Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Biol Reprod ; 107(3): 790-799, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-35596251

RESUMO

Fanconi anemia complementation group B (FANCB) protein is a major component of the Fanconi anemia (FA) core complex and plays an important role in hematopoiesis and germ cell development. Deletion of Fancb gene causes the defect of primordial germ cell (PGC) development and infertility in male mice. However, it remains unknown whether Fancb is required for female germ cell development. In this study, we found that the fertility of Fancb knockout male mice in C57/ICR mixed backgrounds was not affected. Female Fancb-/- mice were obtained by crossing Fancb+/- females with Fancb-/Y males. The number of PGCs was dramatically decreased in Fancb-/- females. Very few oocytes were observed after birth and the primordial follicle pool was completely depleted at 6 weeks of age in Fancb-/- females. However, the remained oocytes from Fancb-/- mice were normal in fertilization and embryonic development from 2-cell to the blastocyst stage. We also found that Fancb and Fancl double-knockout males were also fertile and the number of sperm in epididymis was not reduced as compared to that of Fancb-/- and Fancl-/- single-knockout mice. Taken together, these results showed that Fancb is also essential for female germ cell development. Inactivation of Fancb causes massive germ cell loss and infertility in adult females. We also found that Fancb and Fancl do not act synergistically in regulating germ cell development.


Assuntos
Proteínas de Grupos de Complementação da Anemia de Fanconi , Infertilidade , Insuficiência Ovariana Primária , Animais , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Feminino , Células Germinativas/metabolismo , Infertilidade/genética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Gravidez , Insuficiência Ovariana Primária/genética , Sêmen
2.
Mol Biol Rep ; 49(6): 4469-4478, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35220512

RESUMO

BACKGROUND: Spermatogonial stem cells (SSCs) are unique stem cells that account for the whole reproductive life of males and transmit genetic information to offspring. SSC maintenance is intricate and the underlying mechanisms are largely unclear. Here, we report that SSC maintenance is driven by the plasminogen receptor (PLGRKT). METHODS AND RESULTS: PLGRKT was located in SSCs, and knockdown of PLGRKT expression in cultured neonatal testis and SSCs impaired the proliferation and promoted the apoptosis of cells. PLGRKT interacted with B lymphoma Mo-MLV insertion region 1 (BMI1), and modulated oxidative stress and p16/p19 signaling in SSCs. CONCLUSIONS: We demonstrated that reactive oxygen species (ROS) and p16/p19 signaling are involved in "PLGRKT-BMI1" co-regulation of SSC maintenance in mice.


Assuntos
Plasminogênio , Testículo , Animais , Células Cultivadas , Masculino , Camundongos , Estresse Oxidativo/genética , Plasminogênio/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/metabolismo , Testículo/metabolismo
3.
J Med Genet ; 58(1): 56-65, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32376790

RESUMO

BACKGROUND: Infertility affects approximately 15% of couples worldwide with male infertility being responsible for approximately 50% of cases. Although accumulating evidence demonstrates the critical role of the X chromosome in spermatogenesis during the last few decades, the expression patterns and potential impact of the X chromosome, together with X linked genes, on male infertility are less well understood. METHODS: We performed X chromosome exome sequencing followed by a two-stage independent population validation in 1333 non-obstructive azoospermia cases and 1141 healthy controls to identify variant classes with high likelihood of pathogenicity. To explore the functions of these candidate genes in spermatogenesis, we first knocked down these candidate genes individually in mouse spermatogonial stem cells (SSCs) using short interfering RNA oligonucleotides and then generated candidate genes knockout mice by CRISPR-Cas9 system. RESULTS: Four low-frequency variants were identified in four genes (BCORL1, MAP7D3, ARMCX4 and H2BFWT) associated with male infertility. Functional studies of the mouse SSCs revealed that knocking down Bcorl1 or Mtap7d3 could inhibit SSCs self-renewal and knocking down Armcx4 could repress SSCs differentiation in vitro. Using CRISPR-Cas9 system, Bcorl1 and Mtap7d3 knockout mice were generated. Excitingly, Bcorl1 knockout mice were infertile with impaired spermatogenesis. Moreover, Bcorl1 knockout mice exhibited impaired sperm motility and sperm cells displayed abnormal mitochondrial structure. CONCLUSION: Our data indicate that the X-linked genes are associated with male infertility and involved in regulating SSCs, which provides a new insight into the role of X-linked genes in spermatogenesis.


Assuntos
Cromossomos Humanos X/genética , Proteínas Repressoras/genética , Espermatogênese/genética , Testículo/crescimento & desenvolvimento , Animais , Sistemas CRISPR-Cas/genética , Exoma/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Motilidade dos Espermatozoides/genética , Espermatogônias/metabolismo , Espermatogônias/patologia , Testículo/patologia , Sequenciamento do Exoma
4.
Cryobiology ; 95: 1-8, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32001217

RESUMO

Melatonin is a ubiquitous indoleamine hormone synthesized primarily by the pineal gland. Diverse biological actions of melatonin involve quite complex mechanisms via its membrane receptors. More recently, studies have focused on the role of melatonin in male fertility preservation and male reproductive system. The protective effects of melatonin on immature testicular tissue freshness and activity maintenance and the preservation of sperm and spermatogonial stem cells (SSCs) have attracted considerable attention in recent years. Furthermore, since melatonin has strong antioxidant and anti-apoptotic properties, researchers have examined its potential role in male reproductive system. In this article, recent progress regarding melatonin's effects on male fertility preservation and its potential role is reviewed.


Assuntos
Preservação da Fertilidade , Melatonina , Antioxidantes/farmacologia , Criopreservação/métodos , Genitália , Masculino , Melatonina/farmacologia
5.
J Cell Biochem ; 120(1): 613-621, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30242874

RESUMO

Cryopreservation of testicular tissue before cancer therapy for fertility preservation in prepubertal boys with cancer is of great interest in reproductive medicine. Isolation of spermatogonial stem cells (SSCs) from cryopreserved tissues would be a suitable cell source to re-establish spermatogenesis after cancer therapy. We herein establish optimized protocols for cryopreservation of human testicular tissue and isolation of SSCs from cryopreserved tissue. We developed a freezing protocol that provided high testicular cell viability and supported structural integrity and tubular epithelium coherence similar to fresh tissue. Then, we established a protocol that allowed efficient isolation of functional SSCs from cryopreserved tissues. Isolated cells were found on the testicular basement membrane after xenotransplantation. Our results demonstrated the preservation of testicular tissue structure and high cell viability with efficient isolation of SSCs after testicular cryopreservation, which is promising for future therapeutic applications in fertility preservation.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Separação Celular/métodos , Criopreservação/métodos , Preservação da Fertilidade/métodos , Medicina Reprodutiva/métodos , Espermatogônias/citologia , Testículo/citologia , Animais , Apoptose , Sobrevivência Celular , Humanos , Masculino , Camundongos , Camundongos Nus , Espermatogênese , Transplante Heterólogo
6.
Gen Comp Endocrinol ; 273: 163-171, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29966660

RESUMO

Undifferentiated spermatogonia (Aund) or spermatogonial stem cells (SSCs) are committed to the establishment and maintenance of spermatogenesis and fertility throughout a male's life and are located in a highly specialized microenvironment called niche that regulates their fate. Although several studies have been developed on SSCs in mammalian testis, little is known about other vertebrate classes. The present study is the first to perform a more detailed investigation on the spermatogonial cells and their niche in a reptilian species. Thus, we characterized Aund/SSCs and evaluated the existence of SSCs niche in the Kinosternon scorpioides, a freshwater turtle found from Mexico to northern and central South America. Our results showed that, in this species, Aund/SSCs exhibited a nuclear morphological pattern similar to those described for other mammalian species already investigated. However, in comparison to other spermatogonial cell types, Aund/SSCs presented the largest nuclear volume in this turtle. Similar to some mammalian and fish species investigated, both GFRA1 and CSF1 receptors were expressed in Aund/SSCs in K. scorpioides. Also, as K. scorpioides Aund/SSCs were preferentially located near blood vessels, it can be suggested that this niche characteristic is a well conserved feature during evolution. Besides being valuable for comparative reproductive biology, our findings represent an important step towards the understanding of SSCs biology and the development of valuable systems/tools for SSCs culture and cryopreservation in turtles. Moreover, we expect that the above-mentioned results will be useful for reproductive biotechnologies as well as for governmental programs aiming at reptilian species conservation.


Assuntos
Escorpiões/citologia , Espermatogônias/citologia , Nicho de Células-Tronco , Tartarugas/metabolismo , Animais , Biomarcadores/metabolismo , Forma Celular , Tamanho Celular , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Escorpiões/metabolismo , América do Sul , Espermatogônias/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
J Cell Biochem ; 118(12): 4844-4853, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28543764

RESUMO

Stra8 (stimulated by retinoic acid gene 8) is a specific gene that is expressed in mammalian germ cells during transition from mitosis to meiosis and plays a key role in the initiation of meiosis in mammals and birds. So, the evaluation of the Stra8 pathway in cSSCs may provide a deeper insight into mammalian spermatogenesis. miRNA was also an important regulating factor for meiosis of SSCs. However, there is currently no data indicating that miRNA regulate the meiosis of SSCs via Stra8. Here, we predicted the prospective miRNA targeting to Stra8 using the online Bioinformatics database-Targetscan, and performed an analysis of the dual-luciferase recombinant vector, pGL3-CMV-LUC-MCS-Stra8-3'UTR. miR-31 mimics (miR-31m), miR-31 inhibitors (miR-31i), Control (NC, scrambled oligonucleotides transfection) were transfected into cSSCs; Stra8 and miRNA were analyzed by RT-qPCR, immunofluorescence, and Western blot. The detection of haploid was conducted by flow cytometry. The results showed that miR-31 regulates meiosis of cSSCs via targeting Stra8 in vitro and in vivo. Our study identifies a new regulatory pathway that miR-31 targets Stra8 and inhibits spermatogenesis. J. Cell. Biochem. 118: 4844-4853, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Proteínas Aviárias/metabolismo , Meiose/fisiologia , MicroRNAs/metabolismo , Espermatogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Células-Tronco Germinativas Adultas/citologia , Animais , Proteínas Aviárias/genética , Embrião de Galinha , Galinhas , Masculino , MicroRNAs/genética
8.
Mol Hum Reprod ; 22(9): 601-12, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27430551

RESUMO

STUDY QUESTION: Do the organ culture conditions, previously defined for in vitro murine male germ cell differentiation, also result in differentiation of rat spermatogonia into post-meiotic germ cells exhibiting specific markers for haploid germ cells? SUMMARY ANSWER: We demonstrated the differentiation of rat spermatogonia into post-meiotic cells in vitro, with emphasis on exhibiting, protein markers described for round spermatids. WHAT IS KNOWN ALREADY: Full spermatogenesis in vitro from immature germ cells using an organ culture technique in mice was first reported 5 years ago. However, no studies reporting the differentiation of rat spermatogonia into post-meiotic germ cells exhibiting the characteristic protein expression profile or into functional sperm have been reported. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Organ culture of testicular fragments of 5 days postpartum (dpp) neonatal rats was performed for up to 52 days. Evaluation of microscopic morphology, testosterone levels, mRNA and protein expression as measured by RT-qPCR and immunostaining were conducted to monitor germ cell differentiation in vitro. Potential effects of melatonin, Glutamax® medium, retinoic acid and the presence of epidydimal fat tissue on the spermatogenic process were evaluated. A minimum of three biological replicates were performed for all experiments presented in this study. One-way ANOVA, ANOVA on ranks and student's t-test were applied to perform the statistical analysis. MAIN RESULTS AND THE ROLE OF CHANCE: Male germ cells, present in testicular tissue pieces grown from 5 dpp rats, exhibited positive protein expression for Acrosin and Crem (cAMP (cyclic adenosine mono phosphate) response element modulator) after 52 days of culture in vitro. Intra-testicular testosterone production could be observed after 3 days of culture, while when epididymal fat tissue was added, spontaneous contractility of cultured seminiferous tubules could be observed after 21 days. However, no supportive effect of the supplementation with any factor or the co-culturing with epididymal fat tissue on germ cell differentiation in vitro or testosterone production was observed. LIMITATIONS, REASONS FOR CAUTION: The human testis is very different in physiology from the rat testis, further investigations are still needed to optimize the organ culture system for future use in humans. WIDER IMPLICATIONS OF THE FINDINGS: The successful differentiation of undifferentiated spermatogonia using the testis explant culture system might be employed in future to produce sperm from human spermatogonia as a clinical tool for fertility preservation in boys and men suffering infertility. LARGE SCALE DATA: None. STUDY FUNDING AND COMPETING INTERESTS: This work was supported financially by the Frimurare Barnhuset in Stockholm, the Paediatric Research Foundation, Jeanssons Foundation, Sällskåpet Barnåvard in Stockholm, Swedish Research Council/Academy of Finland, Emil and Wera Cornells Foundation, Samariten Foundation, the Swedish Childhood Cancer Foundation as well as through the regional agreement on medical training and clinical research (ALF) between Stockholm County Council and Karolinska Institutet. All authors declare no conflicts of interests.


Assuntos
Diferenciação Celular/fisiologia , Espermátides/citologia , Espermatogênese/fisiologia , Espermatogônias/citologia , Animais , Diferenciação Celular/genética , Preservação da Fertilidade , Células Germinativas , Masculino , Meiose/genética , Meiose/fisiologia , Ratos , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Espermátides/metabolismo , Espermatogênese/genética , Espermatogônias/metabolismo , Testículo/citologia , Testículo/metabolismo
9.
J Pak Med Assoc ; 66(3): 285-91, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26968278

RESUMO

OBJECTIVE: To investigate the effects of collagen and growth factors on in vitro proliferation of human spermatogonial stem cells obtained from patients with non-obstructive azoospermia. METHODS: The experimental cross-sectional study was conducted from February 2013 to April 2015 after obtaining approval from the ethics committee of Ahvaz Jundishapur University of Medical Sciences, Iran. Testicular sperm extractions of non-obstructive azoospermic patients were obtained from the Clinical Urology and Embryology, In Vitro Fertilization Department of Imam Khomeini Hospital. Spermatogonial stem cells and Sertoli cells, obtained from human testis biopsies by a two-step enzymatic digestion method, were purified using fluorescence- activated cell-sorting and daturastramonium-lectin, and were cultured separately. To investigate a more direct influential factor on colony formation, one control and two experimental groups were formed. Group 1 acted as the control in which spermatogonial stem cells were co-cultured with Sertoli cells alone. In group 2 they were co-cultured with Sertoli cells and growth factors such as leukaemia inhibitory factor, epidermal growth factor and glial cell-derived neurotrophic factor, and in group 3 with Sertoli cells along with growth factors in the presence of collagen-coated dishes. Number and diameter of the colonies were evaluated after 7 weeks. RESULTS: Specimens obtained related to 21 patients. Number and diameter of the colonies in group 3 (18±2.6 and 276.6±45.5) were significantly more than both groups 1 (3.5±1 and D1:81.6±12) and group 2(11±2.2 and 165.2±32.5) (p<0.05 each). Also, the number and diameter of colony in group 2 were significantly better than the control group (p<0.05).Expression profile of the VASA, promyelocytic leukaemia zinc-finger (PLZF), Octamer-binding transcription factor 4 (OCT4) and integrin a6 (INTGa6) were detected in all groups. Based on cytochemical findings, OCT4 was expressed in the colonies of all three groups. CONCLUSIONS: According to positive effects of collagen and growth factors on the colonisation of spermatogonial stem cells, it seems that using the cells may lead to better colonisation of this type of stem cells.


Assuntos
Células-Tronco Germinativas Adultas/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/farmacologia , Células-Tronco Germinativas Adultas/citologia , Azoospermia , Técnicas de Cultura de Células , Técnicas de Cocultura , Estudos Transversais , RNA Helicases DEAD-box/efeitos dos fármacos , RNA Helicases DEAD-box/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Citometria de Fluxo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Humanos , Integrina alfa6/efeitos dos fármacos , Integrina alfa6/metabolismo , Fatores de Transcrição Kruppel-Like/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/metabolismo , Fator Inibidor de Leucemia/farmacologia , Masculino , Fator 3 de Transcrição de Octâmero/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Células de Sertoli
10.
J Cell Biochem ; 115(2): 232-42, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24038201

RESUMO

miRNAs are expressed in many mammalian cells, acting specific roles in regulating gene expression or mediating special mRNAs cleavage by targeting their 3'-untranslated region (3'UTR). Some miRNAs are essential and important for animal development. However, it is still unclear what the relationship is between miR-34c and mammalian spermatogonial stem cells (SSCs). We found that a conserved microRNA-34c through its target-Nanos2, regulating SSCs' differentiation in mouse. Immunohistochemistry analysis of Nanos2 and miR-34c FISH results revealed the opposite expression trends between them. Seven bioinformatics websites and programs predicted that miR-34c has interaction sites in Nanos2's 3'UTR. Dual-luciferase reporter vector and mutated dual-luciferase reporter vector analysis validated that they are interacted. After transfection miR-34c mimics into mouse SSCs, or miR-34c lentiviral vector in vitro co-cultivation with seminiferous tubules, and Western blot analysis demonstrated that miR-34c over-expression could suppress Nanos2 expression in post-transcription level. Our experiments identified that miR-34c may promote meiosis process by interacting with Nanos2 leading up-regulation of Stra8 in mouse spermatogonial stem cells.


Assuntos
Diferenciação Celular/genética , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Meiose , Camundongos , MicroRNAs/genética , Proteínas de Ligação a RNA/genética
11.
J Pers Med ; 14(2)2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38392569

RESUMO

Infertility is a condition defined by the failure to establish a clinical pregnancy after 12 months of regular, unprotected sexual intercourse or due to an impairment of a person's capacity to reproduce either as an individual or with their partner. The authors have set out to succinctly investigate, explore, and assess infertility treatments, harnessing the potential of stem cells to effectively and safely treat infertility; in addition, this paper will present the legal and regulatory complexities at the heart of stem cell research, with an overview of the legislative state of affairs in six major European countries. For couples who cannot benefit from assisted reproductive technologies (ART) to treat their infertility, stem-cells-based approaches have been shown to be a highly promising approach. Nonetheless, lingering ethical and immunological uncertainties require more conclusive findings and data before such treatment avenues can become mainstream and be applied on a large scale. The isolation of human embryonic stem cells (ESCs) is ethically controversial, since their collection involves the destruction of human embryonic tissue. Overall, stem cell research has resulted in important new breakthroughs in the treatment of infertility. The effort to untangle the complex web of ethical and legal issues associated with such therapeutic approaches will have to rely on evidence-based, broadly shared standards, guidelines, and best practices to make sure that the procreative rights of patients can be effectively reconciled with the core values at the heart of medical ethics.

12.
Biol Reprod ; 89(6): 147, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24227752

RESUMO

A strong correlation exists between increasing paternal age and a decline in reproductive function. Testis aging is associated with testicular atrophy, increased DNA damage, and de novo mutations. It is unclear whether these problems arise from the spermatogonial stem cells (SSCs), a buildup of anomalies as older germ cells progress through spermatogenesis, or both. We hypothesize that with the continual divisions of SSCs that maintain the germ cell population, an alteration of these cells occurs over time. To test this, we utilized young (4-mo-old) and aged (18- and 21-mo-old) transgenic rats that express GFP in germ cells only. We first examined the number and activity of SSCs from the different age groups by transplantation. Aged rats had numerically fewer SSCs than young rats (<50%; not significant) despite the lack of testicular atrophy, and 21-mo-old rats show a significant reduction in colony length, suggesting that the quality of SSCs also deteriorates. To evaluate any molecular changes occurring in the early cells of spermatogenesis with age, we isolated an SSC-enriched population of CD9-positive (CD9(+)) cells using fluorescence-activated cell sorting (confirmed by transplantation studies) and extracted RNA for microarray analysis. In the aged CD9(+) cells, 60 transcripts were upregulated and more than 500 downregulated compared to the young cells. An altered expression was found for transcripts involved in mitosis and in DNA damage response. These results suggest molecular alterations in the SSC-enriched population of aged CD9(+) cells, implying that reproductive aging originates in the undifferentiated cells of spermatogenesis.


Assuntos
Células-Tronco Adultas/citologia , Envelhecimento/genética , Proliferação de Células , Expressão Gênica , Células-Tronco Adultas/metabolismo , Animais , Contagem de Células , Células Cultivadas , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Masculino , Ratos , Ratos Transgênicos , Espermatogônias/citologia , Espermatogônias/metabolismo , Tetraspanina 29/metabolismo
13.
Sex Dev ; 2023 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-37611547

RESUMO

INTRODUCTION: Spermatogonial stem cells (SSCs) offer remarkable competencies for animal reproduction and overcoming human disease as a result of their differentiation capability. We evaluated the effect of small molecule pifithrin-mu (PFT-µ) as a well-known inhibitor of P53 on SSC biological processes such as viability, apoptosis, and gene expression pattern. METHODS: The SSCs were isolated from the testes of adult NMRI mice and then cultured in DMEM / F12 medium containing 10% FBS. Then, they were characterized by the immunocytochemistry (ICC) technique by high PLZF and low c-Kit expressions. SSCs colony formation assay was carried out and their viability was estimated by MTT (Methylthiazolyldiphenyl-tetrazolium bromide, or 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide) assay upon exposure to PFT-µ (0, 0.6, 1.2, 2.5, and 5µM). The apoptosis percentages also were measured using FACS analysis, and finally, Oct4 and Stra8 expression at mRNA levels was assessed using real-time quantitative PCR. RESULTS: The 0.6 and 1.2µM PFT-µ improved the viability of SSC based on MTT assay results; however, 2.5 and 5µM PFT-µ reduced SSC viability compared with the control group. Moreover, PFT-µ at lower concentration enhanced the colony size of SSCs and diminished their apoptosis. As well, as exposure to PFT-µ up-regulated Oct4 expression, while down-regulating the meiotic entry marker, Stra8. CONCLUSION: Based on findings, optimized concentrations of PFT-µ can decrease SSCs apoptosis, and conversely potentiate their pluripotency and self-renewal capacities in vitro.

14.
Reprod Sci ; 30(6): 1687-1700, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36417177

RESUMO

Destruction of spermatogonial stem cells in juvenile men survivors of pediatric cancers leads to infertility as a side effect of gonadotoxic therapies. Sperm freezing before cancer treatment is commonly used in the clinic for fertility preservation, but this method is not applicable for prepubertal boys due to the lack of mature sperm. In these cases, cryopreservation of testicular tissues is the only option for fertility preservation. Although controlled slow freezing (CSF) is the most common procedure for testicular tissue cryopreservation, vitrification can be used as an alternative method. Controlled vitrification has prevented cell damage and formation of ice crystals. Procedures were done easily and quickly with a brief exposure time to high concentration of cryoprotectants without expensive equipment. Different studies used vitrification of testicular tissues and they assessed the morphology of seminiferous tubules, apoptosis, and viability of spermatogonial cells. Transplantation of vitrified testicular tissue into infertile recipient mice as well as in vitro culture of vitrified tissues was done in previous studies and their findings showed complete spermatogenesis and production of mature sperm. Review articles usually have compared controlled slow freezing with vitrification. In this review, we focused only on the vitrification method and its results. Despite promising results, many studies have been done for finding an optimal cryopreservation protocol in order to successfully preserve fertility in prepubertal boys.


Assuntos
Preservação da Fertilidade , Masculino , Animais , Camundongos , Preservação da Fertilidade/métodos , Vitrificação , Testículo , Criopreservação/métodos , Espermatozoides
15.
Life (Basel) ; 14(1)2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38276265

RESUMO

This review aims to explore advanced reproductive technologies for male fertility preservation, underscoring the essential role that animal models have played in shaping these techniques through historical contexts and into modern applications. Rising infertility concerns have become more prevalent in human populations recently. The surge in male fertility issues has prompted advanced reproductive technologies, with animal models playing a pivotal role in their evolution. Historically, animal models have aided our understanding in the field, from early reproductive basic research to developing techniques like artificial insemination, multiple ovulation, and in vitro fertilization. The contemporary landscape of male fertility preservation encompasses techniques such as sperm cryopreservation, testicular sperm extraction, and intracytoplasmic sperm injection, among others. The relevance of animal models will undoubtedly bridge the gap between traditional methods and revolutionary next-generation reproductive techniques, fortifying our collective efforts in enhancing male fertility preservation strategies. While we possess extensive knowledge about spermatogenesis and its regulation, largely thanks to insights from animal models that paved the way for human infertility treatments, a pressing need remains to further understand specific infertility issues unique to humans. The primary aim of this review is to provide a comprehensive analysis of how animal models have influenced the development and refinement of advanced reproductive technologies for male fertility preservation, and to assess their future potential in bridging the gap between current practices and cutting-edge fertility techniques, particularly in addressing unique human male factor infertility.

16.
Acta Inform Med ; 30(3): 205-212, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36311149

RESUMO

Background: Infertility is a serious health issue that affects people all around the world. One of the most common reasons for male infertility is sperm abnormalities. Researchers and scientists have been searching for a novel genetic marker to detect or recognize the genetic malfunction that causes sperm abnormalities. Micro-RNA (miRNAs) are small non-coded RNA molecules that present intra and extra-cellular and regulate gene expression. Objective: This studies began to search for a relation between miRNA expression levels and other diseases that may be related to them, considering that the main role of miRNAs was the down-regulation of genes. Methods: The main technique used in this study was to synthesize a complementary DNA (cDNA) (revers transcription method) of extracted total RNA by TRIzol then amplification of candidates' miRNAs genes by Reverse Transcriptase Quantitative Polymerase Chain Reaction RT-qPCR. Results: Studies found that miRNAs have a role in defining sperm qualities such as sperm count, motility, and shape. In this study, we chose the most miRNAs referred to in the previous study as a potential seminal fluid marker (miR-10a, miR-10b, miR-135a and miR-135b) to test them as potential infertility-related miRNAs markers (Asthenospermia AS, Oligospermia OS, Astheno-Oligospermia ASOS) in addition to health normal sperm NS. Conclusion: the main aim of this study was to find the miRNAs expression pattern to find a way to help scientists track the genetic causes of male infertility issues and a novel method to distinguish infertility genetically diseases. Conclusion: The findings may serve as a potential genetic marker for male infertility and provide a background for future research that targeted miRNAs as a molecular marker for medical and forensic fields, also as an infertility disease potential treatment.

17.
Stem Cell Rev Rep ; 18(5): 1718-1727, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34410593

RESUMO

Compared to embryonic and induced pluripotent stem cells, mesenchymal stem/stromal cells (MSCs) have made their presence felt with good therapeutic promise and safety profile. Transplanting MSCs has successfully helped to reverse infertility and resulted in live births in animal models and also in humans. But the underlying mechanism for their therapeutic potential is not yet clear. MSCs are not pluripotent and hence lack plasticity to differentiate into multiple adult cell types. They rather act as 'paracrine providers' to the tissue-resident stem cells since similar beneficial effects are also observed when their secretome (microvesicles or exosomes) is transplanted. Cytokines, growth factors, signaling lipids, mRNAs, and miRNAs secreted by MSCs enables tissue-resident stem cells to undergo differentiation into specific cell types. Tissue-resident stem cells include pluripotent, very small embryonic-like stem cells (VSELs) and progenitors [spermatogonial (SSCs), ovarian (OSCs) and endometrial (EnSCs) stem cells in testes, ovary and uterus respectively] which function in a subtle manner to maintain life-long tissue homeostasis and regenerate damaged (non-functional) reproductive tissues by differentiating into sperm, oocytes and endometrial epithelial cells respectively. Similar to restoring spermatogenesis, primordial follicles numbers are increased upon transplanting MSCs. Published literature suggests that MSCs do not differentiate into epithelial cells in the endometrium. Nuclear OCT-4 positive VSELs and cytoplasmic OCT-4, AXIN2 and KERATIN-19 positive epithelial progenitors have a greater role during endometrial regeneration. We propose, transplantation of MSCs simply provides growth factors/cytokines essential for the tissue-resident stem/progenitor cells to undergo differentiation into sperm, eggs and endometrial epithelial cells in the reproductive tissues.


Assuntos
Células-Tronco Embrionárias , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Citocinas , Feminino , Masculino , Ovário
18.
Reprod Sci ; 28(5): 1508-1522, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33481217

RESUMO

Spermatogonial stem cells (SSCs) are essential to the initiation of spermatogenesis. Cryopreservation, long-term maintenance, and auto-transplantation of SSCs could be a new treatment for infertility. The aim of this study was to add melatonin to the basic freezing medium and to evaluate its effect on the efficiency of the thawed SSCs after transplantation into the testicles of azoospermic mice. SSCs were isolated from newborn NMRI mice, and the cells were enriched to assess morphological features. The thawed SSCs were evaluated for survival, apoptosis, and ROS level before transplantation, and the proliferation (MVH and ID4) and differentiation (c-Kit, SCP3, TP1, TP2, and Prm1) markers of SSCs were examined using immunofluorescence, western blot, and quantitative real-time polymerase chain reaction (PCR) after transplantation. It was found that the survival rate of SSCs after thawing was significantly higher in the melatonin group compared with the cryopreservation group containing basic freezing medium, and the rate of apoptosis and level of ROS production also decreased significantly in the cryopreservation group with melatonin (p < 0.05). The expression of proliferation and differentiation markers after transplantation was significantly higher in the cryopreservation group with melatonin compared to the cryopreservation group (p < 0.05). The results suggest that adding melatonin to the basic freezing medium can effectively protect the SSCs by increasing the viability and reducing the ROS production and apoptosis and improve the transplantation efficiency of SSCs after cryopreservation, which will provide a significant suggestion for fertility protection in the clinic.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Células-Tronco Germinativas Adultas/transplante , Azoospermia/prevenção & controle , Criopreservação/métodos , Meiose , Melatonina/administração & dosagem , Torção do Cordão Espermático/complicações , Células-Tronco Germinativas Adultas/efeitos dos fármacos , Animais , Azoospermia/complicações , Células Cultivadas , Meios de Cultura/farmacologia , Modelos Animais de Doenças , Masculino , Meiose/efeitos dos fármacos , Camundongos
19.
Front Endocrinol (Lausanne) ; 12: 761249, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721307

RESUMO

To achieve spermatogenesis in vitro, one of the most challenging processes to mimic is meiosis. Meiotic problems, like incomplete synapsis of the homologous chromosomes, or impaired homologous recombination, can cause failure of crossover formation and subsequent chromosome nondisjunction, eventually leading to aneuploid sperm. These meiotic events are therefore strictly monitored by meiotic checkpoints that initiate apoptosis of aberrant spermatocytes and lead to spermatogenic arrest. However, we recently found that, in vitro derived meiotic cells proceeded to the first meiotic division (MI) stage, despite displaying incomplete chromosome synapsis, no discernible XY-body and lack of crossover formation. We therefore optimized our in vitro culture system of meiosis from male germline stem cells (mGSCs) in order to achieve full chromosome synapsis, XY-body formation and meiotic crossovers. In comparison to previous culture system, the in vitro-generated spermatocytes were transferred after meiotic initiation to a second culture dish. This dish already contained a freshly plated monolayer of proliferatively inactivated immortalized Sertoli cells supporting undifferentiated mGSCs. In this way we aimed to simulate the multiple layers of germ cell types that support spermatogenesis in vivo in the testis. We found that in this optimized culture system, although independent of the undifferentiated mGSCs, meiotic chromosome synapsis was complete and XY body appeared normal. However, meiotic recombination still occurred insufficiently and only few meiotic crossovers were formed, leading to MI-spermatocytes displaying univalent chromosomes (paired sister chromatids). Therefore, considering that meiotic checkpoints are not necessarily fully functional in vitro, meiotic crossover formation should be closely monitored when mimicking gametogenesis in vitro to prevent generation of aneuploid gametes.


Assuntos
Pareamento Cromossômico/fisiologia , Cromossomos/fisiologia , Meiose/fisiologia , Aneuploidia , Animais , Azoospermia/congênito , Azoospermia/fisiopatologia , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Células de Sertoli/fisiologia , Espermatócitos/fisiologia , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Testículo/fisiologia
20.
Theriogenology ; 162: 84-94, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33450717

RESUMO

The in vitro propagation and differentiation of spermatogonial stem cells (SSCs) has many potential applications within reproductive science and medicine. We established a two-dimensional (2D) cell culture system to proliferate and differentiate prepubertal mouse SSCs as a model capable of maximizing on a small number of donor SSCs. We also investigated the effects of retinol on in vitro SSC differentiation. Testis cells were cultured for 10 days in a serum-free medium. This produced SSC colonies which were then dissociated and sub-cultured for an additional 20 days in a differentiation medium. Before inducing differentiation, colonies expressed genes specific for undifferentiated spermatogonia (Ngn3, Plzf). After 10 days in the differentiation medium, Stra8 expression was upregulated. After 20 days, Acr expression was upregulated, indicating the completion of meiosis. Immunofluorescence, RT-PCR and flow cytometry confirmed the presence of haploid male germ cells (4.4% of all cells). When retinol was added to the differentiation medium the proportion of haploid germ cells increased (8.1% of cells). We concluded that, under serum-free culture conditions, prepubertal SSCs will generate colonies that can differentiate into haploid germ cells in a 2D culture system. These cells demonstrate a relatively high efficiency of haploid-cell production, which can be further improved with retinol.


Assuntos
Células-Tronco Germinativas Adultas , Espermatogônias , Animais , Técnicas de Cultura de Células/veterinária , Diferenciação Celular , Células Cultivadas , Haploidia , Masculino , Camundongos , Espermatogênese , Testículo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA