Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Reproduction ; 168(2)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38912966

RESUMO

In brief: This study reveals that orthotopic transplantation of 3D hUC-MSC spheroids is more effective than monolayer-cultured hUC-MSCs in improving POF and distinctly reducing oxidative stress through the paracrine effect, thereby preventing apoptosis and autophagy of GCs. Abstract: Premature ovarian failure (POF) is a common reproductive disease in women younger than 40 years old, and studies have demonstrated that the application of human umbilical cord mesenchymal stem cells (hUC-MSCs) is a promising therapy strategy for POF. Given the previously established therapeutic advantages of 3D MSC spheroids, and to evaluate their effectiveness, both 3D hUC-MSC spheroids and monolayer-cultured hUC-MSCs were employed to treat a cyclophosphamide-induced POF rat model through orthotopic transplantation. The effects of these two forms on POF were subsequently assessed by examining apoptosis, autophagy, and oxidative damage in ovarian granulosa cells (GCs). The results indicated that hUC-MSC spheroids exhibited superior treatment effects on resisting autophagy, apoptosis, and oxidative damage in GCs compared to monolayer-cultured hUC-MSCs. To further elucidate the impact of hUC-MSC spheroids in vitro, a H2O2-induced KGN cells model was established and co-cultured with both forms of hUC-MSCs. As expected, the hUC-MSC spheroids also exhibited superior effects in resisting apoptosis and autophagy caused by oxidative damage. Therefore, this study demonstrates that 3D hUC-MSC spheroids have potential advantages in POF therapy; however, the detailed mechanisms need to be further investigated. Furthermore, this study will provide a reference for the clinical treatment strategy of POF.


Assuntos
Apoptose , Autofagia , Modelos Animais de Doenças , Células da Granulosa , Células-Tronco Mesenquimais , Estresse Oxidativo , Insuficiência Ovariana Primária , Esferoides Celulares , Feminino , Animais , Ratos , Células da Granulosa/patologia , Células da Granulosa/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/induzido quimicamente , Humanos , Transplante de Células-Tronco Mesenquimais , Ratos Sprague-Dawley , Cordão Umbilical/citologia , Células Cultivadas
2.
Ecotoxicol Environ Saf ; 276: 116319, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38615642

RESUMO

Di-hexyl phthalate (2-ethylhexyl) (DEHP) has been confirmed to cause female reproductive toxicity in humans and model animals by affecting the survival of ovarian granulosa cells (GCs), but the interrelationships between DEHP's on autophagy, apoptosis, and inflammation in GCs are not clear. Our previous study demonstrated that DEHP exposure resulted in the disturbance of intestinal flora associated with serum LPS release, which in turn led to impaired ovarian function. LPS has also been shown to determine cell fate by modulating cellular autophagy, apoptosis, and inflammation. Therefore, this study investigated the role and link between LPS and autophagy, apoptosis, and inflammation of GCs in DEHP-induced ovarian injury. Here, we constructed an in vivo injury model by continuous gavage of 0-1500 mg/kg of DEHP in female mice for 30 days and an in vitro injury model by treatment of human ovarian granulosa cells (KGN) cells with mono-2- ethylhexyl ester (MEHP, an active metabolite of DEHP in vivo). In addition, the expression of relevant pathway molecules was detected by immunohistochemistry, immunofluorescence, qRT-PCR, and Western blotting after the addition of the autophagy inhibitor 3-methyladenine (3-MA), the apoptosis inhibitor Z-VAD- FMK and the NF-κB inhibitor BAY11-7082. The current study found that autophagy and apoptosis were significantly activated in GCs of DEHP-induced atretic follicles in vivo and found that MEHP-induced KGN cells autophagy and apoptosis were independent and potentially cytotoxic of each other in vitro. Further studies confirmed that DEHP exposure resulted in LPS release from the intestinal tract and entering the ovary, thereby participating in DEHP-induced inflammation of GCs. In addition, we found that exogenous LPS synergized with MEHP could activate the NF-κB signaling pathway to induce inflammation and apoptosis of GCs in a relatively prolonged exposure condition. Meanwhile, inhibition of inflammatory activation could rescue apoptosis and estrogen secretion function of GCs induced by MEHP combined with LPS. These results indicated that the increased LPS influenced by DEHP might cooperate with MEHP to induce inflammatory apoptosis of GCs, an important cause of ovarian injury in mice.


Assuntos
Apoptose , Autofagia , Dietilexilftalato , Dietilexilftalato/análogos & derivados , Células da Granulosa , Inflamação , Lipopolissacarídeos , Feminino , Animais , Dietilexilftalato/toxicidade , Autofagia/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Lipopolissacarídeos/toxicidade , Apoptose/efeitos dos fármacos , Camundongos , Inflamação/induzido quimicamente , Inflamação/patologia , Reprodução/efeitos dos fármacos , Humanos
3.
Proc Natl Acad Sci U S A ; 117(9): 4792-4801, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32075915

RESUMO

Angiogenesis, the formation of new blood vessels, is tightly regulated by gene transcriptional programs. Yin Ying 1 (YY1) is a ubiquitously distributed transcription factor with diverse and complex biological functions; however, little is known about the cell-type-specific role of YY1 in vascular development and angiogenesis. Here we report that endothelial cell (EC)-specific YY1 deletion in mice led to embryonic lethality as a result of abnormal angiogenesis and vascular defects. Tamoxifen-inducible EC-specific YY1 knockout (YY1iΔEC ) mice exhibited a scarcity of retinal sprouting angiogenesis with fewer endothelial tip cells. YY1iΔEC mice also displayed severe impairment of retinal vessel maturation. In an ex vivo mouse aortic ring assay and a human EC culture system, YY1 depletion impaired endothelial sprouting and migration. Mechanistically, YY1 functions as a repressor protein of Notch signaling that controls EC tip-stalk fate determination. YY1 deficiency enhanced Notch-dependent gene expression and reduced tip cell formation. Specifically, YY1 bound to the N-terminal domain of RBPJ (recombination signal binding protein for Ig Kappa J region) and competed with the Notch coactivator MAML1 (mastermind-like protein 1) for binding to RBPJ, thereby impairing the NICD (intracellular domain of the Notch protein)/MAML1/RBPJ complex formation. Our study reveals an essential role of endothelial YY1 in controlling sprouting angiogenesis through directly interacting with RBPJ and forming a YY1-RBPJ nuclear repression complex.


Assuntos
Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Morfogênese/fisiologia , Neovascularização Patológica/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Proteínas Nucleares , Ligação Proteica , Receptores Notch/metabolismo , Vasos Retinianos/metabolismo , Transdução de Sinais , Fatores de Transcrição , Fator de Transcrição YY1/genética
4.
Ecotoxicol Environ Saf ; 265: 115534, 2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37776821

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP) is widely used in various plastics but has been demonstrated to cause female reproductive toxicity. However, the exact mechanism underlying the ovarian damage induced by DEHP remains unclear. In this study, DEHP was administered orally to 5-week-old female mice for 30 days at doses of 0, 250, 500, and 1000 mg/kg/day. The findings demonstrated that DEHP exposure disrupted ovarian function and follicular development as well as induced oxidative stress and autophagy in ovarian granulosa cells (GCs). Further, 200 µM mono-(2-ethylhexyl) phthalate (MEHP), the primary metabolite of DEHP in vivo, induced autophagy in both human ovarian granulosa cells line (KGN) and mouse primary GCs within 24 h in vitro. However, it did not affect the p62-dependent autophagy flux. Furthermore, MEHP-induced autophagy was inhibited by the autophagy inhibitor 3-MA and exacerbated by the autophagy activator rapamycin, indicating that MEHP induces excessive autophagy in GCs. Subsequently, we found that MEHP-induced autophagic cell death was primarily attributed to oxidative damage from elevated intracellular ROS levels. Meanwhile, MEHP exposure induced nuclear translocation of erythroid-derived factor 2-related factor (Nrf2), a key regulator of antioxidant activity resulting in activating antioxidant effects. Interestingly, we also found that MEHP-induced increase in p62 competitively binds Keap1, thereby facilitating nuclear translocation of Nrf2 and establishing a positive feedback loop in antioxidant regulation. Therefore, this study demonstrated that inhibition of Nrf2 could aggravate oxidative damage and enhance excessive autophagy caused by MEHP, while activation of Nrf2 could reverse the trend. These findings have also been reinforced in studies of cultured ovaries in vitro. Our study suggests that the p62-Keap1-Nrf2 pathway may serve as a potential protective mechanism against DEHP-induced oxidative stress and excessive autophagy in mouse GCs.

5.
Environ Toxicol ; 38(6): 1277-1291, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36880397

RESUMO

Microplastics (MPs) and nanoplastics (NPs) are novel environmental pollutants that are ubiquitous in the environment and everyday life. NPs can easily enter the tissues and have more significant potential health risks due to their smaller diameter. Previous studies have shown that NPs can induce male reproductive toxicity, but the detailed mechanisms remain uncertain. In this study, intragastric administration treated mice with polystyrene NPs (PS-NPs, 50, and 90 nm) at 3 and 15 mg/mL/day doses for 30 days. Then, the fresh fecal samples were collected from those mice that the exposure doses of 50 nm PS-NPs at 3 mg/mL/day and 90 nm at 15 mg/mL/day for subsequent investigations of 16S rRNA and metabolomics according to significant toxicological effects (Sperm number, viability, abnormality, and testosterone level). The conjoint analysis findings indicated that PS-NPs disrupted the homeostasis of the gut microbiota, metabolism, and male reproduction, suggesting that abnormal gut microbiota-metabolite pathways may be important in PS-NPs-induced male reproductive toxicity. Meanwhile, the common differential metabolites such as 4-deoxy-Erythronic acid, 8-iso-15-keto-PGE2, apo-10'-violaxanthin, beta-D-glucosamine, isokobusone, oleamide, oxoadipic acid, sphingosine induced by 50 and 90 nm PS-NPs might be used as biomarkers to explore PS-NPs-induced male reproductive toxicity. In addition, this study systematically demonstrated that nano-scale PS-NPs induced male reproductive toxicity via the crosstalk of gut microbiota and metabolites. It also provided valuable insights into the toxicity of PS-NPs, which was conducive to reproductive health risk assessment for public health prevention and treatment.


Assuntos
Microbiota , Poluentes Químicos da Água , Masculino , Animais , Camundongos , Microplásticos , Poliestirenos/toxicidade , Plásticos/toxicidade , RNA Ribossômico 16S , Sêmen , Metaboloma
6.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36769070

RESUMO

Female infertility is caused by premature ovarian failure (POF), which is triggered by the endoplasmic reticulum (ER) stress-mediated apoptosis of granulosa cells. The ER unfolded protein response (UPRer) is initiated to promote cell survival by alleviating excessive ER stress, but cellular apoptosis is induced by persistent or strong ER stress. Recent studies have reported that reticulophagy is initiated by ER stress. Whether reticulophagy is activated in the ER stress-mediated apoptosis of granulosa cells and which pathway is initiated to activate reticulophagy during the apoptosis of granulosa cells are unknown. Therefore, the role of reticulophagy in granulosa cell death and the relationship between ER stress and reticulophagy were investigated in this work. Our results suggest that the ER stress inducer tunicamycin causes POF in mice, which is attributed to the apoptosis of granulosa cells and is accompanied by the activation of UPRer and reticulophagy. Furthermore, granulosa cells were treated with tunicamycin, and granulosa cell apoptosis was triggered and increased the expression of UPRer and reticulophagy molecules. The expression of ATF4 was then downregulated by RNAi, which decreased the levels of autophagy and the reticulophagy receptor CCGP1. Furthermore, ATF4 targets MAP1LC3A, as revealed by the ChIP sequencing results, and co-IP results demonstrated that MAP1LC3A interacts with CCPG1. Therefore, reticulophagy was activated by ER stress through the ATF4-MAP1LC3A-CCPG1 pathway to mitigate ER stress. Additionally, the role of reticulophagy in granulosa cells was investigated by the knockdown of CCPG1 with RNAi. Interestingly, only a small number of granulosa cells died by apoptosis, whereas the death of most granulosa cells occurred by necroptosis triggered by STAT1 and STAT3 to impair ER proteostasis and the ER protein quality control system UPRer. Taken together, the results indicate that the necroptosis of granulosa cells is triggered by up- and downregulating the reticulophagy receptor CCPG1 through STAT1/STAT3-(p)RIPK1-(p)RIPK3-(p)MLKL and that reticulophagy is activated by ER stress through the ATF4-MAP1LC3A-CCPG1 pathway.


Assuntos
Estresse do Retículo Endoplasmático , Necroptose , Feminino , Camundongos , Animais , Tunicamicina/farmacologia , Resposta a Proteínas não Dobradas , Autofagia/genética , Apoptose , Células da Granulosa
7.
Hum Mol Genet ; 29(14): 2451-2459, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32469048

RESUMO

Rare coding variants have been proven to be one of the significant factors contributing to spermatogenic failure in patients with non-obstructive azoospermia (NOA) and severe oligospermia (SO). To delineate the molecular characteristics of idiopathic NOA and SO, we performed whole-exome sequencing of 314 unrelated patients of Chinese Han origin and verified our findings by comparing to 400 fertile controls. We detected six pathogenic/likely pathogenic variants and four variants of unknown significance, in genes known to cause NOA/SO, and 9 of which had not been earlier reported. Additionally, we identified 20 novel NOA candidate genes affecting 25 patients. Among them, five (BRDT, CHD5, MCM9, MLH3 and ZFX) were considered as strong candidates based on the evidence obtained from murine functional studies and human single-cell (sc)RNA-sequencing data. These genetic findings provide insight into the aetiology of human NOA/SO and pave the way for further functional analysis and molecular diagnosis of male infertility.


Assuntos
Azoospermia/genética , Predisposição Genética para Doença , Infertilidade Masculina/genética , Oligospermia/genética , Adulto , Animais , Azoospermia/patologia , DNA Helicases/genética , Humanos , Infertilidade Masculina/patologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Proteínas de Manutenção de Minicromossomo/genética , Proteínas MutL/genética , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Oligospermia/patologia , Espermatogênese/genética , Sequenciamento do Exoma
8.
Endocr J ; 69(7): 863-875, 2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35228471

RESUMO

Polycystic ovary syndrome (PCOS) is a common gynecological disease accompanied by a variety of clinical features, including anovulation, hyperandrogenism, and ovarian abnormalities, resulting in infertility. PCOS affects approximately 6%-15% of all reproductive-age women worldwide. Metformin, a popular drug used to treat PCOS in patients, has beneficial effects in reducing hyperandrogenism and inducing ovulation; however, the mechanisms by which metformin ameliorates PCOS are not clear. Hence, we aimed to explore the mechanisms of metformin in treating PCOS. In the present study, we first treated a letrozole-induced PCOS rat model with metformin, detected the pathological recovery of PCOS, and then assessed the effects of metformin on H2O2-induced autophagy in ovarian granulosa cells (GCs) by detecting the level of oxidative stress and the expression of autophagy-associated proteins and key proteins in the PI3K/AKT/mTOR pathway. We demonstrated that metformin ameliorated PCOS in a rat model by downregulating autophagy in GCs, and metformin decreased the levels of oxidative stress and autophagy in H2O2-induced GCs and affected the PI3K/AKT/mTOR signaling pathway. Taken together, our results indicate that metformin ameliorates PCOS in a rat model by decreasing excessive autophagy in GCs via the PI3K/AKT/mTOR pathway, and this study provides evidence for targeted reduction of excessive autophagy of ovarian granulosa cells and improvement of PCOS.


Assuntos
Hiperandrogenismo , Metformina , Síndrome do Ovário Policístico , Animais , Autofagia , Feminino , Células da Granulosa , Humanos , Peróxido de Hidrogênio/metabolismo , Hiperandrogenismo/complicações , Metformina/farmacologia , Metformina/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Serina-Treonina Quinases TOR/metabolismo
9.
Hepatology ; 72(5): 1717-1734, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32048304

RESUMO

BACKGROUND AND AIMS: Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS: This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS: PKD3 deletion in mice drives LF through the profibrotic macrophage activation.


Assuntos
Cirrose Hepática Experimental/patologia , Cirrose Hepática/patologia , Proteína Quinase C/deficiência , Animais , Tetracloreto de Carbono/toxicidade , Células Cultivadas , Progressão da Doença , Células Estreladas do Fígado/metabolismo , Humanos , Fígado/citologia , Fígado/patologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/diagnóstico , Cirrose Hepática Experimental/genética , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Miofibroblastos/metabolismo , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Índice de Gravidade de Doença , Análise Serial de Tecidos , Fator de Crescimento Transformador beta/metabolismo
10.
Environ Toxicol ; 36(6): 1226-1242, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33665894

RESUMO

Di-(2-ethylhexyl) phthalate (DEHP) is one of the most commonly used plasticizers, and it is widely applied in various plastic products. DEHP is an endocrine-disrupting chemical (EDC) that has been shown to disrupt the function of reproductive system in females. Although many studies have shown that DEHP potentially causes female reproductive toxicity, including depletion of the primordial follicle and decreased sex hormone production, the specific mechanisms by which DEHP affects female reproduction remain unknown. In recent years, research focused on the intestinal flora has provided an idea to eliminate our confusion, and gut bacterial dysbiosis may contribute to female reproductive toxicity. In the present study, the feces of DEHP-exposed mice were collected and analyzed using 16S rRNA amplicon sequencing and untargeted global metabolite profiling of metabolomics. DEHP obviously causes reproductive toxicity, including the ovarian organ coefficient, estradiol level, histological features of the ovary and estrus. Furthermore, DEHP exposure alters the structure of the intestinal microbiota community and fecal metabolite profile in mice, suggesting that the reproductive toxicity may be caused by gut bacterial dysbiosis and altered metabolites, such as changes in the levels of short-chain fatty acid (SCFA). Additionally, it is well known that changes in gut microbiota and fecal metabolites cause inflammation and tissue oxidative stress, expectedly, we found oxidative stress in the ovary and systemic inflammation in DEHP exposed mice. Thus, based on our findings, DEHP exposure may cause gut bacterial dysbiosis and altered metabolite profiles, particularly SCFA profiles, leading to oxidative stress in the ovary and systemic inflammation to ultimately induce female reproductive toxicity.


Assuntos
Dietilexilftalato , Microbioma Gastrointestinal , Animais , Dietilexilftalato/toxicidade , Fezes , Feminino , Camundongos , Ácidos Ftálicos , RNA Ribossômico 16S , Reprodução
11.
Biochem Biophys Res Commun ; 526(2): 424-430, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32228888

RESUMO

Cryopreservation and transplantation of the ovarian tissue is an alternative method by which malignant tumor survivors can recover fertility. Previously, it was reported that follicle stimulating hormone (FSH) promoted the survival and functioning of the ovarian tissue after in vitro cultivation. In this study, the expression of the luteinizing hormone receptor (LHR) was observed on the granule cell membrane after luteinizing hormone (LH) (0.3 IU/mL) was supplied as an exogenous hormone into the cultivation medium during ovarian vitrification in the postnatal period (PND) (1, 7, 14, 21, 28, 42, and 56 days PND). The expression of vascular endothelial growth factor (VEGF) and Connexins (Cx), and the recovery of ovarian functions were then assessed in mice models. The results showed that LH increased the production of normal follicles, and upregulated the expression of VEGF, Cx37, and Cx43 in vitrified ovaries. LH administration also shortened the recovery time of the estrus cycle in mice models. Additionally, no difference was observed in the rate of pregnancy and size of the first litter between the experimental and control groups. In conclusion, LH could promote the survival and functioning of the ovaries by upregulating the expression of VEGF, Cx43, and Cx37 during ovarian cryopreservation and transplantation.


Assuntos
Criopreservação , Hormônio Luteinizante/metabolismo , Ovário/fisiologia , Ovário/transplante , Animais , Feminino , Masculino , Camundongos , Ovário/citologia , Gravidez , Transplante
12.
Clin Proteomics ; 17: 15, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32489333

RESUMO

BACKGROUND: Human umbilical cord-derived MSCs (hUC-MSCs) have been identified as promising seeding cells in tissue engineering and clinical applications of regenerative medicine due to their advantages of simple acquisition procedure and the capability to come from a young tissue donor over the other MSCs sources. In clinical applications, large scale production is required and optimal cryopreservation and culture conditions are essential to autologous and allogeneic transplantation in the future. However, the influence of cryopreserved post-thaw and long-term culture on hUC-MSCs remains unknown, especially in terms of specific protein expression. Therefore, biological characteristics and proteomic profiles of hUC-MSCs after cryopreserving and long-term culturing were investigated. METHODS: Firstly, hUC-MSCs were isolated from human umbilical cord tissues and identified through morphology, surface markers and tri-lineage differentiation potential at passage 3, and then the biological characteristics and proteomic profiles were detected and compared after cryopreserving and long-term culturing at passage 4 and continuously cultured to passage 10 with detection occurring here as well. The proteomic profiles were tested by using the isobaric tags for relative and absolute quantification (iTRAQ) labeling technique and differential protein were confirmed by mass spectrometry. RESULTS: The results showed no significant differences in phenotypes including morphology, surface marker and tri-lineage differentiation potential but have obvious changes in translation level, which is involved in metabolism, cell cycle and other pathways. CONCLUSION: This suggests that protein expression may be used as an indicator of hUC-MSCs security testing before applying in clinical settings, and it is also expected to provide the foundation or standardization guide of hUC-MSCs applications in regenerative medicine.

13.
Biol Pharm Bull ; 42(8): 1282-1294, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31105116

RESUMO

The purpose of the paper is to study the differences in cell death mechanism of MGC-803 induced by "dextran-magnetic layered double hydroxide-fluorouracil" (DMF) drug delivery system and 5-Fluorouracil (5-Fu), respectively. The inhibitory effect on the proliferation was detected via CCK-8. The morphology of cell death was detected by transmission electron microscopy (TEM). Intracellular ATP, mitochondrial membrane potential (MMP), reactive oxygen species (ROS) and Cytosolic Free Ca (Ca2+) level were detected via some methods. The result showed that DMF had more obvious effect in suppressing proliferation compared with 5-Fu, and changed cell death pattern of 5-Fu from apoptosis to oncosis. The ATP decrease, MMP loss, Ca2+ increase, the activation of uncoupling protein-2 (UCP-2) and calpain-1 were significant after DMF exposure. However, DMF did not result in ROS accumulation. DMF could involve in activation of porimin, and the cascade reaction of caspases-3, -7, -9, and -12 and poly ADP-ribose polymerase (PARP) through Western blot. DMF showed a stronger injury on nuclear membrane in the cascade reaction of caspases-6, -8 and lamin-A. DMF triggered rapid depletion of ATP, which was consistent with the phenotype of oncosis. Endogenous mitochondrial apoptosis might not be the main cause of cell swelling. DMF could induce strong endoplasmic reticulum stress (ERS) effect, there might be some signaling pathways related with ERS during the process of oncosis. The calpain system might not be a key factor for structural damage in oncosis induced by DMF. DMF could induce the caspases cascade reactions similar to apoptosis, but inflicted a more strong damage on nuclear membrane and PARP.


Assuntos
Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dextranos/farmacologia , Fluoruracila/farmacologia , Hidróxidos/farmacologia , Apoptose/efeitos dos fármacos , Calpaína/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Lamina Tipo A/metabolismo , Mitocôndrias/metabolismo , Fenótipo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas
14.
Molecules ; 24(2)2019 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-30669620

RESUMO

Microglia mediate multiple facets of neuroinflammation. They can be phenotypically divided into a classical phenotype (pro-inflammatory, M1) or an alternative phenotype (anti-inflammatory, M2) with different physiological characteristics and biological functions in the inflammatory process. Betaine has been shown to exert anti-inflammatory effects. In this study, we aimed to verify the anti-inflammatory effects of betaine and elucidate its possible molecular mechanisms of action in vitro. Lipopolysaccharide (LPS)-activated microglial cells were used as an inflammatory model to study the anti-inflammatory efficacy of betaine and explore its mechanism of regulating microglial polarisation by investigating the morphological changes and associated inflammatory changes. Cytokine and inflammatory mediator expression was also measured by ELISA, flow cytometry, immunofluorescence, and western blot analysis. Toll-like receptor (TLR)-myeloid differentiation factor 88 (Myd88)-nuclear factor-kappa B (NF-κB) p65, p-NF-κB p65, IκB, p-IκB, IκB kinase (IKK), and p-IKK expression was determined by western blot analysis. Betaine significantly mitigated the production of pro-inflammatory cytokines and increased the release of anti-inflammatory cytokines. It promoted the conversion of the microglia from M1 to M2 phenotype by decreasing the expression of inducible nitric oxide synthase and CD16/32 and by increasing that of CD206 and arginase-1. Betaine treatment inhibited the TLR4/NF-κB pathways by attenuating the expression of TLR4-Myd88 and blocking the phosphorylation of IκB and IKK. In conclusion, betaine could significantly alleviate LPS-induced inflammation by regulating the polarisation of microglial phenotype; thus, it might be an effective therapeutic agent for neurological disorders.


Assuntos
Betaína/farmacologia , Lipopolissacarídeos/imunologia , Microglia/efeitos dos fármacos , Microglia/fisiologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Óxido Nítrico , Fenótipo
15.
J Transl Med ; 16(1): 38, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29471858

RESUMO

BACKGROUND: Breast cancer is one of the most frequent malignancies and the second leading cause of cancer-related mortality in women. MicroRNAs play a key role in breast cancer development and progression. microRNA(miR)-8084 has been observed an aberrant expression in breast cancer. However, the functions and regulatory axes of miR-8084, particularly in breast cancer, were not entirely clear. METHODS: miR-8084 expression in breast cancer were investigated in a GEO dataset by in silico analysis and in 42 paired tumor tissues by qPCR. The effects of deregulation of miR-8084 on breast cancer cell proliferation, migration and invasion in vitro and tumorigenicity in vivo were examined by colony-formation assay, wound healing assay, transwell assay and nude mouse subcutaneous tumor formation model. The target gene of miR-8084 were predicted by TargetScan and miRDB, and confirmed by luciferase reporter system. The roles of miR-8084 in the breast cancer cell proliferation, apoptosis and epithelial-mesenchymal transition (EMT) were investigated by MTS, FACS and associated-marker detection by western blot. RESULTS: miR-8084 is significantly up-regulated in both serum and malignant tissues from the source of breast cancer patients. miR-8084 promotes the proliferation of breast cancer cells by activating ERK1/2 and AKT. Meanwhile miR-8084 inhibits apoptosis by decreasing p53-BAX related pathway. miR-8084 also enhances migration and invasion by inducing EMT. Moreover, the tumor suppressor ING2 is a potential target of miR-8084, and miR-8084 regulatory axes contribute to pro-tumor effect, at least partially through regulating ING2. CONCLUSION: Our results strongly suggest that miR-8084 functions as an oncogene that promotes the development and progression of breast cancer, and miR-8084 is a potential new diagnostic marker and therapeutic target of breast cancer.


Assuntos
Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Células Clonais , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima/genética , Cicatrização , Proteína X Associada a bcl-2/metabolismo
16.
Cell Biochem Funct ; 35(3): 184-191, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28436141

RESUMO

MicroRNAs (miRs) are functionally important in spermatogenesis, which is the self-renewal or differentiation of spermatogonial stem cells (SSCs). Here, we report a novel role for miR-10b in regulating the self-renewal of mouse SSCs. We showed that miR-10b was highly expressed in mouse SSCs in vitro and enhanced SSC proliferation. Knockdown of miR-10b significantly increased the apoptosis of SSCs compared with controls. Kruppel-like factor 4 was found to be a target gene of miR-10b in the enhancement of SSC proliferation. These findings further our understanding of the self-renewal and differentiation of SSCs and provide a basis for the diagnosis, treatment, and prevention of male infertility.


Assuntos
Diferenciação Celular , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs/genética , Espermatogênese/fisiologia , Espermatogônias/citologia , Células-Tronco/citologia , Testículo/citologia , Animais , Apoptose , Proliferação de Células , Autorrenovação Celular , Células Cultivadas , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogônias/metabolismo , Células-Tronco/metabolismo , Testículo/metabolismo
17.
Biol Reprod ; 94(6): 137, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26911431

RESUMO

Ovarian cryopreservation by vitrification is a highly useful method for preserving female fertility during radiotherapy and chemotherapy. However, cryoinjury, osmotic stress during vitrification, and ischemia/reperfusion during transplantation lead to loss of ovarian follicles. Ovarian follicle loss may be partially reduced by several methods; however, studies regarding the mechanism of ovarian follicle loss have only investigated cell apoptosis, which consists of type I programmed cell death. Autophagy is type II programmed cell death, and cell homeostasis is maintained by autophagy during conditions of stress. The role of autophagy during cryopreservation by vitrification has rarely been reported. The potential role of autophagy during ovarian cryopreservation by vitrification is reviewed in this article.


Assuntos
Autofagia , Criopreservação , Ovário , Vitrificação , Feminino , Humanos , Ovário/transplante
18.
Cell Tissue Res ; 363(3): 589-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26022337

RESUMO

Endoplasmic reticulum stress (ERS) activates a protective pathway, called the unfold protein response, for maintaining cellular homeostasis, but cellular apoptosis is triggered by excessive or persistent ERS. Several recent studies imply that the ERS response might have broader physiological roles in the various reproductive processes of female mammals, including embryo implantation, decidualization, preimplantation embryonic development, follicle atresia, and the development of the placenta. This review summarizes the existing data concerning the molecular and biological roles of the ERS response. The study of the functions of the ERS response in mammalian reproduction might provide novel insights into and an understanding of reproductive cell survival and apoptosis under physiological and pathological conditions. The ERS response is a novel signaling pathway for reproductive cell survival and apoptosis. Infertility might be a result of disturbing the ERS response during the process of female reproduction.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Mamíferos/fisiologia , Reprodução/fisiologia , Animais , Decídua/fisiologia , Implantação do Embrião , Feminino , Atresia Folicular/fisiologia , Humanos , Mamíferos/embriologia
19.
Zhonghua Nan Ke Xue ; 22(1): 6-11, 2016 Jan.
Artigo em Zh | MEDLINE | ID: mdl-26931018

RESUMO

OBJECTIVE: To investigate the effects of single heat stress treatment on spermatogenic cells in mice. METHODS: We randomly divided 36 C57 male mice into a control and a heat stress treatment group and submerged the lower part of the torso in water at 25 °C and 43 °C, respectively, both for 15 minutes. At 1, 7, and 14 days after treatment, we obtained the testicular organ indexes, observed the changes in testicular morphology by HE staining, and determined the location and expression levels of the promyelocytic leukemia zinc finger (PLZF) and synaptonemal comlex protein-3 (SCP-3) in the testis tissue by immunohistochemistry and Western blot. RESULTS: The testicular organ index was significantly lower in the heat stress treatment than in the control group (P < 0.05). Compared with the controls, the heat shock-treated mice showed loosely arranged spermatogenic cells scattered in the seminiferous tubules at 1 day after heat stress treatment, atrophied, loosely arranged and obviously reduced number of spermatogenic cells at 7 days, and relatively closely arranged seminiferous tubules and increased number and layers of spermatogenic cells at 14 days. The number of SCP-3 labelled spermatocytes obviously decreased in the heat stress-treated animals at 1 and 7 days and began to increase at 14 days. The PLZF protein expression was significantly reduced in the heat stress treatment group at 1 day as compared with that in the control (0.19 ± 0.12 vs 0.64 ± 0.03, P < 0.01), but elevated to 0.77 ± 0.02 at 7 and 14 days, even remarkably higher than in the control animals (P < 0.01). CONCLUSION: Heat stress treatment can induce short-term dyszoospermia in mice, which can be recovered with the prolonged time after treatment.


Assuntos
Temperatura Alta , Proteínas Nucleares/metabolismo , Espermatócitos/patologia , Testículo/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Western Blotting , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Imuno-Histoquímica , Masculino , Camundongos , Proteína da Leucemia Promielocítica , Túbulos Seminíferos/citologia , Espermatócitos/citologia
20.
J Immunoassay Immunochem ; 36(2): 170-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24785531

RESUMO

In this study, the spatiotemporal expression of SerpinB11 in the mouse testis from postnatal 1-60 d was checked, the SerpinB11 protein strongly localized in the intermediate spermatogonia, B-type spermatogonium, preleptotene spermatocyte, leptonema spermatocyte, zygotene spermatocyte, but weakly localized in the pachytene spermatocyte, diplotene spermatocyte, sphere sperm, and the apoptotic sperm was positive stained of SerpinB11 protein, the localization of cell cycle marker CDK4 and meiosis marker SCP3 were investigated, and the SCP3 and SerpinB11 colocalized in the intermediate spermatogonia, B-type spermatogonium, preleptotene spermatocyte. Taken together, these results suggested that SerpinB11 might involved in spermatogenesis and apoptosis.


Assuntos
Apoptose/fisiologia , Serpinas/metabolismo , Espermatogênese/fisiologia , Testículo/metabolismo , Animais , Proteínas de Ciclo Celular , Quinase 4 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA , Masculino , Camundongos Endogâmicos ICR , Proteínas Nucleares/metabolismo , Espermatozoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA