Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell Tissue Res ; 394(2): 309-323, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37572164

ABSTRACT

Subclinical hypothyroidism (SCH) affects 10% of the global population, which is most prevalent in women and the elderly. However, it remains debatable whether the elderly with subclinical hypothyroidism needs thyroxine supplement. Human amnion-derived mesenchymal stem cells (hAMSCs) could play important roles in autoimmune diseases, suggesting that hAMSC be a candidate to regulate the thyroid function of female age-related subclinical hypothyroidism. Herein, we established the model of SCH in the aged female mice. This study was designed to investigate whether human amnion-derived mesenchymal stem cells (hAMSC) could effect on immune regulation, apoptosis inhibition of thyroid cells, thyroid function, blood lipid levels, and heart function. In addition, qualified hAMSCs were intravenously injected into aged female SCH mice via the tail vein on day 0 and day 10. The levels of thyroid hormone and blood lipids as well as cardiac function, serum immunological indexes, and apoptosis of thyroid cells were then analyzed on day 5, 10, 15, and 20; meanwhile, the quantity of Th1, Th2, Th17, and Treg immune cells in peripheral blood was evaluated before and on day 20 post-injection. Our study demonstrated that after hAMSC transplantation, the thyroid functions, blood lipid levels, and heart function indexes of age-related SCH (AR-SCH) mice were significantly improved. Consistent with this, Th1 and Treg cells increased significantly, while Th2 and Th17 cells decreased in peripheral blood. Apoptosis was also suppressed in the thyroid cells. In summary, hAMSC delivery can potentially be a safe and effective therapy for treating SCH in the elderly, improving related complications by immunomodulatory and apoptosis inhibition.


Subject(s)
Hypothyroidism , Mesenchymal Stem Cells , Aged , Humans , Female , Mice , Animals , Amnion , Hypothyroidism/therapy , Apoptosis , Lipids , Immunocompetence
2.
Ren Fail ; 45(1): 2218483, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37293809

ABSTRACT

Calciphylaxis is a rare cutaneous vascular disease that manifests with intolerable pains, non-healing skin wounds, histologically characterized by calcification, fibrointimal hyperplasia, and microvessel thrombosis. Currently, there are no standardized guidelines for this disease. Recent studies have recognized a high prevalence of thrombophilias and hypercoagulable conditions in calciphylaxis patients. Here, we report a case of uremic calciphylaxis patient whom was refractory to conventional treatments and then received a salvage strategy with intravenous and local hAMSC application. In order to investigate the therapeutic mechanism of hAMSCs from the novel perspective of hypercoagulability, coagulation-related indicators, wound status, quality of life and skin biopsy were followed up. Polymerase chain reaction (PCR) was performed to determine the distribution of hAMSCs in multiple tissues including lung, kidney and muscle after infusion of hAMSCs for 24 h, 1 week and 1 month in mice aiming to investigate whether hAMSCs retain locally active roles after intravenous administration. Improvement of hypercoagulable condition involving correction of platelet, D-dimer and plasminogen levels, skin regeneration and pain alleviation were revealed after hAMSC administration over one-year period. Skin biopsy pathology suggested regenerative tissues after 1 month hAMSC application and full epidermal regeneration after 20 months hAMSC treatment. PCR analysis indicated that hAMSCs were homing in lung, kidney and muscle tissues of mice even until tail vein injection of hAMSCs for 1 month. We propose that hypercoagulability is a promising therapeutic target of calciphylaxis patients, which can be effectively improved by hAMSC treatment.


Subject(s)
Calciphylaxis , Mesenchymal Stem Cells , Thrombophilia , Humans , Mice , Animals , Amnion , Calciphylaxis/etiology , Calciphylaxis/therapy , Quality of Life , Thrombophilia/etiology
3.
Am J Respir Cell Mol Biol ; 67(1): 61-75, 2022 07.
Article in English | MEDLINE | ID: mdl-35507777

ABSTRACT

Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have been evaluated in many studies as promising therapeutic agents for pulmonary hypertension (PH). However, low yields and heterogeneity are major barriers in the translational utility of EVs for clinical studies. To address these limitations, we fabricated MSC-derived nanovesicles (MSC-NVs) by serial extrusion through filters, resulting in MSC-NVs with characteristics similar to conventional EVs but with much higher production yields. Herein, we examined the therapeutic efficacy of MSC-NVs in preclinical models of PH in vitro and in vivo. Intervention with MSC-NVs improved the core pathologies of monocrotaline-induced PH in rats. Intravenous administration of MSC-NVs resulted in significant uptake within hypertensive lungs, pulmonary artery lesions, and especially pulmonary artery smooth muscle cells (PASMCs). In vitro, MSC-NVs inhibited PDGF-induced proliferation, migration, and phenotype switching of PASMCs. miRNA-sequencing analysis of the genetic cargo of MSC-NVs revealed that miR-125b-5p and miR-100-5p are highly abundant, suggesting that they might account for the therapeutic effects of MSC-NVs in PH. Depletion of miR-125b-5p and miR-100-5p in MSCs almost completely abolished the beneficial effects of MSC-NVs in protecting PASMCs from PDGF-stimulated changes in vitro and also diminished the protective effects of MSC-NVs in monocrotaline-induced PH in vivo. These data highlight the efficacy and advantages of MSC-NVs over MSC-EVs as a promising therapeutic strategy against PH.


Subject(s)
Extracellular Vesicles , Hypertension, Pulmonary , Mesenchymal Stem Cells , MicroRNAs , Animals , Disease Models, Animal , MicroRNAs/genetics , Monocrotaline , Rats
4.
Biochem Biophys Res Commun ; 636(Pt 1): 155-161, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36334439

ABSTRACT

BACKGROUND: Peroxiredoxin 4 (Prdx4) in the endoplasmic reticulum (ER) is the only secretory member of the antioxidant Prdx family. Our previous studies demonstrated that Prdx4 in cumulus cells (CCs) ameliorated the maturation of oocytes in vitro and enhanced oocyte developmental competence by preventing CCs apoptosis caused by oxidative stress (OS) through gap junctions. In this study, we aimed to determine whether Prdx4 released by CCs can repair meiotic defects in mouse oocytes by co-culturing immature (germinal vesicle) oocytes with CCs from mature oocytes in the absence of gap junctions. RESULTS: The OS-induced meiotic defects in mouse oocytes were impeded by co-culture with CCs, as evidenced by the increased first polar body (PB1) extrusion rate and decreased ROS level. CCs increased Prdx4 expression and lowered IRE1α, Bip expression in H2O2-treated oocytes. After knockdown of Prdx4 expression in CCs, the rate of PB1 extrusion in the oocytes was significantly reduced to the level detected in H2O2 group, and ER stress was not alleviated. CO-IP and immunofluorescence co-localization experiments demonstrated that Prdx4 interacted with PDIA6 in the oocytes and the Pearson's R value was 0.69 calculated using ImageJ. CONCLUSIONS: Cumulus cells can promote the maturation of oocytes in vitro by secreting Prdx4 in a paracrine manner and serve as a promising therapeutic antioxidant for improving the quality of oocytes, especially aging oocytes, in clinical in vitro maturation (IVM).


Subject(s)
Cumulus Cells , In Vitro Oocyte Maturation Techniques , Peroxiredoxins , Animals , Female , Mice , Antioxidants/metabolism , Endoribonucleases/metabolism , Hydrogen Peroxide/metabolism , Oocytes/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Protein Serine-Threonine Kinases
5.
Mol Hum Reprod ; 28(6)2022 05 27.
Article in English | MEDLINE | ID: mdl-35583302

ABSTRACT

Maternal-effect genes (MEGs) play an important role in maintaining the survival and development of mammalian embryos at the cleavage stage after fertilization. Despite long-term efforts, the MEGs that regulate preimplantation embryo development remain largely unknown. Here, using whole-exome sequencing and homozygosity mapping, we identified a potential candidate gene associated with early embryo development: nucleoporin37 (NUP37), a nucleoporin gene that encodes a member of the nuclear pore complexes and regulates nuclear pore permeability and nucleocytoplasmic transport. Moreover, we determined the temporal and spatial expression patterns of Nup37 in mouse oocytes and early embryos, and explored the role of NUP37 in oocyte maturation and preimplantation embryo development. Immunoprecipitation assays confirmed that yes-associated protein-1 (YAP1) binds to TEA domain transcription factor 4 (TEAD4) and NUP37. Furthermore, Nup37 gene knockdown reduced the nuclear import of YAP1 and down-regulated the expression of YAP1-TEAD pathway downstream genes Rrm2 and Rpl13 in early embryos. Our study provides evidence that maternal NUP37 contributes to the nuclear import of YAP1 and then activates the YAP1-TEAD pathway, a signalling pathway essential for zygotic genome activation. Nup37 may be a key gene involved in preimplantation embryo development in mammals.


Subject(s)
Embryonic Development , Zygote , Animals , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Humans , Mammals/genetics , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neoplasm Proteins/genetics , Oocytes/metabolism , Oogenesis , Ribosomal Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Reprod Fertil Dev ; 34(15): 957-970, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36031717

ABSTRACT

CONTEXT: Low ovarian putrescine levels and decreased peak values following luteinising hormone peaks are related to poor oocyte quantity and quality in ageing women. AIMS: To investigate the effects of putrescine supplementation in in vitro maturation (IVM) medium on oocyte quality and epigenetic modification. METHODS: Germinal vesicle oocytes retrieved from the ovaries of 8-week-old and 9-month-old mice were divided into four groups (the young, young+difluoromethylornithine (DFMO), ageing and ageing+putrescine groups) and cultured in IVM medium with or without 1mM putrescine or DFMO for 16h. The first polar body extrusion (PBE), cleavage and embryonic development were evaluated. Spindles, chromosomes, mitochondria and reactive oxygen species (ROS) were measured. The expression levels of SIRT1, H3K9ac, H3K9me2, H3K9me3, and 5mC levels were evaluated. Sirt1 and imprinted genes were detected. RESULTS: The PBE was higher in the ageing+putrescine group than in the ageing group. Putrescine increased the total and inner cell mass cell numbers of blastocysts in ageing oocytes. Putrescine decreased aberrant spindles and chromosome aneuploidy, increased the mitochondrial membrane potential and decreased ROS levels. Putrescine increased SIRT1 expression and attenuated the upregulation of H3K9ac levels in ageing oocytes. Putrescine did not affect 5mC, H3K9me2 or H3K9me3 levels or imprinted gene expression. CONCLUSIONS: Putrescine supplementation during IVM improved the maturation and quality of ageing oocytes and promoted embryonic development by decreasing ROS generation, maintaining mitochondrial and spindle function and correcting aberrant epigenetic modification. IMPLICATIONS: Putrescine shows application potential for human-assisted reproduction, especially for IVM of oocytes from ageing women.


Subject(s)
Putrescine , Sirtuin 1 , Animals , Eflornithine/metabolism , Eflornithine/pharmacology , Epigenesis, Genetic , Female , Humans , In Vitro Oocyte Maturation Techniques , Infant , Luteinizing Hormone/metabolism , Mice , Oocytes/metabolism , Pregnancy , Putrescine/pharmacology , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism
7.
Dev Dyn ; 250(7): 974-985, 2021 07.
Article in English | MEDLINE | ID: mdl-33449399

ABSTRACT

BACKGROUND: Genetic programs underlying preimplantation development and early lineage segregation are highly conserved across mammals. It has been suggested that nonhuman primates would be better model organisms for human embryogenesis, but a limited number of studies have investigated the monkey preimplantation development. In this study, we collect single cells from cynomolgus monkey preimplantation embryos for transcriptome profiling and compare with single-cell RNA-seq data derived from human and mouse embryos. RESULTS: By weighted gene-coexpression network analysis, we found that cynomolgus gene networks have greater conservation with human embryos including a greater number of conserved hub genes than that of mouse embryos. Consistently, we found that early ICM/TE lineage-segregating genes in monkeys exhibit greater similarity with human when compared to mouse, so are the genes in signaling pathways such as LRP1 and TCF7 involving in WNT pathway. Last, we tested the role of one conserved pre-EGA hub gene, SIN3A, using a morpholino knockdown of maternal RNA transcripts in monkey embryos followed by single-cell RNA-seq. We found that SIN3A knockdown disrupts the gene-silencing program during the embryonic genome activation transition and results in developmental delay of cynomolgus embryos. CONCLUSION: Taken together, our study provided new insight into evolutionarily conserved and divergent transcriptome dynamics during mammalian preimplantation development.


Subject(s)
Blastomeres/metabolism , Embryonic Development/genetics , Macaca fascicularis/embryology , Adult , Animals , Blastocyst , Blastomeres/cytology , Cell Lineage/genetics , Cells, Cultured , Embryo, Mammalian , Embryonic Development/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks/physiology , Humans , Macaca fascicularis/genetics , Macaca mulatta , Male , Mice , Pregnancy , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Sin3 Histone Deacetylase and Corepressor Complex/physiology , Single-Cell Analysis/veterinary , Transcriptome/genetics
8.
Zhonghua Nan Ke Xue ; 26(6): 487-498, 2020 Jun.
Article in Zh | MEDLINE | ID: mdl-33356036

ABSTRACT

OBJECTIVE: To observe the effects of different concentrations of testosterone on the differentiation of human embryonic stem cells (hESCs) into early male germ cells and investigate the potential impact of high-level androgen exposure in early pregnancy in women with polycystic ovary syndrome (PCOS) on the fertility and primordial germ cell reserve of the male offspring in adulthood. METHODS: We used 2 µmol/L retinoic acid to induce the differentiation of hESCs (46, XY) into male germ cells in vitro and meanwhile treated them with testosterone (T) at 0 mol/L, 3×10-7 mol/L, 5×10-7 mol/L, 15×10-7 mol/L, 45×10-7 mol/L, and 135×10-7 mol/L, respectively. We collected the cell samples at 0, 4, 7 and 14 days to determine the expressions of the specific genes and compare the differentiation process and efficiency of the male germ cells in different stages. RESULTS: There was no difference in the morphology of the hESCs treated with different concentrations of testosterone in the same differentiation stage. The expression of the marker gene DAZL in the primordial germ cells peaked on the 4th day of differentiation, significantly higher in the 15×10-7, 45×10-7 and 135×10-7 mol/L groups than in the 3×10-7 mol/L group (P < 0.05), and that of the specific gene SCP3 in the early-meiosis germ cells began to increase on the same day, more significantly in the 45×10-7mol/L than in the 3×10-7 mol/L and 5×10-7 mol/L groups (P < 0.01), and peaked on the 7th day, dramatically higher in the 15×10-7, 45×10-7 and 135×10-7 mol/L groups than in the 3×10-7 mol/L group (P < 0.01). Immunofluorescence staining and flow cytometry showed a T concentration-dependent increase in the expression of DAZL at 4 days and those of SCP3 and VASA at 7 days. Moreover, the expression of the androgen receptor (AR) in the hESCs began to rise on the 4th day and kept going up till the 14th day, higher in the high-concentration than in the low-concentration T groups in the same stage of differentiation, though with no statistically significant difference (P > 0.05). CONCLUSIONS: Exposure to high-level androgen during the differentiation of hESCs into early male germ cells can induce earlier expression of AR and earlier differentiation of hESCs into early male germ cells, which may result in insufficient reserve of male primary germ cells in the male offspring of PCOS women and affect their fertility after adulthood. hESCs can be used as an in vitro model to study the effects of intrauterine hyperandrogen on the reproductive development of male offspring in PCOS patients, which is also contributive to researches on the etiology of male infertility.


Subject(s)
Androgens/pharmacology , Cell Differentiation , Germ Cells/cytology , Human Embryonic Stem Cells/drug effects , Cell Cycle Proteins/physiology , Cells, Cultured , DEAD-box RNA Helicases/physiology , DNA-Binding Proteins/physiology , Human Embryonic Stem Cells/cytology , Humans , Male , Meiosis , RNA-Binding Proteins/physiology
9.
Zhonghua Nan Ke Xue ; 24(1): 6-13, 2018.
Article in Zh | MEDLINE | ID: mdl-30157353

ABSTRACT

OBJECTIVE: To assess the risk of male infertility in the offspring conceived through assisted reproductive technology (ART) byin vitroinductionof the differentiation of embryonic stem cells (ESCs) derived from the embryos of the couples with male asthenozoospermia and Robertsonian translocation (RT) into germ cells. METHODS: We established a CCRM16ESC line with the karyotype of 46, XY, +14, rob(13; 14) (q10; q10) from the embryo donated by a patientwithasthenozoospermiaand RT and his wife by isolation of the inner cell mass of blastula, culturing, passaging, and amplification,followed by in vitro induction and differentiationof the ESCs into germ cells with ratinoic acid(RA) at 2 mol/L. Then, we analyzed the process of differentiation and the expressions of its related genes and compared them with those in the normal CCRM23ESCs. RESULTS: CCRM16 showed the typical characteristics of ESCs, expressing the pluripotency makers of NANOG, OCT4, TRA-1-181 and SSEA4, forming embryoid bodies, and differentiating into three germlayer tissues in vitro and in vivo. Intervention with 2 mol/LRAinduced direct differentiation of the ESCs into germ cells. The expressions of the primordial germ cell marker geneDAZLand the meiosis marker geneSCP3were markedly decreased in the CCRM16 as compared with those in the normal CCRM23 ESCs. CONCLUSIONS: The CCRM16ESC linewith the karyotype of46, XY, +14, rob(13; 14) (q10; q10) has thetypical characteristics of ESCs but an abnormal process of differentiation into germ cells in the early stage. In vitroinductionof the differentiation of ESCs into germ cells can be used for assessing the risk of male infertility in the offspring conceived through ART for asthenozoospermia patients.


Subject(s)
Abnormal Karyotype , Asthenozoospermia/pathology , Blastocyst Inner Cell Mass , Cell Differentiation/genetics , Chromosomes, Human, 13-15/genetics , Embryonic Stem Cells/cytology , Germ Cells/cytology , Infertility, Male/etiology , Translocation, Genetic/genetics , Animals , Asthenozoospermia/genetics , Cell Line , Genetic Markers , Humans , Male , Reproductive Techniques, Assisted , Risk , Stage-Specific Embryonic Antigens
10.
Zhonghua Nan Ke Xue ; 23(5): 468-472, 2017 May.
Article in Zh | MEDLINE | ID: mdl-29717842

ABSTRACT

Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), have the potential of differentiating into all types of adult cells. Today, mature functional sperm can be derived from mouse PSCs in vitro, and meanwhile primordial germ cells (PGCs) and meiotic prophase sperm cells can be generated from human ESCs/iPSCs (hESCs/hiPSCs). It is proposed that non-genetic azoospermia might be cured if functional sperm could be obtained from human PSCs (hPSCs) in vitro. It is also possible that healthy functional sperm could be derived from the patient with genetic factor-induced azoospermia by combining iPSCs and gene editing technology. IPSC-derived functional sperm have a higher clinical value for the avoidance of the sperm source and the issue of medical ethics. This article summarizes recent advances in the differentiation of PSCs into male germ cells in vitro, aiming to provide some reference for the treatment of male infertility with PSCs.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Infertility, Male/therapy , Spermatozoa/cytology , Animals , Humans , Male , Meiosis , Mice , Pluripotent Stem Cells/cytology
11.
Stem Cell Res Ther ; 15(1): 115, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38650029

ABSTRACT

BACKGROUND: Studies have shown that chemotherapy and radiotherapy can cause premature ovarian failure and loss of fertility in female cancer patients. Ovarian cortex cryopreservation is a good choice to preserve female fertility before cancer treatment. Following the remission of the disease, the thawed ovarian tissue can be transplanted back and restore fertility of the patient. However, there is a risk to reintroduce cancer cells in the body and leads to the recurrence of cancer. Given the low success rate of current in vitro culture techniques for obtaining mature oocytes from primordial follicles, an artificial ovary with primordial follicles may be a good way to solve this problem. METHODS: In the study, we established an artificial ovary model based on the participation of mesenchymal stem cells (MSCs) to evaluate the effect of MSCs on follicular development and oocyte maturation. P2.5 mouse ovaries were digested into single cell suspensions and mixed with bone marrow derived mesenchymal stem cells (BM-MSCs) at a 1:1 ratio. The reconstituted ovarian model was then generated by using phytohemagglutinin. The phenotype and mechanism studies were explored by follicle counting, immunohistochemistry, immunofluorescence, in vitro maturation (IVM), in vitro fertilization (IVF), real-time quantitative polymerase chain reaction (RT-PCR), and Terminal-deoxynucleotidyl transferase mediated nick end labeling(TUNEL) assay. RESULTS: Our study found that the addition of BM-MSCs to the reconstituted ovary can enhance the survival of oocytes and promote the growth and development of follicles. After transplanting the reconstituted ovaries under kidney capsules of the recipient mice, we observed normal folliculogenesis and oocyte maturation. Interestingly, we found that BM-MSCs did not contribute to the formation of follicles in ovarian aggregation, nor did they undergo proliferation during follicle growth. Instead, the cells were found to be located around growing follicles in the reconstituted ovary. When theca cells were labeled with CYP17a1, we found some overlapped staining with green fluorescent protein(GFP)-labeled BM-MSCs. The results suggest that BM-MSCs may participate in directing the differentiation of theca layer in the reconstituted ovary. CONCLUSIONS: The presence of BM-MSCs in the artificial ovary was found to promote the survival of ovarian cells, as well as facilitate follicle formation and development. Since the cells didn't proliferate in the reconstituted ovary, this discovery suggests a potential new and safe method for the application of MSCs in clinical fertility preservation by enhancing the success rate of cryo-thawed ovarian tissues after transplantation.


Subject(s)
Mesenchymal Stem Cells , Oocytes , Ovary , Female , Animals , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Ovary/cytology , Oocytes/cytology , Oocytes/metabolism , Mesenchymal Stem Cell Transplantation/methods , Ovarian Follicle/metabolism , Ovarian Follicle/cytology
12.
J Cell Biochem ; 114(9): 2016-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23564289

ABSTRACT

A normal fertilized human zygote contains two pronuclei, but zygotes may also display one, three, or even more pronuclei resulting from irregular insemination or meiotic division. Today diploid and triploid human embryonic stem cell (hESC) lines have been derived from tripronuclear (3PN) triploid zygotes, and an in-vitro fertilization (IVF) baby was born from a rescued diploid zygote by removing the extra male pronucleus of the 3PN zygote. However, whether hESCs can be derived from a rescued 3PN zygote is still unknown. Here, by microsurgical pronuclear removal, we restored 61 diploid zygotes from 3PN zygotes donated by 35 couples, and 11 blastocysts developed with a blastocyst rate of 18.0%, which seems higher than that of nonrescued 3PN zygotes according to previous reports. After the whole zona pellucida free embryos were plated onto feeder cells to grow and passage, 2 hESC lines (CCRM-hESC-22 and CCRM-hESC-23) were generated and both carried normal karyotype (46, XY). The hESC lines were then characterized by morphology, expansion in vitro, and expression of specific markers of alkaline phosphatase, OCT4, SSEA4, TRA-1-60 and TRA-1-81. Furthermore, the pluripotency of these 2 hESC lines was confirmed by in vitro embryoid body formation and in vivo teratoma production. Our study indicates that depronucleared 3PN zygotes can improve the blastocysts formation rate, and normal hESC lines can be derived from those corrected 2PN embryos. Based on their multi-directional differentiation potential in vitro, the established hESC lines could be applied to the developmental risk assessment for IVF babies born from restored zygotes.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Zygote/metabolism , Cell Line , Cells, Cultured , Female , Fertilization in Vitro , Humans , Karyotype , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 30(3): 270-3, 2013 Jun.
Article in Zh | MEDLINE | ID: mdl-23744312

ABSTRACT

OBJECTIVE: To identify potential mutation of ectodysplasin A (EDA) gene in a Chinese family affected with X-linked hypohidrotic ectodermal dysplasia. METHODS: Blood samples were collected from the affected male proband, his family members and 103 unrelated individuals. Following extraction of genomic DNA, coding sequence of the EDA gene was amplified with PCR, and DNA sequencing was performed to detect potential mutation. RESULTS: A novel missense mutation, c.822G>T (p.W274C), was identified in exon 7 of the EDA gene in the proband, whilst his mother was found to be a heterozygous carrier. The same mutation was also found in 5 other family members including one affected male and four females, but was absent in unaffected males and 103 unrelated individuals. CONCLUSION: A c.822G>T mutation in exon 7 of the EDA gene probably underlies the disease in this Chinese family.


Subject(s)
Asian People/genetics , Ectodermal Dysplasia 1, Anhidrotic/diagnosis , Ectodermal Dysplasia 1, Anhidrotic/genetics , Ectodysplasins/genetics , Mutation , Base Sequence , China , Exons , Female , Humans , Male , Pedigree , Phenotype , Young Adult
14.
Stem Cell Res ; 68: 103049, 2023 04.
Article in English | MEDLINE | ID: mdl-36805323

ABSTRACT

GATA binding protein 6 (GATA6) is an important transcription factor of cardiovascular endothelial cells, has the potential to regulate the process of cardiac development. Consequently, its abnormal expression is related to congenital heart disease.Human GATA6 gene clones were on chromosome 18 q11.1- q 11.2, consists of 7 exons and 6 intron.Now, a human embryonic stem cell line with GATA6 c.620_647del (p.S208Afs*77) mutation was generated. Interestingly, the ESC line displayed a normal karyotype, pluripotency and morphology of stem cells.This cell line has the ability to undergo differentiation into three germ layers in vivo.


Subject(s)
Heart Defects, Congenital , Human Embryonic Stem Cells , Humans , Endothelial Cells , Cell Line , Embryonic Stem Cells/metabolism , Blastocyst , Cell Differentiation/genetics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism
15.
Front Endocrinol (Lausanne) ; 14: 859204, 2023.
Article in English | MEDLINE | ID: mdl-36950692

ABSTRACT

Objectives: This study aimed to investigate the potential mechanism of hyperoestrogensim elicited by ovulation induction affects endometrial receptivity and leads to embryo implantation abnormality or failure. Study design: Establishment of ovulation induction mouse model. Changes in mouse body weight, ovarian weight, serum E2 level and oestrous cycle were observed. During the peri-implantation period, morphological changes in the mouse uterus and implantation sites and the localization and protein levels of oestrogen receptors ERα and ERß, the tight junction factors CLDN3 and OCLN, the aquaporins AQP3, AQP4 and AQP8, and the sodium channel proteins SCNN1α, SCNN1ß and SCNN1γ were observed. The expression and cellular localization of ERα, CLDN3, AQP8 and SCNN1 ß in RL95-2 cell line were also detected by western blotting and immunofluorescence. Results: Ovarian and body weights were significantly higher in the 5 IU and 10 IU groups than in the CON group. The E2 level was significantly higher in the 10 IU group than in the CON group. The mice in the 10 IU group had a disordered oestrous cycle and were in oestrus for a long time. At 5.5 dpc, significantly fewer implantation sites were observed in the 10 IU group than in the CON (p<0.001) and 5 IU (p<0.05) groups. The probability of abnormal implantation and abortion was higher in the 10 IU group than in the CON and 5 IU groups. CLDN3, OCLN, AQP8 and SCNN1ß in the mouse endometrium were localized on the luminal epithelium and glandular epithelium and expression levels were lower in the 10 IU group than in the CON group. The protein expression level of ERα was increased by 50% in the 10 IU group compared to the CON group. The expressions of CLDN3, AQP8, SCNN1ß in RL95-2 cell line were significantly depressed by the superphysiological E2, ERα agonist or ERß agonist, which could be reversed by the oestrogen receptor antagonist. Conclusion: ART-induced hyperoestrogenism reduces CLDN3, AQP8 and SCNN1ß expression through ERα, thereby destroying tight junctions and water and sodium channels in the endometrial cavity epithelium, which may cause abnormal implantation due to abnormal uterine fluid secretion and absorption.


Subject(s)
Estrogen Receptor alpha , Estrogen Receptor beta , Pregnancy , Female , Mice , Animals , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Embryo Implantation , Uterus/metabolism , Receptors, Estrogen/metabolism , Reproductive Techniques, Assisted
16.
Front Pharmacol ; 14: 1149659, 2023.
Article in English | MEDLINE | ID: mdl-37388446

ABSTRACT

Acute lung injury (ALI) is one of the most common clinical emergencies with limited effective pharmaceutical treatment in the clinic, especially when it progresses to acute respiratory distress syndrome (ARDS). Currently, mesenchymal stem cells (MSCs) exhibit specific superiority for ALI/ARDS treatment. However, stem cells from different sources may result in controversial effects on similar disease conditions. This study aimed to determine the effects of human amnion-derived mesenchymal stem cells (hAMSCs) on two different ALI mice model. The administered hAMSCs effectively accumulated in the lung tissues in all hAMSC-treated groups. Compared with the model and 1% human serum albumin (HSA) groups, high-dose hAMSCs (1.0 × 106 cells) group significantly alleviated alveolar-capillary permeability, oxidative stress, inflammatory factors level and histopathological damage. In addition, the NF-κB signaling pathway is one of the key pathways activated during lipopolysaccharide (LPS) or paraquat (PQ)-induced lung injury. Our results indicated that hAMSCs (1.0 × 106 cells) obviously inhibited the expression of p-IKKα/ß, p-IκBα, and p-p65 in the lung tissue (p < 0.05). The high-dose (HD) hAMSC treatment exerted beneficial therapeutic effects on ALI mice models without detectable adverse reactions. The therapeutic effect of hAMSCs might involve NF-κB signaling pathway inhibition. hAMSC treatment is a potential candidate therapy for ALI.

17.
Front Cell Dev Biol ; 11: 1164757, 2023.
Article in English | MEDLINE | ID: mdl-37427383

ABSTRACT

The non-invasive and rapid assessment of the developmental potential of embryos is of great clinical importance in assisted reproductive technology (ART). In this retrospective study, we analyzed the metabolomics of 107 samples provided by volunteers and utilized Raman spectroscopy to detect the substance composition in the discarded culture medium of 53 embryos resulting in successful pregnancies and 54 embryos that did not result in pregnancy after implantation. The culture medium from D3 cleavage-stage embryos was collected after transplantation and a total of 535 (107 × 5) original Raman spectra were obtained. By combining several machine learning methods, we predicted the developmental potential of embryos, and the principal component analysis-convolutional neural network (PCA-CNN) model achieved an accuracy rate of 71.5%. Furthermore, the chemometric algorithm was used to analyze seven amino acid metabolites in the culture medium, and the data showed significant differences in tyrosine, tryptophan, and serine between the pregnancy and non-pregnancy groups. The results suggest that Raman spectroscopy, as a non-invasive and rapid molecular fingerprint detection technology, shows potential for clinical application in assisted reproduction.

18.
Stem Cell Res Ther ; 14(1): 188, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37507770

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are widely used in a variety of tissue regeneration and clinical trials due to their multiple differentiation potency. However, it remains challenging to maintain their replicative capability during in vitro passaging while preventing their premature cellular senescence. Forkhead Box P1 (FOXP1), a FOX family transcription factor, has been revealed to regulate MSC cell fate commitment and self-renewal capacity in our previous study. METHODS: Mass spectra analysis was performed to identify acetylation sites in FOXP1 protein. Single and double knockout mice of FOXP1 and HDAC7 were generated and analyzed with bone marrow MSCs properties. Gene engineering in human embryonic stem cell (hESC)-derived MSCs was obtained to evaluate the impact of FOXP1 key modification on MSC self-renewal potency. RESULTS: FOXP1 is deacetylated and potentiated by histone deacetylase 7 (HDAC7) in MSCs. FOXP1 and HDAC7 cooperatively sustain bone marrow MSC self-renewal potency while attenuating their cellular senescence. A mutation within human FOXP1 at acetylation site (T176G) homologous to murine FOXP1 T172G profoundly augmented MSC expansion capacity during early passages. CONCLUSION: These findings reveal a heretofore unanticipated mechanism by which deacetylation of FOXP1 potentiates self-renewal of MSC and protects them from cellular senescence. Acetylation of FOXP1 residue T172 as a critical modification underlying MSC proliferative capacity. We suggest that in vivo gene editing of FOXP1 may provide a novel avenue for manipulating MSC capability during large-scale expansion in clinical trials.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells , Animals , Humans , Mice , Cell Differentiation/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Histone Deacetylases/genetics , Mesenchymal Stem Cells/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
19.
Stem Cell Res ; 59: 102625, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34952435

ABSTRACT

The absence of azoospermia factor c (AZFc) is a common molecular cause of sperm failure. In men with non-obstructive azoospermia or severe oligospermia, the incidence of AZFc is around 10%. The AZFc region is located at the far end of the Yqll chromosome which has three non-overlapping sub-regions with a high frequency of deletion. Now, we generated a human embryonic stem cell line (SKLRMe001-A) that carries a deleted AZFc gene on the Y chromosome. The ESC line maintains a stem cell-like morphology, pluripotency, and has a normal karyotype. The cells can also differentiate into all three germ layers in vivo.

20.
J Mol Cell Biol ; 14(2)2022 06 17.
Article in English | MEDLINE | ID: mdl-35142858

ABSTRACT

Calciphylaxis is a rare disease characterized histologically by microvessel calcification and microthrombosis, with high mortality and no proven therapy. Here, we reported a severe uremic calciphylaxis patient with progressive skin ischemia, large areas of painful malodorous ulcers, and mummified legs. Because of the worsening symptoms and signs refractory to conventional therapies, treatment with human amnion-derived mesenchymal stem cells (hAMSCs) was approved. Preclinical release inspections of hAMSCs, efficacy, and safety assessment, including cytokine secretory ability, immunocompetence, tumorigenicity, and genetics analysis in vitro, were introduced. We further performed acute and long-term hAMSC toxicity evaluations in C57BL/6 mice and rats, abnormal immune response tests in C57BL/6 mice, and tumorigenicity tests in neonatal Balbc-nu nude mice. After the preclinical research, the patient was treated with hAMSCs by intravenous and local intramuscular injection and external supernatant application to the ulcers. When followed up to 15 months, the blood-based markers of bone and mineral metabolism improved, with skin soft tissue regeneration and a more favorable profile of peripheral blood mononuclear cells. Skin biopsy after 1-month treatment showed vascular regeneration with mature noncalcified vessels within the dermis, and 20 months later, the re-epithelialization restored the integrity of the damaged site. No infusion or local treatment-related adverse events occurred. Thus, this novel long-term intravenous combined with local treatment with hAMSCs warrants further investigation as a potential regenerative treatment for uremic calciphylaxis due to effects of inhibiting vascular calcification, stimulating angiogenesis and myogenesis, anti-inflammatory and immune modulation, multidifferentiation, re-epithelialization, and restoration of integrity.


Subject(s)
Calciphylaxis , Mesenchymal Stem Cells , Amnion , Animals , Calciphylaxis/complications , Calciphylaxis/therapy , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred C57BL , Mice, Nude , Rats , Ulcer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL