Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Curr Eye Res ; 47(12): 1631-1640, 2022 12.
Article in English | MEDLINE | ID: mdl-36218352

ABSTRACT

PURPOSE: Cataract is the leading cause of visual impairment and reversible blindness. Despite advances in surgical removal of cataracts, cataract continues to be a leading public-health issue due to the complications after surgery. Circular RNAs (circRNAs) have been showed to be implicated in the pathophysiology of age-related cataract (ARC). Herein, this work elucidated the role and mechanism of circMED12L in the process of ARC. METHODS: Human lens epithelial cells (HLECs) were exposed to hydrogen peroxide (H2O2) in experimental groups. Levels of genes and proteins were measured by qRT-PCR and western blotting. Cell growth was evaluated by Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. The oxidative stress was assessed by detecting the activity of malondialdehyde, catalase, and superoxide dismutase. The interaction between miR-34a-5p and circMED12L or ALCAM (activated leukocyte cell adhesion molecule) was validated using dual-luciferase reporter and RNA immunoprecipitation assays. RESULTS: CircMED12L expression was lower in the lens epithelium of ARC patients and H2O2-induced HLECs compared with the normal individuals and untreated cells. Functionally, forced expression of circMED12L could alleviate H2O2-induced viability inhibition, as well as apoptotic and oxidative injury in HLECs. Mechanistically, circMED12L/miR-34a-5p/ALCAM constituted a feedback loop in HLECs. MiR-34a-5p was increased, while ALCAM was decreased in ARC patients and H2O2-induced HLECs. High expression of miR-34a-5p reversed the protective effects of circMED12L on HLECs under H2O2 treatment. Besides, inhibition of miR-34a-5p could repress H2O2-induced apoptotic and oxidative injury in HLECs, which were abolished by subsequent ALCAM knockdown. CONCLUSION: Overexpression of circMED12L could protect against H2O2-induced apoptosis and oxidative stress in HLECs by miR-34a-5p/ALCAM axis.


Subject(s)
Cataract , MicroRNAs , Humans , Hydrogen Peroxide/toxicity , Hydrogen Peroxide/metabolism , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Activated-Leukocyte Cell Adhesion Molecule/pharmacology , Oxidative Stress , Epithelial Cells/metabolism , Cataract/genetics , Cataract/metabolism , Apoptosis , MicroRNAs/genetics , MicroRNAs/pharmacology , Fetal Proteins , Antigens, CD/metabolism , Cell Adhesion Molecules, Neuronal
2.
Exp Cell Res ; 398(1): 112382, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33253709

ABSTRACT

Radiotherapy is a conventional approach for anti-cancer treatment, killing tumor cells through damaging cellular DNA. While increasing studies have demonstrated that tumors generated the tolerance to radiation and tumor immune system was found to be correlated to radiotherapy resistance. Therefore, it is critical to identify potential immune factors associated with the efficacy of radiotherapy. Here in this study, we evaluated the sensitivities of different tumor cells to radiation and determined HEp-2 cells as the radio-resistant tumor cells for further investigation. IFNgamma as a key regulator of host immune response showed the potential to sensitize tumors to ionizing radiation (IR). Besides, IFNgamma-induced CXC chemokine ligand 10 (CXCL10) was found to be necessary for effective IR-induced killing of cultured HEp-2 cells. Increased clonogenic survival was observed in CXCL10-depleted HEp-2 cells and CXCL10-KO cells. Additionally, the loss of CXCL10 in HEp-2 cells showed less progression of the G0/G1 phase to G2/M when exposed to IR (8 Gy). Local IR (20 Gy) to nude mice bearing HEp-2 tumors significantly reduced tumor burden, while fewer effects on tumor burden in mice carrying CXCL10-KO tumors were observed. We furtherly evaluated the possible roles the chemokine receptor CXCR3 plays in mediating the sensitivity of cultured HEp-2 cells to IR. Altered expression of CXCR3 in HEp-2 cells affected IR-induced killing of HEp-2 cells. Our data suggest the IFNgamma-activated CXCL10/CXCR3 axis may contribute to the effective radiation-induced killing of HEp-2 cells in vitro.


Subject(s)
Chemokine CXCL10/metabolism , Interferon-gamma/metabolism , Radiation, Ionizing , Receptors, CXCR3/metabolism , Cell Line , Humans , Interferon-gamma/deficiency , Recombinant Proteins/metabolism
3.
Cell Death Differ ; 25(4): 749-766, 2018 03.
Article in English | MEDLINE | ID: mdl-29305586

ABSTRACT

Generation of functional spermatids from human spermatogonial stem cells (SSCs) in vitro is of utmost importance for uncovering mechanisms underlying human germ cell development and treating infertility. Here we report a three-dimensional-induced (3D-I) system by which human SSCs were efficiently differentiated into functional haploid spermatids. Human SSCs were isolated and identified phenotypically. Meiotic chromatin spreads and DNA content assays revealed that spermatocytes and haploid cells were effectively generated from human SSCs by 3D-I system. Haploid cells derived from human SSCs harbored normal chromosomes and excluded Y chromosome microdeletions. RNA sequencing and bisulfite sequencing analyses reflected similarities in global gene profiles and DNA methylation in human SSCs-derived spermatids and normal round spermatids. Significantly, haploid spermatids generated from human SSCs via 3D-I system were capable of fertilizing mouse oocytes, which subsequently enabled the development of hybrid embryos. This study thus provides invaluable human male gametes for treating male infertility.


Subject(s)
Cell Differentiation , Haploidy , Infertility, Male/metabolism , Sex Chromosome Disorders of Sex Development/metabolism , Spermatids/metabolism , Spermatogenesis , Stem Cells/metabolism , Adolescent , Adult , Animals , Cell Culture Techniques , Chromosome Deletion , Chromosomes, Human, Y/genetics , Chromosomes, Human, Y/metabolism , Female , Humans , Infertility, Male/genetics , Infertility, Male/pathology , Male , Mice , Middle Aged , Sex Chromosome Aberrations , Sex Chromosome Disorders of Sex Development/genetics , Sex Chromosome Disorders of Sex Development/pathology , Spermatids/pathology , Stem Cells/pathology
4.
Sci Rep ; 7: 45298, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387750

ABSTRACT

Sertoli cells are essential for regulating normal spermatogenesis. However, the mechanisms underlying human Sertoli cell development remain largely elusive. Here we examined the function and signaling pathways of BMP6 in regulating human Sertoli cells. RT-PCR, immunocytochemistry and Western blots revealed that BMP6 and its multiple receptors were expressed in human Sertoli cells. CCK-8 and EDU assays showed that BMP6 promoted the proliferation of Sertoli cells. Conversely, BMP6 siRNAs inhibited the division of these cells. Annexin V/PI assay indicated that BMP6 reduced the apoptosis in human Sertoli cells, whereas BMP6 knockdown assumed reverse effects. BMP6 enhanced the expression levels of ZO1, SCF, GDNF and AR in human Sertoli cells, and ELISA assay showed an increase of SCF by BMP6 and a reduction by BMP6 siRNAs. Notably, Smad2/3 phosphorylation and cyclin D1 were enhanced by BMP6 and decreased by BMP6 siRNAs in human Sertoli cells. The levels of DACH1 and TFAP2A were increased by BMP6 and reduced by BMP6 siRNAs, and the growth of human Sertoli cells was inhibited by these siRNAs. Collectively, these results suggest that BMP6 regulates the proliferation and apoptosis of human Sertoli cells via activating the Smad2/3/cyclin D1 and DACH1 and TFAP2A pathway.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Cyclin D1/metabolism , Eye Proteins/genetics , Sertoli Cells/cytology , Smad Proteins/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Apoptosis , Bone Morphogenetic Protein 6/genetics , Cell Proliferation , Cells, Cultured , Eye Proteins/metabolism , Humans , Male , Phosphorylation , Sertoli Cells/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism , Transcriptional Activation
6.
Oncotarget ; 8(9): 14576-14592, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28107194

ABSTRACT

Here we proposed a new concept that human spermatogonial stem cells (SSCs) can transdifferentiate into hepatocytes in vivo. We first established liver injury model of mice by carbon tetrachloride to provide proper environment for human SSC transplantation. Liver mesenchymal cells were isolated from mice and identified phenotypically. Human SSC line was recombined with liver mesenchymal cells, and they were transplanted under renal capsules of nude mice with liver injury. The grafts expressed hepatocyte hallmarks, including ALB, AAT, CK18, and CYP1A2, whereas germ cell and SSC markers VASA and GPR125 were undetected in these cells, implicating that human SSCs were converted to hepatocytes. Furthermore, Western blots revealed high levels of PCNA, AFP, and ALB, indicating that human SSCs-derived hepatocytes had strong proliferation potential and features of hepatocytes. In addition, ALB-, CK8-, and CYP1A2- positive cells were detected in liver tissues of recipient mice. Significantly, no obvious lesion or teratomas was observed in several important organs and tissues of recipient mice, reflecting that transplantation of human SSCs was safe and feasible. Collectively, we have for the first time demonstrated that human SSCs can be transdifferentiated to hepatocyte in vivo. This study provides a novel approach for curing liver diseases using human SSC transplantation.


Subject(s)
Cell Transdifferentiation , Hepatocytes/cytology , Spermatogonia/cytology , Stem Cell Transplantation/methods , Stem Cells/cytology , Albumins/metabolism , Animals , Blotting, Western , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/therapy , Cytochrome P-450 CYP1A2/metabolism , Hepatocytes/metabolism , Humans , Immunohistochemistry , Male , Mesenchymal Stem Cell Transplantation/methods , Mice, Nude , Microscopy, Fluorescence , Proliferating Cell Nuclear Antigen/metabolism , Stem Cells/metabolism , Transplantation, Heterologous , Transplantation, Homologous , alpha 1-Antitrypsin/metabolism
7.
Reproduction ; 153(2): 211-220, 2017 02.
Article in English | MEDLINE | ID: mdl-27864336

ABSTRACT

Generation of male germ cells from pluripotent cells could provide male gametes for treating male infertility and offer an ideal model for unveiling molecular mechanisms of spermatogenesis. However, the influence and exact molecular mechanisms, especially downstream effectors of BMP4 signaling pathways, in male germ cell differentiation of the induce pluripotent stem (iPS) cells, remain unknown. This study was designed to explore the role and mechanism of BMP4 signaling in the differentiation of mouse iPS cells to male germ cells. Embryoid body (EB) formation and recombinant BMP4 or Noggin were utilized to evaluate the effect of BMP4 on male germ cell generation from mouse iPS cells. Germ cell-specific genes and proteins as well as the downstream effectors of BMP4 signaling pathway were assessed using real-time PCR and Western blots. We found that BMP4 ligand and its multiple receptors, including BMPR1a, BMPR1b and BMPR2, were expressed in mouse iPS cells. Real-time PCR and Western blots revealed that BMP4 could upregulate the levels of genes and proteins for germ cell markers in iPS cells-derived EBs, whereas Noggin decreased their expression in these cells. Moreover, Smad1/5 phosphorylation, Gata4 transcription and the transcripts of Id1 and Id2 were enhanced by BMP4 but decreased when exposed to Noggin. Collectively, these results suggest that BMP4 promotes the generation of male germ cells from iPS cells via Smad1/5 pathway and the activation of Gata4, Id1 and Id2 This study thus offers novel insights into molecular mechanisms underlying male germ cell development.


Subject(s)
Bone Morphogenetic Protein 4/physiology , Cell Differentiation/physiology , Germ Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Blotting, Western , Bone Morphogenetic Protein 4/genetics , Cell Line , GATA4 Transcription Factor/physiology , Gene Expression , Induced Pluripotent Stem Cells/physiology , Inhibitor of Differentiation Protein 1/physiology , Inhibitor of Differentiation Protein 2/physiology , Male , Mice , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Smad1 Protein/physiology , Smad5 Protein/physiology , Spermatozoa/cytology
8.
Oncotarget ; 7(8): 8879-95, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26840458

ABSTRACT

To generate functional human hepatocytes from stem cells and/or extra-hepatic tissues could provide an important source of cells for treating liver diseases. Spermatogonial stem cells (SSCs) have an unlimited plasticity since they can dedifferentiate and transdifferentiate to other cell lineages. However, generation of mature and functional hepatocytes from human SSCs has not yet been achieved. Here we have for the first time reported direct transdifferentiation of human SSCs to mature and functional hepatocytes by three-step induction using the defined condition medium. Human SSCs were first transdifferentiated to hepatic stem cells, as evidenced by their morphology and biopotential nature of co-expressing hepatocyte and cholangiocyte markers but not hallmarks for embryonic stem cells. Hepatic stem cells were further induced to differentiate into mature hepatocytes identified by their morphological traits and strong expression of CK8, CK18, ALB, AAT, TF, TAT, and cytochrome enzymes rather than CK7 or CK19. Significantly, mature hepatocytes derived from human SSCs assumed functional attributes of human hepatocytes, because they could produce albumin, remove ammonia, and uptake and release indocyanine green. Moreover, expression of ß-CATENIN, HNF4A, FOXA1 and GATA4 was upregulated during the transdifferentiation of human SSCs to mature hepatocytes. Collectively, human SSCs could directly transdifferentiate to mature and functional hepatocytes. This study could offer an invaluable source of human hepatocytes for curing liver disorders and drug toxicology screening and provide novel insights into mechanisms underlying human liver regeneration.


Subject(s)
Azoospermia/pathology , Biomarkers/metabolism , Cell Lineage , Germ Cells/cytology , Hepatocytes/cytology , Spermatogonia/cytology , Testis/cytology , Azoospermia/metabolism , Cell Differentiation , Cell Transdifferentiation , Cells, Cultured , Culture Media, Conditioned , Flow Cytometry , Germ Cells/metabolism , Hepatocytes/metabolism , Humans , Immunoenzyme Techniques , Male , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spermatogonia/metabolism , Testis/metabolism , beta Catenin/metabolism
9.
Oncotarget ; 7(3): 2201-19, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26755652

ABSTRACT

Sertoli cells play critical roles in regulating spermatogenesis and they can be reprogrammed to the cells of other lineages, highlighting that they have significant applications in reproductive and regenerative medicine. The fate determinations of Sertoli cells are regulated precisely by epigenetic factors. However, the expression, roles, and targets of microRNA (miRNA) in human Sertoli cells remain unknown. Here we have for the first time revealed that 174 miRNAs were distinctly expressed in human Sertoli cells between Sertoli-cell-only syndrome (SCOS) patients and obstructive azoospermia (OA) patients with normal spermatogenesis using miRNA microarrays and real time PCR, suggesting that these miRNAs may be associated with the pathogenesis of SCOS. MiR-133b is upregulated in Sertoli cells of SCOS patients compared to OA patients. Proliferation assays with miRNA mimics and inhibitors showed that miR-133b enhanced the proliferation of human Sertoli cells. Moreover, we demonstrated that GLI3 was a direct target of miR-133b and the expression of Cyclin B1 and Cyclin D1 was enhanced by miR-133b mimics but decreased by its inhibitors. Gene silencing of GLI3 using RNA inference stimulated the growth of human Sertoli cells. Collectively, miR-133b promoted the proliferation of human Sertoli cells by targeting GLI3. This study thus sheds novel insights into epigenetic regulation of human Sertoli cells and the etiology of azoospermia and offers new targets for treating male infertility.


Subject(s)
Azoospermia/pathology , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Sertoli Cell-Only Syndrome/pathology , Sertoli Cells/cytology , Spermatogenesis/genetics , Azoospermia/genetics , Cell Proliferation/genetics , Cells, Cultured , Cyclin B1/biosynthesis , Cyclin D1/biosynthesis , Humans , Male , RNA Interference , RNA, Small Interfering/genetics , Sertoli Cell-Only Syndrome/genetics , Zinc Finger Protein Gli3
10.
Sci Rep ; 5: 16922, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26585066

ABSTRACT

Spermatogonial stem cells (SSCs) have significant applications in both reproductive and regenerative medicine. However, primary human SSCs are very rare, and a human SSC line has not yet been available. In this study, we have for the first time reported a stable human SSC line by stably expressing human SV40 large T antigen. RT-PCR, immunocytochemistry, and Western blots revealed that this cell line was positive for a number of human spermatogonial and SSC hallmarks, including VASA, DAZL, MAGEA4, GFRA1, RET, UCHL1, GPR125, PLZF and THY1, suggesting that these cells are human SSCs phenotypically. Proliferation analysis showed that the cell line could be expanded with significant increases of cells for 1.5 years, and high levels of PCNA, UCHL1 and SV40 were maintained for long-term culture. Transplantation assay indicated that human SSC line was able to colonize and proliferate in vivo in the recipient mice. Neither Y chromosome microdeletions of numerous genes nor tumor formation was observed in human SSC line although there was abnormal karyotype in this cell line. Collectively, we have established a human SSC line with unlimited proliferation potentials and no tumorgenesis, which could provide an abundant source of human SSCs for their mechanistic studies and translational medicine.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Cell Proliferation/genetics , Gene Expression , Adult , Adult Stem Cells/transplantation , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Blotting, Western , Cell Line, Transformed , Humans , Immunohistochemistry , Male , Mice , Microscopy, Fluorescence , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation/methods , Transplantation, Heterologous
11.
Reproduction ; 150(1): R25-34, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25852155

ABSTRACT

Spermatogenesis is composed of three distinctive phases, which include self-renewal of spermatogonia via mitosis, spermatocytes undergoing meiosis I/II and post-meiotic development of haploid spermatids via spermiogenesis. Spermatogenesis also involves condensation of chromatin in the spermatid head before transformation of spermatids to spermatozoa. Epigenetic regulation refers to changes of heritably cellular and physiological traits not caused by modifications in the DNA sequences of the chromatin such as mutations. Major advances have been made in the epigenetic regulation of spermatogenesis. In this review, we address the roles and mechanisms of epigenetic regulators, with a focus on the role of microRNAs and DNA methylation during mitosis, meiosis and spermiogenesis. We also highlight issues that deserve attention for further investigation on the epigenetic regulation of spermatogenesis. More importantly, a thorough understanding of the epigenetic regulation in spermatogenesis will provide insightful information into the etiology of some unexplained infertility, offering new approaches for the treatment of male infertility.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Meiosis/genetics , MicroRNAs/metabolism , Mitosis/genetics , Spermatogenesis/genetics , Animals , Humans , Male , MicroRNAs/genetics
12.
Cell Commun Signal ; 13: 20, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25880873

ABSTRACT

BACKGROUND: Sertoli cells play key roles in regulating spermatogenesis and testis development by providing structural and nutritional supports. Recent studies demonstrate that Sertoli cells can be converted into functional neural stem cells. Adult Sertoli cells have previously been considered the terminally differentiated cells with a fixed and unmodifiable population after puberty. However, this concept has been challenged. Since the number of adult human Sertoli cells is limited, it is essential to culture these cells for a long period and expand them to obtain sufficient cells for their basic research and clinic applications. Nevertheless, the studies on human Sertoli cells are restricted, because it is difficult to get access to human testis tissues. RESULTS: Here we isolated adult human Sertoli cells with a high purity and viability from obstructive azoospermia patients with normal spermatogenesis. Adult human Sertoli cells were cultured with DMEM/F12 and fetal bovine serum for 2 months, and they could be expanded with a 59,049-fold increase of cell numbers. Morphology, phenotypic characteristics, and the signaling pathways of adult human Sertoli cells from different passages were compared. Significantly, adult human Sertoli cells assumed similar morphological features, stable global gene expression profiles and numerous proteins, and activation of AKT and SMAD1/5 during long-period culture. CONCLUSIONS: This study demonstrates that adult human Sertoli cells can be cultured for a long period and expanded with remarkable increase of cell numbers whilst maintaining their primary morphology, phenotype and signaling pathways. This study could provide adequate human Sertoli cells for reproductive and regenerative medicine.


Subject(s)
Gene Expression Regulation , Proto-Oncogene Proteins c-akt/metabolism , Sertoli Cells/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Adult , Animals , Azoospermia/metabolism , Azoospermia/pathology , Cattle , Cell Culture Techniques , Cells, Cultured , Humans , Male , Sertoli Cells/pathology , Time Factors
13.
Semin Cell Dev Biol ; 29: 66-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24718316

ABSTRACT

Spermatogenesis is a complex process by which spermatogonial stem cells (SSCs) self-renew and differentiate into spermatozoa under the elaborate coordination of testicular microenvironment, namely, niche. Sertoli cells, which locate around male germ cells, are the most critical component of the niche. Significant progress has recently been made by peers and us on uncovering the effects of Sertoli cells on regulating fate determinations of SSCs. Here we addressed the roles and regulation of Sertoli cells in normal and abnormal spermatogenesis. Specifically, we summarized the biological characteristics of Sertoli cells, and we emphasized the roles of Sertoli cells in mediating the self-renewal, differentiation, apoptosis, de-differentiation, and trans-differentiation of SSCs. The association between abnormal function of Sertoli cells and impaired spermatogenesis was discussed. Finally, we highlighted several issues to be addressed for further investigation on the effects and mechanisms of Sertoli cells in spermatogenesis. Since Sertoli cells are the key supportive cells for SSCs and they are very receptive to modification, a better understanding of the roles and regulation of Sertoli cells in SSC biology and spermatogenesis would make it feasible to identify novel targets for gene therapy of male infertility as well as seek more efficient and safer strategies for male contraception.


Subject(s)
Adult Stem Cells/cytology , Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatogonia/cytology , Spermatozoa/cytology , Animals , Apoptosis , Azoospermia/pathology , Cell Differentiation/physiology , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Male , Mice , Phagocytosis
14.
Reproduction ; 147(6): R179-88, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24534952

ABSTRACT

Infertility is a major and largely incurable disease caused by disruption and loss of germ cells. It affects 10-15% of couples, and male factor accounts for half of the cases. To obtain human male germ cells 'especially functional spermatids' is essential for treating male infertility. Currently, much progress has been made on generating male germ cells, including spermatogonia, spermatocytes, and spermatids, from various types of stem cells. These germ cells can also be used in investigation of the pathology of male infertility. In this review, we focused on advances on obtaining male differentiated germ cells from different kinds of stem cells, with an emphasis on the embryonic stem (ES) cells, the induced pluripotent stem (iPS) cells, and spermatogonial stem cells (SSCs). We illustrated the generation of male differentiated germ cells from ES cells, iPS cells and SSCs, and we summarized the phenotype for these stem cells, spermatocytes and spermatids. Moreover, we address the differentiation potentials of ES cells, iPS cells and SSCs. We also highlight the advantages, disadvantages and concerns on derivation of the differentiated male germ cells from several types of stem cells. The ability of generating mature and functional male gametes from stem cells could enable us to understand the precise etiology of male infertility and offer an invaluable source of autologous male gametes for treating male infertility of azoospermia patients.


Subject(s)
Fertility , Infertility, Male/surgery , Spermatogenesis , Spermatozoa/transplantation , Stem Cell Transplantation , Animals , Biomarkers/metabolism , Cell Lineage , Cell Proliferation , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Phenotype , Spermatozoa/metabolism , Stem Cell Transplantation/methods
15.
Respirology ; 16(8): 1228-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21883677

ABSTRACT

BACKGROUND AND OBJECTIVE: The adenosine triphosphate (ATP)-binding cassette, sub-family B, member 1 (ABCB1) gene encodes P-glycoprotein (Pgp), which plays an important role in drug disposition by limiting intracellular uptake of paclitaxel. ABCB1 gene polymorphisms may alter the expression and function of Pgp, thereby influencing the response to chemotherapy. A panel of 17 non-small cell lung cancer (NSCLC) cell lines was used to investigate whether alterations in the ABCB1 gene or its mRNA expression correlated with in vitro chemosensitivity to paclitaxel. METHODS: Polymorphisms in the ABCB1 gene were evaluated by direct sequencing. mRNA expression levels were assessed by quantitative real-time reverse transcription PCR. In vitro chemosensitivity to paclitaxel was expressed as half-maximal inhibitory concentration values, using a tetrazolium (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)-based colorimetric assay. RESULTS: The variant allele frequencies for four ABCB1 gene polymorphisms were 14.71% for 2677G>T/A, 32.35% for 2734T>C, 23.53% for 3396C>T and 76.47% for 3435C>T. There was a significant positive correlation between ABCB1 mRNA expression and half-maximal inhibitory concentration values for paclitaxel (r=0.5322, P=0.0279). None of the four ABCB1 gene polymorphisms were associated with paclitaxel chemosensitivity or ABCB1 mRNA expression in the 17 cell lines. CONCLUSIONS: These in vitro results suggest that high ABCB1 mRNA expression may be a predictive biomarker for poor chemosensitivity to paclitaxel. The panel of NSCLC cell lines may provide clues and indications for establishing clinically useful relationships between a given polymorphism or level of gene expression and chemosensitivity to an anti-cancer agent.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Paclitaxel/pharmacology , Polymorphism, Genetic , ATP Binding Cassette Transporter 1 , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA, Neoplasm/genetics , Gene Frequency , Humans , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...