Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Sci China Life Sci ; 65(5): 969-987, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34586576

RESUMO

The surrogate reproduction technique, such as inter-specific spermatogonial stem cells (SSCs) transplantation (SSCT), provides a powerful tool for production of gametes derived from endangered species or those with desirable traits. However, generation of genome-edited gametes from a different species or production of gametes from a phylogenetically distant species such as from a different subfamily, by SSCT, has not succeeded. Here, using two small cyprinid fishes from different subfamilies, Chinese rare minnow (gobiocypris rarus, for brief: Gr) and zebrafish (danio rerio), we successfully obtained Gr-derived genome-edited sperm in zebrafish by an optimized SSCT procedure. The transplanted Gr SSCs supported the host gonadal development and underwent normal spermatogenesis, resulting in a reconstructed fertile testis containing Gr spermatids and zebrafish testicular somatic cells. Interestingly, the surrogate spermatozoa resembled those of host zebrafish but not donor Gr in morphology and swimming behavior. When pou5f3 and chd knockout Gr SSCs were transplanted, Gr-derived genome-edited sperm was successfully produced in zebrafish. This is the first report demonstrating surrogate production of gametes from a different subfamily by SSCT, and surrogate production of genome-edited gametes from another species as well. This method is feasible to be applied to future breeding of commercial fish and livestock.


Assuntos
Células-Tronco Germinativas Adultas , Peixe-Zebra , Células-Tronco Germinativas Adultas/transplante , Animais , Masculino , Espermatogênese/genética , Espermatogônias/transplante , Espermatozoides , Transplante de Células-Tronco/métodos , Testículo , Peixe-Zebra/genética
2.
Reprod Sci ; 29(2): 526-539, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34494233

RESUMO

Preserving the spermatogonial stem cells (SSCs) in long periods of time during the treatment of male infertility using stem cell banking systems and transplantation is an important issue. Therefore, this study was conducted to develop an optimal cryopreservation protocol for SSCs using 10 mM pentoxifylline (PTX) as an antioxidant in basal freezing medium. Testicular torsion-a mouse model for long-term infertility-was used to transplant fresh SSCs (n = 6), fresh SSCs treated with PTX (n = 6), cryopreserved SSCs with basal freezing medium (n = 6), and cryopreserved SSCs treated with PTX (n = 6). Eight weeks after germ cell transplantation, samples were assessed for proliferation, through evaluation of Ddx4 and Id4 markers, and differentiation via evaluation of C-Kit and Sycp3, Tnp1, Tnp2, and Prm1 markers. According to morphological and flow cytometry results, SSCs are able to form colonies and express Gfra1, Id4, α6-integrin, and ß1-integrin markers. We found positive influence from PTX on proliferative and differentiative markers in SSCs transplanted to azoospermic mice. In the recipient testis, donor SSCs formed spermatogenic colonies and sperm. Respecting these data, adding pentoxifylline is a practical way to precisely cryopreserve germ cells enriched for SSCs in cryopreservation, and this procedure could become an efficient method to restore fertility in a clinical setup. However, more studies are needed to ensure its safety in the long term.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Azoospermia/etiologia , Crioprotetores/uso terapêutico , Pentoxifilina/uso terapêutico , Torção do Cordão Espermático/complicações , Células-Tronco Germinativas Adultas/efeitos dos fármacos , Animais , Criopreservação , Modelos Animais de Doenças , Masculino , Camundongos
4.
Andrology ; 9(5): 1603-1616, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33960147

RESUMO

BACKGROUND: Cancer treatment of prepubertal patients impacts future fertility due to the abolition of spermatogonial stem cells (SSCs). In macaques, spermatogenesis could be regenerated by intratesticular transplantation of SSCs, but no studies have involved cytotoxic treatment before puberty and transplantation after puberty, which would be the most likely clinical scenario. OBJECTIVES: To evaluate donor-derived functional sperm production after SSC transplantation to adult monkeys that had received testicular irradiation during the prepubertal period. MATERIALS AND METHODS: We obtained prepubertal testis tissue by unilaterally castrating six prepubertal monkeys and 2 weeks later irradiated the remaining testes with 6.9 Gy. However, because spermatogenic recovery was observed, we irradiated them again 14 months later with 7 Gy. Three of the monkeys were treated with GnRH-antagonist (GnRH-ant) for 8 weeks. The cryopreserved testis cells from the castrated testes were then allogeneically transplanted into the intact testes of all monkeys. Tissues were harvested 10 months later for analyses. RESULTS: In three of the six monkeys, 61%, 38%, and 11% of the epididymal sperm DNA were of the donor genotype. The ability to recover donor-derived sperm production was not enhanced by the GnRH-ant pretreatment. However, the extent of filling seminiferous tubules during the transplantation procedure was correlated with the eventual production of donor spermatozoa. The donor epididymal spermatozoa from the recipient with 61% donor contribution were capable of fertilizing rhesus eggs and forming embryos. Although the transplantation was done into the rete testis, two GnRH-ant-treated monkeys, which did not produce donor-derived epididymal spermatozoa, displayed irregular tubular cords in the interstitium containing testicular spermatozoa derived from the transplanted donor cells. DISCUSSION AND CONCLUSION: The results further support that sperm production can be restored in non-human primates from tissues cryopreserved prior to prepubertal and post-pubertal gonadotoxic treatment by transplantation of these testicular cells after puberty into seminiferous tubules.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Puberdade/efeitos da radiação , Lesões Experimentais por Radiação/terapia , Espermatogênese/efeitos da radiação , Transplante de Células-Tronco , Animais , Criopreservação , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/administração & dosagem , Macaca mulatta , Masculino , Lesões Experimentais por Radiação/fisiopatologia , Túbulos Seminíferos , Espermatozoides/efeitos da radiação , Testículo/fisiopatologia , Testículo/efeitos da radiação
5.
Reprod Sci ; 28(6): 1733-1745, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33834375

RESUMO

Infertility is a major health problem, and despite improved treatments over the years, there are still some conditions that cannot be treated successfully using a conventional approach. Therefore, new options are being considered and one of them is cell therapy using stem cells. Stem cell treatments for infertility can be divided into two major groups, the first one being direct transplantation of stem cells or their paracrine factors into reproductive organs and the second one being in vitro differentiation into germ cells or gametes. In animal models, all of these approaches were able to improve the reproductive potential of tested animals, although in humans there is still too little evidence to suggest successful use. The reasons for lack of evidence are unavailability of proper material, the complexity of explored biological processes, and ethical considerations. Despite all of the above-mentioned hurdles, researchers were able to show that in women, it seems to be possible to improve some conditions, but in men, no similar clinically important improvement was achieved. To conclude, the data presented in this review suggest that the treatment of infertility with stem cells seems plausible, because some types of treatments have already been tested in humans, achieving live births, while others show great potential only in animal studies, for now.


Assuntos
Infertilidade/terapia , Células-Tronco/fisiologia , Células-Tronco Germinativas Adultas/transplante , Animais , Técnicas de Cultura de Células em Três Dimensões , Tubas Uterinas/fisiologia , Feminino , Humanos , Infertilidade Feminina/etiologia , Infertilidade Feminina/terapia , Infertilidade Masculina/terapia , Masculino , Células-Tronco Mesenquimais , Mitocôndrias/transplante , Oócitos/fisiologia , Oócitos/ultraestrutura , Oogênese , Gravidez , Insuficiência Ovariana Primária/complicações , Insuficiência Ovariana Primária/terapia , Regeneração , Espermatogênese , Transplante de Células-Tronco , Útero/fisiologia
6.
Stem Cell Reports ; 16(3): 597-609, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33636117

RESUMO

One cause of human male infertility is a scarcity of spermatogonial stem cells (SSCs) in testes with Sertoli cells that neither produce adequate amounts of GDNF nor form the Sertoli-Sertoli junctions that form the blood-testis barrier (BTB). These patients raise the issue of whether a pool of SSCs, depleted due to inadequate GDNF stimulation, will expand if normal signaling is restored. Here, we reduce adult mouse SSC numbers by 90% using a chemical-genetic approach that reversibly inhibits GDNF signaling. Signal resumption causes all remaining SSCs to replicate immediately, but they primarily form differentiating progenitor spermatogonia. Subsequently, self-renewing replication restores SSC numbers. Testicular GDNF levels are not increased during restoration. However, SSC replication decreases as numbers of SSCs and progenitors increase, suggesting important regulatory interactions among these cells. Finally, sequential loss of SSCs and then pachytene spermatocytes causes dissolution of the BTB, thereby recapitulating another important characteristic of some infertile men.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Autorrenovação Celular , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Infertilidade Masculina/metabolismo , Células de Sertoli/metabolismo , Transdução de Sinais , Células-Tronco Germinativas Adultas/transplante , Animais , Contagem de Células , Diferenciação Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Células-Tronco
7.
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
8.
Proc Natl Acad Sci U S A ; 117(39): 24195-24204, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32929012

RESUMO

Spermatogonial stem cell transplantation (SSCT) is an experimental technique for transfer of germline between donor and recipient males that could be used as a tool for biomedical research, preservation of endangered species, and dissemination of desirable genetics in food animal populations. To fully realize these potentials, recipient males must be devoid of endogenous germline but possess normal testicular architecture and somatic cell function capable of supporting allogeneic donor stem cell engraftment and regeneration of spermatogenesis. Here we show that male mice, pigs, goats, and cattle harboring knockout alleles of the NANOS2 gene generated by CRISPR-Cas9 editing have testes that are germline ablated but otherwise structurally normal. In adult pigs and goats, SSCT with allogeneic donor stem cells led to sustained donor-derived spermatogenesis. With prepubertal mice, allogeneic SSCT resulted in attainment of natural fertility. Collectively, these advancements represent a major step toward realizing the enormous potential of surrogate sires as a tool for dissemination and regeneration of germplasm in all mammalian species.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Proteínas de Ligação a RNA/fisiologia , Espermatogênese , Animais , Bovinos , Feminino , Cabras , Masculino , Camundongos , Camundongos Knockout , Suínos , Testículo/anatomia & histologia , Testículo/fisiologia , Transplante Homólogo
9.
Reprod Biol ; 20(4): 447-459, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32900639

RESUMO

Stem cell therapy and exosome therapy are the two experimental methods that are now at the center of attention. Various types of stem cells, especially mesenchymal stem cells and spermatogonial stem cells have been widely administrated in reproductive medicine. However, due to the limitation of injecting living cells, using their paracrine secretions such as exosomes seems to be a better option. Exosomes show regenerative, pro-angiogenic, anti-apoptotic, anti-inflammatory, anti-hypoxic, and anti-fibrotic characteristics. They can induce cell proliferation, cell viability, migration, oogenesis, spermatogenesis, capacitation, acrosome reaction, and embryonic implantation. Exosomes have shown promising results in regenerative medicine such as liver fibrosis, stroke, cardiac ischemia, and skin injuries. Exosomes have been used to treat reproductive diseases such as erectile dysfunction and primary ovarian insufficiency. However, the study of exosomes in reproductive medicine is limited. In this article, we are going to review some of the researches on the use of stem cells and exosomes in reproductive medicine and suggest administration of a combination of exosomes for alleviating the symptoms of endometriosis and asthenozoospermia based on previous studies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Exossomos/transplante , Medicina Reprodutiva/métodos , Transplante de Células-Tronco , Células-Tronco Germinativas Adultas/transplante , Animais , Desenvolvimento Embrionário , Disfunção Erétil/terapia , Feminino , Humanos , Infertilidade/terapia , Masculino , Transplante de Células-Tronco Mesenquimais , Oogênese , Insuficiência Ovariana Primária/terapia , Medicina Regenerativa/métodos , Espermatogênese , Células-Tronco/ultraestrutura
10.
Andrology ; 8(6): 1923-1934, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32691968

RESUMO

BACKGROUND: Spermatogonial stem cells (SSCs), as tissue-specific stem cells, are capable of both self-renewal and differentiation and supporting the continual and robust spermatogenesis for male fertility. As a rare sub-fraction of undifferentiated spermatogonia, SSCs share most molecular markers with the progenitor spermatogonia. Thus, the heterogeneity of the progenitor cells often obscures the characteristics of stem cells. Distinguishing SSCs from the progenitors is of paramount importance to understand the regulatory mechanisms governing their actions. OBJECTIVES: The present study was designed to reveal that SSEA4 can be a marker for putative porcine SSCs that distinguished it from the progenitors and to build a sorting program for efficient enrichment of porcine SSCs. METHODS: To explore expression of SSEA4 within the undifferentiated spermatogonial population, we performed co-immunofluorescent staining for SSEA4 and common molecular markers (VASA, DBA, PLZF, c-KIT, and SOX9) in the 7-, 90-, and 150-day-old porcine testicular tissues. SSEA4-positive cells were isolated from the 90-day-old porcine testes by flow cytometry. Immunofluorescent, RNA-sequencing, and transplantation analysis were used to reveal that SSEA4-positive fraction holds the stem cell capacity. RESULTS: We found that SSEA4 was expressed in a rare sub-fraction of porcine undifferentiated spermatogonia, and RNA-sequencing analysis revealed that the genes for stem cell maintenance and SSC-specific markers (ID4 and PAX7) were up-regulated in the SSEA4-sorted fraction, compared with undifferentiated spermatogonia. In addition, germ cell transplantation assay demonstrated that SSEA4-positive spermatogonia colonized in the recipient testicular tubules. Sorting of the undifferentiated spermatogonia with anti-SSEA4 antibody resulted in a 2.5-fold enrichment of SSCs compared with the germ cell gate group, and 21-fold enrichment of SSCs compared with the SSEA4-negative spermatogonia group. CONCLUSIONS: Our findings revealed that SSEA4 is a new surface marker for porcine undifferentiated spermatogonia. This finding helps to elucidate the characteristics of porcine SSCs and facilitates the culture and manipulation of SSCs.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/transplante , Espermatogênese/fisiologia , Espermatogônias/metabolismo , Antígenos Embrionários Estágio-Específicos/metabolismo , Células-Tronco Germinativas Adultas/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Xenoenxertos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espermatogônias/citologia , Espermatozoides/crescimento & desenvolvimento , Suínos , Testículo/metabolismo , Transplante Heterólogo
11.
Andrology ; 8(5): 1428-1441, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32351003

RESUMO

BACKGROUND: In male pre-pubertal cancer patients, radiation and chemotherapy impact future fertility by eradication of spermatogonial stem cells (SSCs). In macaques, spermatogenesis could be regenerated by intratesticular transplantation of SSCs, but only a small percentage of spermatozoa produced were of donor origin. Transient hormone suppression with a GnRH antagonist (GnRH-ant) enhanced spermatogenic recovery from transplanted SSCs. OBJECTIVES: To evaluate donor-derived and endogenous spermatogenic recovery after SSC transplantation into irradiated monkeys and to test whether hormone suppression around the time of transplantation facilitates spermatogenic recovery. MATERIALS AND METHODS: Testes of 15 adult rhesus monkeys were irradiated with 7 Gy and 4 months later transplanted, to one of the testes, with cryopreserved testicular cells containing SSCs from unrelated monkeys. Monkeys were either treated with GnRH-ant for 8 weeks before transplantation, GnRH-ant from 4 weeks before to 4 weeks after transplantation, or with no GnRH-ant. Tissues were harvested 10 months after transplantation. RESULTS: Two of the 15 monkeys, a control and a pre-transplantation GnRH-ant-treated, showed substantially higher levels of testicular spermatogenesis and epididymal sperm output in the transplanted side as compared to the untransplanted. Over 84% of epididymal spermatozoa on the transplanted side had the donor genotype and were capable of fertilizing eggs after intracytoplasmic sperm injection forming morulae of the donor paternal origin. Low levels of donor spermatozoa (~1%) were also identified in the epididymis of three additional monkeys. Transplantation also appeared to enhance endogenous spermatogenesis. DISCUSSION AND CONCLUSION: We confirmed that SSC transplantation can be used for restoration of fertility in male cancer survivors exposed to irradiation as a therapeutic agent. The success rate of this procedure, however, is low. The success of filling the tubules with the cell suspension, but not the GnRH-ant treatment, was related to the level of colonization by transplanted cells.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Espermatogênese/fisiologia , Espermatogônias/transplante , Transplante de Células-Tronco/métodos , Testículo/efeitos da radiação , Animais , Macaca mulatta , Masculino , Lesões Experimentais por Radiação
12.
Urol Clin North Am ; 47(2): 227-244, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32272995

RESUMO

Infertility caused by chemotherapy or radiation treatments negatively impacts patient-survivor quality of life. The only fertility preservation option available to prepubertal boys who are not making sperm is cryopreservation of testicular tissues that contain spermatogonial stem cells (SSCs) with potential to produce sperm and/or restore fertility. SSC transplantation to regenerate spermatogenesis in infertile adult survivors of childhood cancers is a mature technology. However, the number of SSCs obtained in a biopsy of a prepubertal testis may be small. Therefore, methods to expand SSC numbers in culture before transplantation are needed. Here we review progress with human SSC culture.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Preservação da Fertilidade/métodos , Infertilidade Masculina/prevenção & controle , Neoplasias/terapia , Espermatogênese/fisiologia , Células-Tronco Germinativas Adultas/fisiologia , Humanos , Infertilidade Masculina/etiologia , Masculino , Espermatogênese/efeitos dos fármacos , Espermatogênese/efeitos da radiação , Transplante de Células-Tronco/métodos
13.
Hum Reprod Update ; 26(3): 368-391, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32163572

RESUMO

BACKGROUND: Although the prognosis of childhood cancer survivors has increased dramatically during recent years, chemotherapy and radiation treatments for cancer and other conditions may lead to permanent infertility in prepubertal boys. Recent developments have shown that spermatogonial stem cell (SSC) transplantation may be a hope for restoring fertility in adult survivors of childhood cancers. For this reason, several centres around the world are collecting and cryopreserving testicular tissue or cells anticipating that, in the near future, some patients will return for SSC transplantation. This review summarizes the current knowledge and utility of SSC transplantation techniques. OBJECTIVE AND RATIONALE: The aim of this narrative review is to provide an overview of the currently used experimental injection techniques for SSC transplantation in animal and human testes. This is crucial in understanding and determining the role of the different techniques necessary for successful transplantation. SEARCH METHODS: A comprehensive review of peer-reviewed publications on this topic was performed using the PubMed and Google Scholar databases. The search was limited to English language work and studies between 1994 (from the first study on SSC transplantation) and April 2019. Key search terms included mouse, rat, boar, ram, dog, sheep, goat, cattle, monkey, human, cadaver, testes, SSC transplantation, injection and technique. OUTCOMES: This review provides an extensive clinical overview of the current research in the field of human SSC transplantation. Rete testis injection with ultrasonography guidance currently seems the most promising injection technique thus far; however, the ability to draw clear conclusions is limited due to long ischemia time of cadaver testis, the relatively decreased volume of the testis, the diminishing size of seminiferous tubules, a lack of intratesticular pressure and leakage into the interstitium during the injection on human cadaver testis. Current evidence does not support improved outcomes from multiple infusions through the rete testes. Overall, further optimization is required to increase the efficiency and safety of the infusion method. WIDER IMPLICATIONS: Identifying a favourable injection method for SSC transplantation will provide insight into the mechanisms of successful assisted human reproduction. Future research could focus on reducing leakage and establishing the optimal infusion cell concentrations and pressure.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Preservação da Fertilidade/métodos , Espermatogênese/fisiologia , Espermatogônias/transplante , Transplante de Células-Tronco/métodos , Animais , Bovinos , Criança , Criopreservação , Cães , Humanos , Masculino , Camundongos , Modelos Animais , Neoplasias/terapia , Ratos , Túbulos Seminíferos/fisiologia , Ovinos , Espermatogônias/citologia , Suínos
14.
Asian J Androl ; 22(2): 184-191, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31187778

RESUMO

An ideal animal model of azoospermia would be a powerful tool for the evaluation of spermatogonial stem cell (SSC) transplantation. Busulfan has been commonly used to develop such a model, but 30%-87% of mice die when administered an intraperitoneal injection of 40 mg kg-1. In the present study, hematoxylin and eosin staining, Western blot, immunofluorescence, and quantitative real-time polymerase chain reaction were used to test the effects of busulfan exposure in a mouse model that received two intraperitoneal injections of busulfan at a 3-h interval at different doses (20, 30, and 40 mg kg-1) on day 36 or a dose of 40 mg kg-1 at different time points (0, 9, 18, 27, 36, and 63 days). The survival rate of the mice was 100%. When the mice were treated with 40 mg kg-1 busulfan, dramatic SSC depletion occurred 18 days later and all of the germ cells were cleared by day 36. In addition, the gene expressions of glial cell line-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), chemokine (C-X-C Motif) ligand 12 (CXCL12), and colony-stimulating factor 1 (CSF1) were moderately increased by day 36. A 63-day, long-term observation showed the rare restoration of endogenous germ cells in the testes, suggesting that the potential period for SSC transplantation was between day 36 and day 63. Our results demonstrate that the administration of two intraperitoneal injections of busulfan (40 mg kg-1 in total) at a 3-h interval to mice provided a nonlethal and efficient method for recipient preparation in SSC transplantation and could improve treatments for infertility and the understanding of chemotherapy-induced gonadotoxicity.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Azoospermia/induzido quimicamente , Bussulfano/toxicidade , Infertilidade Masculina/induzido quimicamente , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Injeções Intraperitoneais , Masculino , Camundongos , Transplante de Células-Tronco/métodos
15.
Theriogenology ; 142: 441-449, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31711692

RESUMO

Genetic modification of spermatogonial stem cells (SSCs) is an alternative method to pronuclear microinjection and somatic cell nuclear transfer for transgenesis in large animals. In the present study, we optimized the process of homologous SSC transplantation in the water buffalo (Bubalus bubalis) using transfected enriched SSCs generated by a non-viral transfection approach. Firstly, the SSC enrichment efficiencies of extracellular matrix components viz. collagen, gelatin, and Datura stramonium agglutinin (DSA) lectin were determined either individually or in combination with Percoll density gradient centrifugation. The highest enrichment was achieved after differential plating with DSA lectin followed by Percoll density gradient centrifugation. Nucleofection showed greater transfection efficiency (68.55 ±â€¯4.56%, P < 0.05) for enriched SSCs in comparison to fugene HD (6.7 ±â€¯0.25%) and lipofectamine 3000 (15.57 ±â€¯0.74%). The transfected enriched SSCs were transplanted into buffalo males under the ultrasound guidance and testis was removed by castration after 7-8 weeks of transplantation. Persistence and localization of donor cells within recipient seminiferous tubules was confirmed using fluorescent microscopy. Further confirmation was done by flow cytometric evaluation of GFP expressing cells among those isolated from two-step enzymatic digestion of recipient testicular parenchyma. In conclusion, we demonstrated for the first time, generation of buffalo transfected enriched SSCs and their successful homologous transplantation in buffaloes. This study represents the first step towards genetic modifications in buffaloes using SSC transplantation technique.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Búfalos , Espermatogônias/transplante , Testículo/citologia , Transfecção , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Animais Geneticamente Modificados , Búfalos/genética , Técnicas de Cultura de Células , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Espermatogônias/citologia , Espermatogônias/metabolismo , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/veterinária , Testículo/metabolismo , Transfecção/métodos , Transfecção/veterinária , Transplante Homólogo/veterinária
16.
Asian Journal of Andrology ; (6): 184-191, 2020.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-1009750

RESUMO

An ideal animal model of azoospermia would be a powerful tool for the evaluation of spermatogonial stem cell (SSC) transplantation. Busulfan has been commonly used to develop such a model, but 30%-87% of mice die when administered an intraperitoneal injection of 40 mg kg-1. In the present study, hematoxylin and eosin staining, Western blot, immunofluorescence, and quantitative real-time polymerase chain reaction were used to test the effects of busulfan exposure in a mouse model that received two intraperitoneal injections of busulfan at a 3-h interval at different doses (20, 30, and 40 mg kg-1) on day 36 or a dose of 40 mg kg-1 at different time points (0, 9, 18, 27, 36, and 63 days). The survival rate of the mice was 100%. When the mice were treated with 40 mg kg-1 busulfan, dramatic SSC depletion occurred 18 days later and all of the germ cells were cleared by day 36. In addition, the gene expressions of glial cell line-derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), chemokine (C-X-C Motif) ligand 12 (CXCL12), and colony-stimulating factor 1 (CSF1) were moderately increased by day 36. A 63-day, long-term observation showed the rare restoration of endogenous germ cells in the testes, suggesting that the potential period for SSC transplantation was between day 36 and day 63. Our results demonstrate that the administration of two intraperitoneal injections of busulfan (40 mg kg-1 in total) at a 3-h interval to mice provided a nonlethal and efficient method for recipient preparation in SSC transplantation and could improve treatments for infertility and the understanding of chemotherapy-induced gonadotoxicity.


Assuntos
Animais , Masculino , Camundongos , Células-Tronco Germinativas Adultas/transplante , Azoospermia/induzido quimicamente , Bussulfano/toxicidade , Modelos Animais de Doenças , Infertilidade Masculina/induzido quimicamente , Injeções Intraperitoneais , Espermatogênese/efeitos dos fármacos , Espermatogônias/efeitos dos fármacos , Transplante de Células-Tronco/métodos
17.
Curr Mol Med ; 19(8): 539-546, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31288721

RESUMO

Worldwide, infertility affects 8-12% of couples of reproductive age and has become a common problem. There are many ways to treat infertility, including medication, intrauterine insemination, and in vitro fertilization. In recent years, stem-cell therapy has raised new hope in the field of reproductive disability management. Stem cells are self-renewing, self-replicating undifferentiated cells that are capable of producing specialized cells under appropriate conditions. They exist throughout a human's embryo, fetal, and adult stages and can proliferate into different cells. While many issues remain to be addressed concerning stem cells, stem cells have undeniably opened up new ways to treat infertility. In this review, we describe past, present, and future strategies for the use of stem cells in reproductive medicine.


Assuntos
Infertilidade Feminina/terapia , Infertilidade Masculina/terapia , Transplante de Células-Tronco , Células-Tronco Germinativas Adultas/transplante , Diferenciação Celular , Técnicas de Reprogramação Celular , Células-Tronco Embrionárias/transplante , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Masculino , Transplante de Células-Tronco Mesenquimais , Oócitos/citologia , Ovário/citologia , Técnicas de Reprodução Assistida/tendências , Espermatócitos/citologia , Células-Tronco/classificação , Células-Tronco/citologia
18.
Methods Mol Biol ; 2005: 205-220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31175655

RESUMO

Spermatogonial stem cell (SSC) culture and transplantation pave the way for clinical restoration of fertility in male prepubertal cancer survivors. In this chapter we detail the steps for isolating and freezing testicular tissue along with protocols for the subsequent recovery from cryopreservation and transplantation of cells into a recipient testis. Transplantation of cultured or thawed SSCs provides not only a functional assay for identification of stem cells, a critical tool for the study of the germline stem cell niche in model organisms, but also a framework for reconstitution of spermatogenesis in humans. As proof of concept, the outlined methods have been performed successfully in the murine model and have the potential to be translated to clinical environments.


Assuntos
Células-Tronco Germinativas Adultas/transplante , Separação Celular/métodos , Criopreservação/métodos , Modelos Biológicos , Espermatogênese , Nicho de Células-Tronco , Transplante de Células-Tronco/métodos , Animais , Humanos , Masculino , Camundongos , Camundongos Transgênicos
19.
Cell Physiol Biochem ; 53(1): 101-120, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31215778

RESUMO

In the recent decades, cardiovascular diseases emerged as the major leading cause of human mortality. However, current clinical approaches still do not encompass a thorough therapeutic solution for improving heart function of the patients who suffered an extensive myocardial injury. Based on this status quo, stem cells could become a novel option, as a natural source of the new myocardium lineage cells, being capable of paracrine factors secretion, protection or even regeneration of the damaged heart muscle. Efficient stem cell-based therapy of the heart should lead to repair or/and replacement of the degenerated tissue with functional myocardial and endothelial cells. Hereon, various types of pluripotent and multipotent stem cells have been already studied in the pre-clinical and clinical settings, demonstrating their cardiomyogenic and regenerative potential. In this context, as a type of male adult stem/ progenitors, spermatogonial stem cells feature a remarkable ability for a formation of cardiovascular lineages, based on our own observations. Presented data supports the presumption, that spermatogonial stem cells not only have a suitable capacity to generate functional heart cells but can also potentially improve the function of an injured myocardium. In this review article, we first describe the essential molecular and pathophysiological mechanisms involved in the heart tissue injury. Afterwards, based on our ongoing study, we review the impact of the stem cell technologies on the regeneration therapy in cardiovascular and myocardial diseases. Particular emphasis is being put on the usability of spermatogonial stem cells in cardiac therapy.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Traumatismos Cardíacos/terapia , Coração/fisiologia , Regeneração , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/transplante , Animais , Diferenciação Celular , Coração/fisiopatologia , Traumatismos Cardíacos/patologia , Traumatismos Cardíacos/fisiopatologia , Humanos , Miocárdio/citologia , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo
20.
In Vitro Cell Dev Biol Anim ; 55(3): 177-188, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30725355

RESUMO

Spermatogonial stem cells (SSCs) are a useful tool for the generation of genetically modified transgenic sperm. As a result, the transfer of specific genes into the cytoplasm of SSCs is crucial for the successful generation of transgenic sperm. Here, we report electroporation conditions optimized for SSCs derived from the porcine testis. The highest transfection efficiency and cell viability were observed in porcine SSCs transfected with 1 µg transgenic vector with a single electric pulse from an electroporator at a voltage of 100 V and a capacitor setting of 250 µF. The transfection efficiency and cell viability were constant regardless of the size of the transgenic vector. Furthermore, we did not detect loss of spermatozoa differentiation potential in the transfected porcine SSCs. From these results, we confirm that this electroporation-based gene delivery system can effectively introduce foreign DNA into the genome of porcine SSCs without any loss of the original porcine SSC characteristics, which will be important in the generation of mosaicism-free transgenic pigs produced from transgenic porcine sperm.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Eletroporação/métodos , Técnicas de Transferência de Genes , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/transplante , Animais , Animais Geneticamente Modificados , Blastocisto/fisiologia , Diferenciação Celular , Sobrevivência Celular , Fertilização in vitro , Vetores Genéticos , Masculino , Espermatozoides/citologia , Espermatozoides/fisiologia , Suínos , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...