Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
Cell Tissue Bank ; 25(1): 231-243, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37676366

RESUMEN

Spermatogonia stem cells (SSCs) are a unique cell population maintaining male spermatogenesis during life, through their potential for proliferation and differentiation. The application of silicon nanoparticles (SNs) and hyaluronic acid (HA) to induce the differentiation of SSCs seems promising. Herein, we investigate the effect of SN and HA scaffolds on the progression of SSCs spermatogenesis in mice. Initially SSCs were isolated from healthy immature mice and cultured on prepared scaffolds (HA, SN, and HA/SN) in a 3D culture system. Then viability of SSCs cultured on scaffolds was examined using MTT assay and Acridine Orange staining. Then SSCs cultured on scaffolds were transplanted into epididymal adipose tissue (EAT) in mature mice and the result was studied by H&E and IHC staining 8 weeks after transplantation. MTT and Acridine Orange analysis revealed that among three different scaffolds HA/SN based scaffold causes considerable toxicity on SSCs (P < 0.05) while H&E staining showed that culture of SSCs on HA, SN, and HA/SN scaffolds has a positive effect on the progression of SSCs spermatogenesis after transplantation into EAT. IHC staining identified TP1, TEKT1, and PLZF as crucial biomarkers in the spermatogenesis development of SSCs transplanted to EAT. According to the presence of these biomarkers in different experimental groups, we found the most spermatogenesis development in SSCs cultured on HA/SN scaffold (PLZF, P < 0.01) (TEKT1, P < 0.01) (TP1, P < 0.001). Our study showed that, although the cytotoxic effect of the HA/SN scaffold decreases the viability rate of SSCs; however, SSCs that survive on HA/SN scaffold showed more ability to progress in spermatogenesis after transplantation into EAT.


Asunto(s)
Ácido Hialurónico , Espermatogonias , Ratones , Animales , Masculino , Espermatogonias/trasplante , Silicio , Naranja de Acridina , Biomarcadores , Células Madre , Proliferación Celular , Testículo
2.
Clin Epigenetics ; 15(1): 58, 2023 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-37029425

RESUMEN

BACKGROUND: Spermatogonial stem cell transplantation (SSCT) is proposed as a fertility therapy for childhood cancer survivors. SSCT starts with cryopreserving a testicular biopsy prior to gonadotoxic treatments such as cancer treatments. When the childhood cancer survivor reaches adulthood and desires biological children, the biopsy is thawed and SSCs are propagated in vitro and subsequently auto-transplanted back into their testis. However, culturing stress during long-term propagation can result in epigenetic changes in the SSCs, such as DNA methylation alterations, and might be inherited by future generations born after SSCT. Therefore, SSCT requires a detailed preclinical epigenetic assessment of the derived offspring before this novel cell therapy is clinically implemented. With this aim, the DNA methylation status of sperm from SSCT-derived offspring, with in vitro propagated SSCs, was investigated in a multi-generational mouse model using reduced-representation bisulfite sequencing. RESULTS: Although there were some methylation differences, they represent less than 0.5% of the total CpGs and methylated regions, in all generations. Unsupervised clustering of all samples showed no distinct grouping based on their pattern of methylation differences. After selecting the few single genes that are significantly altered in multiple generations of SSCT offspring compared to control, we validated the results with quantitative Bisulfite Sanger sequencing and RT-qPCRin various organs. Differential methylation was confirmed only for Tal2, being hypomethylated in sperm of SSCT offspring and presenting higher gene expression in ovaries of SSCT F1 offspring compared to control F1. CONCLUSIONS: We found no major differences in DNA methylation between SSCT-derived offspring and control, both in F1 and F2 sperm. The reassuring outcomes from our study are a prerequisite for promising translation of SSCT to the human situation.


Asunto(s)
Metilación de ADN , Espermatogonias , Niño , Humanos , Masculino , Animales , Ratones , Adulto , Espermatogonias/metabolismo , Espermatogonias/trasplante , Semen/metabolismo , Espermatozoides/metabolismo , Células Madre/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
3.
Nihon Yakurigaku Zasshi ; 157(3): 168-171, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-35491111

RESUMEN

Infertility is one of the late side effects of cancer treatment. Expansion of anti-cancer treatment allow patients to have more life time, however infertility is becoming a matter damaging QOL during the young cancer survivors. The passive strategy such as avoiding the gonad-toxic drug or decreasing the total volume of them and shielding the gonads against cancer therapy has been conducted. To preserve the fertility of young female, ovary tissue cryopreservation is becoming a standard over the world after the success of offspring from cryopreserved ovary tissue autograft was reported. Sperm preservation method is established for the male fertility preservation method, however this is only applicable for sexually matured male patients. For the sake of preserving fertility of sexually immature male patients, many trials using cryopreserved testis tissues or testicular cells have been undergone. Recently, in vitro gametogenesis from stem cell of the human and the mouse to primordial germ cell like cell has been achieved. Here the previous challenges and the latest reports for obtaining functional sperm from immature testis and the reconstruction of spermatogonial niche as a potential approach for preserving fertility procedure are described.


Asunto(s)
Preservación de la Fertilidad , Infertilidad , Animales , Femenino , Preservación de la Fertilidad/métodos , Humanos , Masculino , Ratones , Calidad de Vida , Espermatogonias/trasplante , Testículo/trasplante
4.
Gene ; 823: 146390, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35248658

RESUMEN

Male fertility relies on continual and robust spermatogenesis. Environmental hypoxia adversely affects reproductive health in humans and animal studies provide compelling evidences that hypoxia impairs spermatogenesis in directly exposed individuals. However, a detail examination of hypoxia induced changes in testicular gene expression is still lacking and spermatogenesis in offspring of hypoxia exposed animals of awaits investigation. In this study, a hypobaric hypoxic chamber was used to simulate hypoxic conditions in mice and effects of hypoxia on spermatogenesis, fertility and testicular gene expression were evaluated. The results showed that hypoxia exposure reduced the number of undifferentiated spermatogonia but did not change the regenerative capacity of spermatogonial stem cells (SSCs) after transplantation. Hypoxia significantly increased the percent of abnormal sperm and these defects were recovered 2 months after returning to the normoxia. Transcriptome analysis of testicular tissues from control and hypoxia treated animals revealed that 766 genes were up-regulated and 965 genes were down-regulated. Surprisingly, expressions of genes that regulate epigenetic modifications were altered, indicating hypoxia-induced damage to spermatogenesis may be intergenerational. Indeed, animals that were sired by hypoxia exposed males exhibited impaired spermatogenesis. Together, these findings suggest that hypoxia exposure alters testicular gene expression and causes long-lasting damage to spermatogenesis.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Hipoxia/genética , Testículo/química , Animales , Epigénesis Genética , Regulación de la Expresión Génica , Masculino , Ratones , Espermatogénesis , Espermatogonias/citología , Espermatogonias/trasplante , Testículo/citología
5.
Stem Cell Reports ; 17(4): 924-935, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35334214

RESUMEN

Gametogenesis requires close interactions between germ cells and somatic cells. Derivation of sperm from spermatogonial stem cells (SSCs) is hampered by the inefficiency of spermatogonial transplantation technique in many animal species because it requires a large number of SSCs and depletion of endogenous spermatogenesis. Here we used mouse testis primordia and organoids to induce spermatogenesis from SSCs. We microinjected mouse SSCs into embryonic gonads or reaggregated neonatal testis organoids, which were transplanted under the tunica albuginea of mature testes. As few as 1 × 104 donor cells colonized both types of transplants and produced sperm. Moreover, rat embryonic gonads supported xenogeneic spermatogenesis from mouse SSCs when transplanted in testes of immunodeficient mice. Offspring with normal genomic imprinting patterns were born after microinsemination. These results demonstrate remarkable flexibility of the germ cell-somatic cell interaction and raise new strategies of SSC manipulation for animal transgenesis and analysis of male infertility.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Testículo , Animales , Masculino , Ratones , Organoides , Ratas , Espermatogénesis/genética , Espermatogonias/trasplante , Trasplante de Células Madre
6.
Sci China Life Sci ; 65(5): 969-987, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34586576

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas , Pez Cebra , Células Madre Germinales Adultas/trasplante , Animales , Masculino , Espermatogénesis/genética , Espermatogonias/trasplante , Espermatozoides , Trasplante de Células Madre/métodos , Testículo , Pez Cebra/genética
8.
Cell Rep ; 36(7): 109550, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407418

RESUMEN

Spermatogonial stem cells (SSCs) are maintained in a special microenvironment called a niche. However, much is unknown about components that constitute the niche. Here, we report that Cdc42 is essential for germline niche development. Sertoli cell-specific Cdc42-deficient mice showed normal premeiotic spermatogenesis. However, germ cells gradually disappeared during haploid cell formation and few germ cells remained in the mature testes. Spermatogonial transplantation experiments revealed a significant loss of SSCs in Cdc42-deficient testes. Moreover, Cdc42 deficiency in Sertoli cells downregulated GDNF, a critical factor for SSC maintenance. Cdc42-deficient Sertoli cells also exhibited lower nuclear MAPK1/3 staining. Inhibition of MAP2K1 or depletion of Pea15a scaffold protein downregulated GDNF expression. A screen of transcription factors revealed that Cdc42-deficient Sertoli cells downregulate DMRT1 and SOX9, both of which are critical for Sertoli cell development. These results indicate that Cdc42 is essential for niche function via MAPK1/3-dependent GDNF secretion.


Asunto(s)
Células Germinativas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Animales , Microambiente Celular , Regulación hacia Abajo , Desarrollo Embrionario , Eliminación de Gen , Regulación de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación , Factores de Transcripción SOX/metabolismo , Células de Sertoli/metabolismo , Espermatogonias/trasplante , Testículo/metabolismo , Factores de Transcripción/metabolismo
9.
Stem Cell Reports ; 16(7): 1832-1844, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34143973

RESUMEN

Spermatogonial transplantation has been used as a standard assay for spermatogonial stem cells (SSCs). After transplantation into the seminiferous tubules, SSCs transmigrate through the blood-testis barrier (BTB) between Sertoli cells and settle in a niche. Unlike in the repair of other self-renewing systems, SSC transplantation is generally performed after complete destruction of endogenous spermatogenesis. Here, we examined the impacts of recipient conditioning on SSC homing. Germ cell ablation downregulated the expression of glial cell line-derived neurotrophic factor, which has been shown to attract SSCs to niches, implying that nonablated niches would attract SSCs more efficiently. As expected, SSCs colonized nonablated testes when transplanted into recipients with the same genetic background. Moreover, although spermatogenesis was arrested at the spermatocyte stage in Cldn11-deficient mice without a BTB, transplantation not only enhanced donor colonization but also restored normal spermatogenesis. The results show promise for the development of a new transplantation strategy to overcome male infertility.


Asunto(s)
Espermatogonias/citología , Espermatogonias/trasplante , Trasplante de Células Madre , Testículo/citología , Animales , Apoptosis , Biomarcadores/metabolismo , Busulfano/farmacología , Claudinas/metabolismo , Citocinas/metabolismo , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Ratones Noqueados , Regeneración/efectos de los fármacos , Espermatogénesis
10.
Res Vet Sci ; 137: 127-137, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33965833

RESUMEN

Rhesus macaque (Macaca mulatta) is widely applied in animal model construction of infertility, spermatogonia stem cell transplantation and male reproductive diseases. In this review, we describe the seasonal changes of the reproductive system in rhesus macaques, the regular pattern of spermatogenesis and spermatozoa maturation, and the differentiation of spermatogonia and spermatocytes. The duration of the M. mulatta spermatogenesis is approximately 10 days and seminiferous epithelium cycles mainly consist of 12 stages, which provide a suitable model for reproductive studies in non-human primates. Here, we summarize the features of gonadal development and sperm maturation in the rhesus monkeys, which provide important information in the studies of reproductive biology. Rhesus macaque is an excellent animal model in spermatogonia stem cell transplantation. We discuss the applications and progresses of assisted reproductive technologies in sperm liquefaction, semen cryopreservation and spermatogonia stem cell transplantation of rhesus macaques. Besides, we sort out recent proteomic analyses of male reproductive systems and semen samples in rhesus macaques. This review mainly focuses on male reproductive biology and application studies using M. mulatta, which would promote the development of new therapeutic interventions on assisted reproduction and reproductive disease studies in the future.


Asunto(s)
Genitales Masculinos/fisiología , Macaca mulatta/fisiología , Espermatogénesis , Espermatogonias/trasplante , Trasplante de Células Madre/veterinaria , Animales , Criopreservación/veterinaria , Masculino , Proteómica , Estaciones del Año , Espermatozoides , Testículo/citología
11.
Fish Physiol Biochem ; 47(3): 767-776, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30937624

RESUMEN

We aimed to develop a simplified protocol for transplantation of Brycon orbignyanus spermatogonial stem cells (SSCs) into Astyanax altiparanae testes. Brycon orbignyanus testes were enzymatically digested and SSC purified by a discontinuous density gradient. Endogenous spermatogenesis was suppressed in A. altiparanae using busulfan or by incubation at 35 °C water, and SSCs from B. orbignyanus labeled with PKH26 were injected into their testes via the urogenital papilla. Twenty-two hours post-transplantation, labeled spermatogonia were observed in A. altiparanae tubular lumen. After 7 days, spermatogonia proliferated in the epithelium, and 21 days post-transplantation, sperm was observed in the lumen. Of surviving host fish, nearly 67% of those treated with busulfan and 85% of those held in warm water showed labeled cells in host germinal epithelium. The present study standardized, by a simple and accessible method, germ cell transplantation between sexually mature Characiformes fish species. This is the first report of xenogenic SSC transplantation in this fish order.


Asunto(s)
Characidae , Espermatogonias/citología , Espermatogonias/trasplante , Trasplante de Células Madre/métodos , Animales , Especies en Peligro de Extinción , Femenino , Masculino , Espermatogénesis , Testículo
12.
Andrology ; 8(5): 1428-1441, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32351003

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Espermatogénesis/fisiología , Espermatogonias/trasplante , Trasplante de Células Madre/métodos , Testículo/efectos de la radiación , Animales , Macaca mulatta , Masculino , Traumatismos Experimentales por Radiación
13.
Proc Natl Acad Sci U S A ; 117(14): 7837-7844, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32229564

RESUMEN

The blood-testis barrier (BTB) is thought to be indispensable for spermatogenesis because it creates a special environment for meiosis and protects haploid cells from the immune system. The BTB divides the seminiferous tubules into the adluminal and basal compartments. Spermatogonial stem cells (SSCs) have a unique ability to transmigrate from the adluminal compartment to the basal compartment through the BTB upon transplantation into the seminiferous tubule. Here, we analyzed the role of Cldn11, a major component of the BTB, in spermatogenesis using spermatogonial transplantation. Cldn11-deficient mice are infertile due to the cessation of spermatogenesis at the spermatocyte stage. Cldn11-deficient SSCs failed to colonize wild-type testes efficiently, and Cldn11-deficient SSCs that underwent double depletion of Cldn3 and Cldn5 showed minimal colonization, suggesting that claudins on SSCs are necessary for transmigration. However, Cldn11-deficient Sertoli cells increased SSC homing efficiency by >3-fold, suggesting that CLDN11 in Sertoli cells inhibits transmigration of SSCs through the BTB. In contrast to endogenous SSCs in intact Cldn11-deficient testes, those from WT or Cldn11-deficient testes regenerated sperm in Cldn11-deficient testes. The success of this autologous transplantation appears to depend on removal of endogenous germ cells for recipient preparation, which reprogrammed claudin expression patterns in Sertoli cells. Consistent with this idea, in vivo depletion of Cldn3/5 regenerated endogenous spermatogenesis in Cldn11-deficient mice. Thus, coordinated claudin expression in both SSCs and Sertoli cells expression is necessary for SSC homing and regeneration of spermatogenesis, and autologous stem cell transplantation can rescue congenital defects of a self-renewing tissue.


Asunto(s)
Fertilidad/genética , Infertilidad/terapia , Espermatogonias/trasplante , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Fertilidad/fisiología , Humanos , Infertilidad/genética , Infertilidad/patología , Masculino , Ratones , Espermatogénesis/genética , Espermatogonias/crecimiento & desarrollo , Espermatozoides/crecimiento & desarrollo , Espermatozoides/trasplante , Células Madre/citología , Trasplante Autólogo/métodos
14.
Hum Reprod Update ; 26(3): 368-391, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32163572

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Preservación de la Fertilidad/métodos , Espermatogénesis/fisiología , Espermatogonias/trasplante , Trasplante de Células Madre/métodos , Animales , Bovinos , Niño , Criopreservación , Perros , Humanos , Masculino , Ratones , Modelos Animales , Neoplasias/terapia , Ratas , Túbulos Seminíferos/fisiología , Ovinos , Espermatogonias/citología , Porcinos
15.
Gen Comp Endocrinol ; 289: 113341, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31954748

RESUMEN

Sprmatogonial stem cells (SSCs) are valuable for preservation of endangered fish species, biological experimentation, as well as biotechnological applications. However, the rarity of SSCs in the testes has been a great obstacle in their application. Thus, establishment of an efficient in-vitro culture system to support continuous proliferation of SSCs is essential. The present study aimed to establish an efficient and simple method for in vitro culture of Caspian trout undifferentiated spermatogonial cells. Using a two-step enzymatic digestion, testicular cells were isolated from immature testes composed of mainly undifferentiated spermatogonial cells with gonadosomatic indices of <0.05%. The spermatogonial cells were purified by differential plating through serial passaging. The purified cells indicated high expression of type A spermatogonia-related genes (Ly75, Gfrα1, Nanos2, Plzf and Vasa). Proliferation of purified cells was confirmed by BrdU incorporation. Co-culture of purified cells with testicular somatic cells as a feeder layer, resulted in continuous proliferation of type A spermatogonia. The cultured cells continued to express type A spermatogonia-specific markers after one month culture. The cultured spermatogonia were successfully incorporated into the germline after being intraperitoneally transplanted into sterile triploid rainbow trout hatchlings. These results, for the first time, demonstrated that the somatic microenvironment of the rainbow trout gonad can support the colonization and survival of intraperitoneally transplanted cells derived from a fish species belonging to a different genus. Therefore, the combination of in vitro culture system and xenotransplantation can be considered as a promising strategy for conservation of Caspian trout genetic resources.


Asunto(s)
Oncorhynchus mykiss/genética , Espermatogonias/trasplante , Animales , Células Cultivadas , Masculino
16.
Theriogenology ; 142: 441-449, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31711692

RESUMEN

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.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Búfalos , Espermatogonias/trasplante , Testículo/citología , Transfección , Células Madre Germinales Adultas/citología , Células Madre Germinales Adultas/metabolismo , Animales , Animales Modificados Genéticamente , Búfalos/genética , Técnicas de Cultivo de Célula , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Espermatogonias/citología , Espermatogonias/metabolismo , Trasplante de Células Madre/métodos , Trasplante de Células Madre/veterinaria , Testículo/metabolismo , Transfección/métodos , Transfección/veterinaria , Trasplante Homólogo/veterinaria
17.
Int J Mol Sci ; 20(23)2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31757040

RESUMEN

Fertility preservation for prepubertal boys relies exclusively on cryopreservation of immature testicular tissue (ITT) containing spermatogonia as the only cells with reproductive potential. Preclinical studies that used a nude mice model to evaluate the development of human transplanted ITT were characterized by important spermatogonial loss. We hypothesized that the encapsulation of testicular tissue in an alginate matrix supplemented with nanoparticles containing a necrosis inhibitor (NECINH-NPS) would improve tissue integrity and germ cells' survival in grafts. We performed orthotopic autotransplantation of 1 mm³ testicular tissue fragments recovered form mice (aged 4-5 weeks). Fragments were either non-encapsulated, encapsulated in an alginate matrix, or encapsulated in an alginate matrix containing NECINH-NPs. Grafts were recovered 5- and 21-days post-transplantation. We evaluated tissue integrity (hematoxylin-eosin staining), germ cells survival (immunohistochemistry for promyelocytic leukemia zinc-finger, VASA, and protein-boule-like), apoptosis (immunohistochemistry for active-caspase 3), and lipid peroxidation (immunohistochemistry for malondialdehyde). NECINH-NPs significantly improved testicular tissue integrity and germ cells' survival after 21 days. Oxidative stress was reduced after 5 days, regardless of nanoparticle incorporation. No effect on caspase-dependent apoptosis was observed. In conclusion, NECINH-NPs in an alginate matrix significantly improved tissue integrity and germ cells' survival in grafts with the perspective of higher reproductive outcomes.


Asunto(s)
Preservación de la Fertilidad/métodos , Nanopartículas/química , Espermatogonias/efectos de los fármacos , Inhibidores del Factor de Necrosis Tumoral/farmacología , Alginatos/química , Animales , Apoptosis , Supervivencia Celular , Peroxidación de Lípido , Masculino , Ratones , Espermatogonias/metabolismo , Espermatogonias/trasplante , Testículo/citología , Testículo/efectos de los fármacos , Testículo/trasplante , Inhibidores del Factor de Necrosis Tumoral/administración & dosificación
18.
Stem Cell Res Ther ; 10(1): 310, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31640769

RESUMEN

BACKGROUND: Spermatogonial stem cell transplantation (SSCT) is a promising therapy in restoring the fertility of childhood cancer survivors. However, the low efficiency of SSCT is a significant concern. SSCT could be improved by co-transplanting transforming growth factor beta 1 (TGFß1)-induced mesenchymal stem cells (MSCs). In this study, we investigated the reproductive efficiency and safety of co-transplanting spermatogonial stem cells (SSCs) and TGFß1-induced MSCs. METHODS: A mouse model for long-term infertility was used to transplant SSCs (SSCT, n = 10) and a combination of SSCs and TGFß1-treated MSCs (MSi-SSCT, n = 10). Both transplanted groups and a fertile control group (n = 7) were allowed to mate naturally to check the reproductive efficiency after transplantation. Furthermore, the testes from transplanted males and donor-derived male offspring were analyzed for the epigenetic markers DNA methyltransferase 3A (DNMT3A) and histone 4 lysine 5 acetylation (H4K5ac). RESULTS: The overall tubular fertility index (TFI) after SSCT (76 ± 12) was similar to that after MSi-SSCT (73 ± 14). However, the donor-derived TFI after MSi-SSCT (26 ± 14) was higher compared to the one after SSCT (9 ± 5; P = 0.002), even after injecting half of the number of SSCs in MSi-SSCT. The litter sizes after SSCT (3.7 ± 3.7) and MSi-SSCT (3.7 ± 3.6) were similar but differed significantly with the control group (7.6 ± 1.0; P < 0.001). The number of GFP+ offspring per litter obtained after SSCT (1.6 ± 0.5) and MSi-SSCT (2.0 ± 1.0) was also similar. The expression of DNMT3A and H4K5ac in germ cells of transplanted males was found to be significantly reduced compared to the control group. However, in donor-derived offspring, DNMT3A and H4K5ac followed the normal pattern. CONCLUSION: Co-transplanting SSCs and TGFß1-treated MSCs results in reproductive efficiency as good as SSCT, even after transplanting half the number of SSCs. Although transplanted males showed lower expression of DNMT3A and H4K5ac in donor-derived germ cells, the expression was restored to normal levels in germ cells of donor-derived offspring. This procedure could become an efficient method to restore fertility in a clinical setup, but more studies are needed to ensure safety in the long term.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Reproducción/fisiología , Espermatogonias/citología , Espermatogonias/trasplante , Acetilación , Animales , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Epigénesis Genética , Histonas/metabolismo , Lisina/metabolismo , Masculino , Ratones Endogámicos C57BL
19.
Stem Cell Res Ther ; 10(1): 135, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31109365

RESUMEN

BACKGROUND: Xenotransplantation of spermatogonial stem cells (SSCs) has become a popular topic in various research fields because manipulating these cells can provide insights into the mechanisms associated with germ cell lines and the entire spermatogenesis process; moreover, these cells can be used in several biotechnology applications. To achieve successful xenotransplantation, the in vitro microenvironment in which SSCs are cultured should be an ideal microenvironment for self-renewal and similar to the in vivo testicular microenvironment. The age of the donor, the correct spermatogenesis cycle, and the quality of the donor tissue are also important. Although cell culture-related factors, such as the in vitro supplementation of hormonal factors, are known to promote successful xenotransplantation in mice, little is known about the influence of these factors on SSCs in vitro or in vivo in other mammalian species, such as dogs (Canis lupus familiaris). In this context, the goals of this study were to test the effect of follicle-stimulating hormone (FSH) on canine spermatogonial stem cell (cSSC) cultures since this hormone is related to the glial cell-derived neurotrophic factor (GDNF) signaling pathway, which is responsible for the self-renewal and maintenance of these cells in vivo, and to investigate the microenvironment of the SSC culture after FSH supplementation. Additionally, in vivo analyses of transplanted FSH-supplemented cSSCs in the testes of infertile mice were performed to assess the capacity of cSSCs to develop, maintain, and restore spermatogenesis. METHODS: SSCs from canine prepubertal testes (aged 3 months) were cultured in vitro in the presence of FSH (10 IU L-1). GFRA1 transcript expression was detected to confirm the spermatogonia population in culture and the effect of FSH on these cells. The protein and transcript levels of late germ cell markers (GFRA1, DAZL, STRA8, PLZF, and CD49f) and a pluripotency marker (OCT4) were detected at 72 and 120 h to confirm the cSSC phenotype. In vivo experiments were performed by transplanting GFP+ cSSCs into infertile mice, and a 10-week follow-up was performed. Histological and immunofluorescence analyses were performed to confirm the repopulation capacity after cSSC xenotransplantation in the testis. RESULTS: Supplementation with FSH in cell culture increased the number of cSSCs positive for GFRA1. The cSSCs were also positive for the pluripotency and early germline marker OCT4 and the late germline markers PLZF, DAZL, C-kit, and GFRA-1. The in vivo experiments showed that the cSSCs xenotransplanted into infertile mouse testes were able to repopulate germline cells in the seminiferous tubules of mice. CONCLUSIONS: In conclusion, our results showed for the first time that the treatment of cSSC cultures with FSH can promote in vitro self-renewal, increase the population of germline cells, and possibly influence the success of spermatogenesis in infertile mice in vivo.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Espermatogénesis/genética , Espermatogonias/trasplante , Trasplante Heterólogo/métodos , Animales , Perros , Masculino , Ratones , Espermatogonias/citología
20.
Curr Opin Endocrinol Diabetes Obes ; 26(3): 166-174, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30998603

RESUMEN

PURPOSE OF REVIEW: This systematic review evaluates the state of the art in terms of strategies used to detect and remove contaminated malignant cells from testicular biopsy prior to spermatogonia stem cells (SSCs) autotransplantation to restore fertility. RECENT FINDINGS: Several trials have been done in past two decades to determine the reliable methods of detecting and purging cancer cells prior to SSCs autotransplantation. SUMMARY: The success in treating childhood cancer has dramatically increased over the past few decades. This leads to increasing demand for a method of fertility preservation for patients with pediatric cancer, as many cancer therapies can be gonadotoxic. Storing the SSCs prior to chemo- or radiation therapies and transplanting them back has been tested as a method of restoring fertility in rodents and nonhuman primate models. This has promise for restoring fertility in childhood cancer survivors. One of the major concerns is the possibility of malignant cell presence in testicular tissue biopsies that could re-introduce cancer to the patient after SSCs autotransplantation. Non-solid cancers - especially hematologic malignancies - have the risk of being transplanted back into patients after SSCs cryopreservation even if they were only present in small number in the stored testicular tissue biopsy.


Asunto(s)
Separación Celular/métodos , Preservación de la Fertilidad , Neoplasias/patología , Espermatogonias/patología , Espermatogonias/trasplante , Trasplante de Células Madre/métodos , Testículo/patología , Separación Celular/tendencias , Niño , Criopreservación/métodos , Criopreservación/tendencias , Preservación de la Fertilidad/métodos , Preservación de la Fertilidad/tendencias , Neoplasias Hematológicas/patología , Humanos , Infiltración Leucémica/patología , Masculino , Preservación de Semen/métodos , Preservación de Semen/normas , Preservación de Semen/tendencias , Trasplante de Células Madre/tendencias , Trasplante Autólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA