDL cells and promoted the proliferation of PDLSCs. Thus, PMSC-CM could be a promising storage medium for avulsed teeth.


Assuntos
Células-Tronco Mesenquimais , Soluções para Preservação de Órgãos , Avulsão Dentária , Animais , Sobrevivência Celular , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Soluções Isotônicas , Leite , Ligamento Periodontal , Placenta , Gravidez , Avulsão Dentária/terapia
12.
Int J Mol Sci ; 21(22)2020 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-33202697

RESUMO

Mitochondrial dynamics are involved in many cellular events, including the proliferation, differentiation, and invasion/migration of normal as well as cancerous cells. Human placenta-derived mesenchymal stem cells (PD-MSCs) were known to regulate the invasion activity of trophoblasts. However, the effects of PD-MSCs on mitochondrial function in trophoblasts are still insufficiently understood. Therefore, the objectives of this study are to analyze the factors related to mitochondrial function and investigate the correlation between trophoblast invasion and mitophagy via PD-MSC cocultivation. We assess invasion ability and mitochondrial function in invasive trophoblasts according to PD-MSC cocultivation by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and extracellular flux (XF) assay. Under PD-MSCs co-cultivation, invasion activity of a trophoblast is increased via activation of the Rho signaling pathway as well as Matrix metalloproteinases (MMPs). Additionally, the expression of mitochondrial function (e.g., reactive oxygen species (ROS), calcium, and adenosine triphosphate (ATP) synthesis) in trophoblasts are increased via PD-MSCs co-cultivation. Finally, PD-MSCs regulate mitochondrial autophagy factors in invasive trophoblasts via regulating the balance between PTEN-induced putative kinase 1 (PINK1) and parkin RBR E3 ubiquitin protein ligase (PARKIN) expression. Taken together, these results demonstrate that PD-MSCs enhance the invasion ability of trophoblasts via altering mitochondrial dynamics. These results support the fundamental mechanism of trophoblast invasion via mitochondrial function and provide a new stem cell therapy for infertility.


Assuntos
Regulação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Transdução de Sinais , Trofoblastos/metabolismo , Linhagem Celular , Humanos , Células-Tronco Mesenquimais/citologia , Trofoblastos/citologia
13.
Int J Mol Sci ; 20(21)2019 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-31653075

RESUMO

Placenta-derived mesenchymal stem cells (PD-MSCs) were highlighted as therapeutic sources in several degenerative diseases. Recently, microRNAs (miRNAs)were found to mediate one of the therapeutic mechanisms of PD-MSCs in regenerative medicine. To enhance the therapeutic effects of PD-MSCs, we established functionally enhanced PD-MSCs with phosphatase of regenerating liver-1 overexpression (PRL-1(+)). However, the profile and functions of miRNAs induced by PRL-1(+) PD-MSCs in a rat model with hepatic failure prepared by bile duct ligation (BDL) remained unclear. Hence, the objectives of the present study were to analyze the expression of miRNAs and investigate their therapeutic mechanisms for hepatic regeneration via PRL-1(+) in a rat model with BDL. We selected candidate miRNAs based on microarray analysis. Under hypoxic conditions, compared with migrated naïve PD-MSCs, migrated PRL-1(+) PD-MSCs showed improved integrin-dependent migration abilitythrough Ras homolog (RHO) family-targeted miRNA expression (e.g., hsa-miR-30a-5p, 340-5p, and 146a-3p). Moreover, rno-miR-30a-5p and 340-5p regulated engraftment into injured rat liver by transplantedPRL-1(+) PD-MSCs through the integrin family. Additionally, an increase inplatelet-derived growth factor receptor A (PDGFRA) by suppressing rno-miR-27a-3p improved vascular structure in rat liver tissues after PRL-1(+) PD-MSC transplantation. Furthermore, decreased rno-miR-122-5p was significantly correlated with increased proliferation of hepatocytes in liver tissues by PRL-1(+) PD-MSCs byactivating the interleukin-6 (IL-6) signaling pathway through the repression of rno-miR-21-5p. Taken together, these findings improve the understandingof therapeutic mechanisms based on miRNA-mediated stem-cell therapy in liver diseases.


Assuntos
Falência Hepática/terapia , Fígado/fisiologia , Transplante de Células-Tronco Mesenquimais , MicroRNAs/metabolismo , Regeneração , Animais , Hipóxia Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Interleucina-6/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Placenta/citologia , Gravidez , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Remodelação Vascular
14.
J Cell Biochem ; 117(5): 1145-57, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26448639

RESUMO

Mesenchymal stem cells (MSCs) are a powerful source for cell therapy in degenerative diseases. The migration ability of MSCs is an important factor that enhances the therapeutic effect of the cells when they are transplanted into target tissues or organs. Hypoxia and the endothelial barrier, which are representative migration microenvironmental factors, are known to be regulated by the integrin-mediated pathway in several cancers. However, their regulatory mechanisms in MSCs remain unclear. Here, the objectives of the study were to compare the expression of markers related to integrin-mediated signaling in placenta-derived MSCs (PDMSCs) dependent on hypoxia and co-cultured with human umbilical vein endothelial cells (HUVECs) and to evaluate their correlations between migration ability and microenvironmetal factors including hypoxia and endothelial cells. The migration abilities of PDMSCs exposed to hypoxic conditions were significantly increased compared with normal fibroblasts (WI-38) and control (P < 0.05). Interestingly, decreased integrin α4 in PDMSCs under hypoxia induce to increase migration abilities of PDMSCs. Also, Rho family-related markers were significantly increased in PDMSCs under hypoxic conditions compared with normoxia (P < 0.05). Furthermore, the migration ability of PDMSCs was decreased by Rho kinase inhibitor treatment (Y-27632) and co-culturing with HUVECs in an ex vivo system. ROCK activity was increased by inhibiting integrin α4 with HUVECs and hypoxia compared with the absence of HUVECs and under normoxia. The findings suggest microenvironment event by hypoxia and the interaction with endothelial cells may be useful as a regulator of MSC migration and provide insight into the migratory mechanism of MSCs in stem cell-based therapy.


Assuntos
Movimento Celular , Células Endoteliais da Veia Umbilical Humana/citologia , Integrina alfa4/metabolismo , Células-Tronco Mesenquimais/citologia , Quinases Associadas a rho/metabolismo , Amidas/farmacologia , Western Blotting , Hipóxia Celular , Linhagem Celular , Células Cultivadas , Microambiente Celular , Técnicas de Cocultura , Inibidores Enzimáticos/farmacologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Integrina alfa4/genética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Células-Tronco Mesenquimais/metabolismo , Placenta/citologia , Gravidez , Piridinas/farmacologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
15.
Biotechnol Lett ; 38(6): 931-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26932904

RESUMO

OBJECTIVES: To explore the effect of placenta-derived mesenchymal stem cells on scar formation as well as the underlying mechanism. RESULTS: The isolated placenta-derived mesenchymal stem cells from mice were distributed in the wounded areas of scalded mouse models, attenuated inflammatory responses and decreased the deposition of collagens, thus performing a beneficial effect against scar formation. Hypoxia enhanced the protective effect of placenta-derived mesenchymal stem cells and hypoxia-inducible factor-1α was involved in the protective effect of placenta-derived mesenchymal stem cells in hypoxic condition. CONCLUSIONS: Hypoxia enhanced the protective effect of placenta-derived mesenchymal stem cells through hypoxia-inducible factor-1α and PMSCs may have a potential application in the treatment of wound.


Assuntos
Cicatriz/prevenção & controle , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Placenta/citologia , Animais , Hipóxia Celular , Cicatriz/metabolismo , Cicatriz/patologia , Colágeno/metabolismo , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Feminino , Inflamação/prevenção & controle , Inflamação/terapia , Células-Tronco Mesenquimais/fisiologia , Camundongos Endogâmicos C57BL , Gravidez , Cicatrização/fisiologia
16.
Int J Med Sci ; 11(9): 870-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25013366

RESUMO

MSCs-based therapy for cancer is a relatively new but rapidly growing area of research. Human term placenta, an attractive source of MSCs (PMSCs), appears to have great advantage due to its easy access without invasive procedures, its lack of ethical issues and its high-throughput and young age. In the present study, we isolated MSCs from placenta and characterized their morphology and differentiation capacities. We next investigated the underlying antitumor effects and the potential mechanism of PMSCs to express endostatin using adenoviral transduction (Ad-Endo) in a colorectal peritoneal carcinomatosis (CRPC) mouse model. For in vitro experiments, the migratory potential of Ad-Endo-PMSCs towards tumor cells was demonstrated using a double-chamber assay, and the anti-angiogenesis ability of endostatin from engineered PMSCs was evaluated using the tube formation assay. For the in vivo study, mice harboring CT26 colorectal cancer indicated a significant reduction in tumor nodules and a prolongation of survival following Ad-Endo-PMSCs therapy. These observations were associated with significantly decreased tumor cell proliferation and blood vessel counts as well as increased tumor cell apoptosis. These data suggested the potential of PMSCs-based gene therapy for the targeted delivery of therapeutic proteins in cancer.


Assuntos
Carcinogênese/genética , Neoplasias Colorretais/genética , Endostatinas/biossíntese , Peritônio/patologia , Adenoviridae , Animais , Neoplasias Colorretais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Placenta/citologia , Gravidez
17.
Cell Transplant ; 33: 9636897241228922, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38334047

RESUMO

To overcome the difficulty of vascular regeneration in exposed tendon wounds, we combined human placenta-derived mesenchymal stem cells (hPMSCs) with an artificial dermal scaffold and assessed their role in promoting vascular regeneration and wound healing in vivo. hPMSCs were isolated from the human placenta and characterized based on their morphology, phenotypic profiles, and pluripotency. New Zealand rabbits were used to establish an exposed tendon wound model, and hPMSCs and artificial dermal scaffolds were transplanted into the wounds. The results of gross wound observations and pathological sections showed that hPMSCs combined with artificial dermal scaffold transplantation increased the vascularization area of the wound, promoted wound healing, and increased the survival rate of autologous skin transplantation. Following artificial dermal scaffold transplantation, hPMSCs accelerated the vascularization of the dermal scaffold, and the number of fibroblasts, collagen fibers, and neovascularization in the dermal scaffold after 1 week were much higher than those in the control group. Immunohistochemical staining further confirmed that the expression of the vascular endothelial cell marker, CD31, was significantly higher in the combined transplantation group than in the dermal scaffold transplantation group. Our findings demonstrated that hPMSCs seeded onto artificial dermal scaffold could facilitate vascularization of the dermal scaffold and improve tendon-exposed wound healing.


Assuntos
Células-Tronco Mesenquimais , Alicerces Teciduais , Humanos , Coelhos , Animais , Cicatrização , Pele/irrigação sanguínea , Tendões
18.
Curr Stem Cell Res Ther ; 19(11): 1484-1496, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38204245

RESUMO

BACKGROUND: The effectiveness of fucoxanthin (Fx) in liver diseases has been reported due to its anti-inflammatory and antifibrotic effects. Mesenchymal stem cells (MSCs)-based therapy has also been proposed as a promising strategy for liver fibrosis treatment. Recent studies have shown that the co-administration of MSCs and drugs demonstrates a pronounced effect on liver fibrosis. AIM: This study aimed to determine the therapeutic potential of placenta-derived MSCs (PD-MSCs) in combination with Fx to treat liver fibrosis and evaluate their impact on the main links of liver fibrosis pathogenesis. METHODS: After PD-MSCs isolation and identification, outbred ICR/CD1 mice were divided into five groups: Control group, CCl4 group (CCl4), Fx group (CCl4+Fx), PD-MSCs group (CCl4+MSCs) and cotreatment group (CCl4+MSCs+Fx). Biochemical histopathological investigations were performed. Semiquantitative analysis of the alpha-smooth muscle actin (α-SMA+), matrix metalloproteinases (MMP-9+, MMP-13+), tissue inhibitor of matrix metalloproteinases-1 (TIMP-1+) areas, and the number of positive cells in them were studied by immunohistochemical staining. Transforming growth factor-beta (TGF-ß), hepatic growth factor (HGF), procollagen-1 (COL1α1) in liver homogenate and proinflammatory cytokines in blood serum were determined using an enzyme immunoassay. RESULTS: Compared to the single treatment with PD-MSCs or Fx, their combined administration significantly reduced liver enzyme activity, the severity of liver fibrosis, the proinflammatory cytokine levels, TGF-ß level, α-SMA+, TIMP-1+ areas and the number of positive cells in them, and increased HGF level, MMP-13+, and MMP-9+ areas. CONCLUSION: Fx enhanced the therapeutic potential of PD-MSCs in CCl4-induced liver fibrosis, but more investigations are necessary to understand the mutual impact of PD-MSCs and Fx.


Assuntos
Tetracloreto de Carbono , Modelos Animais de Doenças , Cirrose Hepática , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Placenta , Inibidor Tecidual de Metaloproteinase-1 , Xantofilas , Animais , Células-Tronco Mesenquimais/metabolismo , Feminino , Placenta/metabolismo , Xantofilas/farmacologia , Camundongos , Gravidez , Cirrose Hepática/terapia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Camundongos Endogâmicos ICR , Metaloproteinase 9 da Matriz/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Metaloproteinase 13 da Matriz/metabolismo , Actinas/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Masculino , Fator de Crescimento Transformador beta1
19.
Front Biosci (Landmark Ed) ; 29(4): 139, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38682178

RESUMO

BACKGROUND: Hypoxic-ischaemic encephalopathy (HIE) is a major cause of neonatal disability and mortality. Although hypothermia therapy offers some neuroprotection, the recovery of neurological function is limited. Therefore, new synergistic therapies are necessary to improve the prognosis. Mesenchymal stem cell-based therapy is emerging as a promising treatment option for HIE. In this study, we studied the therapeutic efficacy of human placenta-derived mesenchymal stem cells (PD-MSCs) in the HIE rat model and analyzed the underlying therapeutic mechanisms. METHODS: Rats were divided into 6 groups (n = 9 for each) as follows: control, HIE model, HIE + normal saline, and HIE + PD-MSC transplantation at days 7, 14 and 28 postpartum. Following PD-MSC transplantation, neurological behavior was evaluated using rotarod tests, traction tests, and the Morris water maze test. The degree of brain tissue damage was assessed by histological examination and Nissl staining. Expression levels of apoptosis-related proteins and inflammatory factors were quantified by Western blotting and enzyme-linked immunosorbent assays. Immunofluorescence was used to investigate the ability of PD-MSCs to repair the morphology and function of hippocampal neurons with hypoxic-ischaemic (HI) injury. RESULTS: PD-MSC transplantation enhanced motor coordination and muscle strength in HIE rats. This treatment also improved spatial memory ability by repairing pathological damage and preventing the loss of neurons in the cerebral cortex. The most effective treatment was observed in the HIE + PD-MSC transplantation at day 7 group. Expression levels of microtubule-associated protein-2 (MAP-2), B-cell lymphoma-2 (BCL-2), interleukin (IL)-10, and transforming growth factor (TGF -ß1) were significantly higher in the HIE + PD-MSC treatment groups compared to the HIE group, whereas the levels of BCL-2-associated X protein (BAX), BCL-2-associated agonist of cell death (BAD), IL-1ß and tumour necrosis factor α (TNF-α) were significantly lower. CONCLUSIONS: We demonstrated that intravenous injection of PD-MSC at 7, 14 and 28 days after intrauterine HI damage in a rat model could improve learning, memory, and motor function, possibly by inhibiting apoptosis and inflammatory damage. These findings indicate that autologous PD-MSC therapy could have potential application for the treatment of HIE.


Assuntos
Apoptose , Hipóxia-Isquemia Encefálica , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Placenta , Ratos Sprague-Dawley , Animais , Feminino , Transplante de Células-Tronco Mesenquimais/métodos , Gravidez , Hipóxia-Isquemia Encefálica/terapia , Humanos , Placenta/citologia , Células-Tronco Mesenquimais/citologia , Ratos , Modelos Animais de Doenças , Hipocampo/metabolismo , Inflamação/terapia , Neurônios/metabolismo , Masculino
20.
Cells ; 13(14)2024 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-39056810

RESUMO

Age-related ocular diseases such as age-related macular degeneration, glaucoma, and diabetic retinopathy are major causes of irreversible vision impairment in the elderly. Conventional treatments focus on symptom relief and disease slowdown, often involving surgery, but fall short of providing a cure, leading to substantial vision loss. Regenerative medicine, particularly mesenchymal stem cells (MSCs), holds promise for ocular disease treatment. This study investigates the synergistic potential of combining placenta-derived MSCs (PD-MSCs) with Achyranthis radix extract (ARE) from Achyranthes japonica to enhance therapeutic outcomes. In a 24-h treatment, ARE significantly increased the proliferative capacity of PD-MSCs and delayed their senescence (* p < 0.05). ARE also enhanced antioxidant capabilities and increased the expression of regeneration-associated genes in an in vitro injured model using chemical damages on human retinal pigment epithelial cell line (ARPE-19) (* p < 0.05). These results suggest that ARE-primed PD-MSC have the capability to enhance the activation of genes associated with regeneration in the injured eye via increasing antioxidant properties. Taken together, these findings support the conclusion that ARE-primed PD-MSC may serve as an enhanced source for stem cell-based therapy in ocular diseases.


Assuntos
Antioxidantes , Células-Tronco Mesenquimais , Placenta , Extratos Vegetais , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Antioxidantes/farmacologia , Extratos Vegetais/farmacologia , Feminino , Placenta/metabolismo , Placenta/efeitos dos fármacos , Gravidez , Achyranthes/química , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/citologia , Proliferação de Células/efeitos dos fármacos , Linhagem Celular
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa