Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38890266

RESUMEN

PURPOSE: To identify the sperm preparation procedure that selects the best sperm population for medically assisted reproduction. METHODS: Prospective observational study comparing the effect of four different sperm selection procedures on various semen parameters. Unused raw semen after routine diagnostic analysis was split in four fractions and processed by four different methods: (1) density gradient centrifugation (DGC), (2) sperm wash (SW), (3) DGC followed by magnetic activated cell sorting (MACS), and (4) using a sperm separation device (SSD). Each fraction was analyzed for progressive motility, morphology, acrosome index (AI), and DNA fragmentation index (DFI). RESULTS: With DGC as standard of care in intraclass correlation coefficient analysis, only SSD was in strong disagreement regarding progressive motility and DFI [0.26, 95%CI (- 0.2, 0.58), and 0.17, 95%CI (- 0.19, 0.45), respectively]. When controlling for abstinence duration, DFI was significantly lower after both MACS and SSD compared to DGC [- 0.27%, 95%CI (- 0.47, - 0.06), p = 0.01, and - 0.6%, 95%CI (- 0.80, - 0.41), p < 0.001, respectively]. Further comparisons between SSD and MACS indicate significantly less apoptotic cells [Median (IQR) 4 (5), 95%CI (4.1, - 6.8) vs Median (IQR) 5 (8), 95%CI (4.9, - 9.2), p < 0.001, respectively] and dead cells [Median (IQR) 9.5 (23.3), 95%CI (13.2, - 22.4) vs Median (IQR) 22 (28), 95%CI (23.1, - 36.8), p < 0.001, respectively] in the SSD group. CONCLUSION: The selection of a population of highly motile spermatozoa with less damaged DNA from unprocessed semen is ideally performed with SSD. Question remains whether this method improves the embryological outcomes in the IVF laboratory.

2.
Adv Exp Med Biol ; 1345: 161-164, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34582022

RESUMEN

Decellularized testicular matrix (DTM) enables researchers to focus on the specific composition of the testicular extracellular matrix (ECM) and elucidate its role in spermatogenesis. Furthermore, it provides the natural architectural arrangement that could guide the reorganization of dissociated testicular cells in vitro. This is a key consideration as the presence of an authentic nutritive and endocrine support has been proven to be essential for in vitro spermatogenesis, at least in the mouse (Oliver and Stukenborg in Andrology 8:825-834, 2020; Richer et al. in Andrology 12741, 2019). Hence, scaffolds of DTM could be harnessed for the development of a human in vitro spermatogenesis culture system, which is a missing link in male fertility preservation and could be a possible treatment for nonobstructive azoospermia (Gassei and Orwig in Steril 105:256-266, 2016).


Asunto(s)
Matriz Extracelular , Preservación de la Fertilidad , Animales , Masculino , Ratones , Espermatogénesis , Testículo , Ingeniería de Tejidos , Andamios del Tejido
3.
Reprod Biomed Online ; 40(2): 261-269, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32001160

RESUMEN

RESEARCH QUESTION: Which cryopreservation method better protects reproductive potential: the cryopreservation of a testicular cell suspension (TCS) or the cryopreservation of testicular tissue (TET)? DESIGN: Two cryopreservation strategies for spermatogonial stem cells (SSCs) were compared in a mouse model: cryopreservation as TET or as TCS. Evaluated outcomes were number of viable cells after thawing, number and length of donor-derived colonies after spermatogonial stem cell transplantation (SSCT), number of litters, litter size and number of donor-derived pups after mating. RESULTS: Compared with cryopreserving TCS, cryopreservation of TET resulted in significantly higher numbers of viable cells after thawing (TET: 13.4 â€¯×  104 ± 7.2 â€¯×  104 versus TCS: 8.2 â€¯×  104 ± 2.7 â€¯×  104; P = 0.0002), more (TET: 47.6 ± 19.2 versus TCS: 18.5 ± 13.0; P = 0.0039) and longer (TET: 5.2 ± 1.0 mm versus TCS: 2.7 ± 1.5 mm; P = 0.0016) donor-derived colonies, and more donor-derived pups per litter (TET: 2.2 ± 0.2 versus TCS: 0.5 ± 0.1; P = 0.0008). CONCLUSIONS: Cryopreservation of TET is the preferred method to cryopreserve SSCs prior to SSCT in a mouse model.


Asunto(s)
Células Madre Germinales Adultas , Preservación de la Fertilidad/métodos , Fertilidad/fisiología , Testículo/trasplante , Animales , Criopreservación , Masculino , Ratones
4.
Hum Mol Genet ; 24(19): 5581-8, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26199321

RESUMEN

Infertility is a global healthcare problem, and despite long years of assisted reproductive activities, a significant number of cases remain idiopathic. Our currently restricted understanding of basic mechanisms driving human gametogenesis severely limits the improvement of clinical care for infertile patients. Using exome sequencing, we identified a nonsense mutation leading to a premature stop in the TEX15 locus (c.2130T>G, p.Y710*) in a consanguineous Turkish family comprising eight siblings in which three brothers were identified as infertile. TEX15 displays testis-specific expression, maps to chromosome 8, contains four exons and encodes a 2789-amino acid protein with uncertain function. The mutation, which should lead to early translational termination at the first exon of TEX15, co-segregated with the infertility phenotype, and our data strongly suggest that it is the cause of spermatogenic defects in the family. All three affected brothers presented a phenotype reminiscent of the one observed in KO mice. Indeed, previously reported results demonstrated that disruption of the orthologous gene in mice caused a drastic reduction in testis size and meiotic arrest in the first wave of spermatogenesis in males while female KO mice were fertile. The data from our study of one Turkish family suggested that the identified mutation correlates with a decrease in sperm count over time. A diagnostic test identifying the mutation in man could provide an indication of spermatogenic failure and prompt patients to undertake sperm cryopreservation at an early age.


Asunto(s)
Proteínas de Ciclo Celular/genética , Codón sin Sentido , Infertilidad Masculina/genética , Análisis de Secuencia de ADN/métodos , Espermatogénesis , Población Blanca/genética , Consanguinidad , Exoma , Predisposición Genética a la Enfermedad , Humanos , Infertilidad Masculina/patología , Masculino , Meiosis , Oligospermia , Tamaño de los Órganos , Linaje , Testículo/anatomía & histología , Factores de Tiempo , Turquía
5.
J Assist Reprod Genet ; 34(5): 683-694, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28401488

RESUMEN

PURPOSE: The purpose of this study was to identify mutations that cause non-syndromic male infertility using whole exome sequencing of family cases. METHODS: We recruited a consanguineous Turkish family comprising nine siblings with male triplets; two of the triplets were infertile as well as one younger infertile brother. Whole exome sequencing (WES) performed on two azoospermic brothers identified a mutation in the melanoma antigen family B4 (MAGEB4) gene which was confirmed via Sanger sequencing and then screened for on control groups and unrelated infertile subjects. The effect of the mutation on messenger RNA (mRNA) and protein levels was tested after in vitro cell transfection. Structural features of MAGEB4 were predicted throughout the conserved MAGE domain. RESULTS: The novel single-base substitution (c.1041A>T) in the X-linked MAGEB4 gene was identified as a no-stop mutation. The mutation is predicted to add 24 amino acids to the C-terminus of MAGEB4. Our functional studies were unable to detect any effect either on mRNA stability, intracellular localization of the protein, or the ability to homodimerize/heterodimerize with other MAGE proteins. We thus hypothesize that these additional amino acids may affect the proper protein interactions with MAGEB4 partners. CONCLUSION: The whole exome analysis of a consanguineous Turkish family revealed MAGEB4 as a possible new X-linked cause of inherited male infertility. This study provides the first clue to the physiological function of a MAGE protein.


Asunto(s)
Antígenos de Neoplasias/genética , Azoospermia/genética , Genes Ligados a X/genética , Infertilidad Masculina/genética , Proteínas de Neoplasias/genética , Oligospermia/genética , Adulto , Azoospermia/patología , Preescolar , Consanguinidad , Frecuencia de los Genes , Homocigoto , Humanos , Infertilidad Masculina/patología , Masculino , Mutación , Oligospermia/patología , Linaje , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , Secuenciación del Exoma
6.
Methods Mol Biol ; 2770: 123-134, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38351451

RESUMEN

Gene editing in the murine germline is a valuable approach to investigate germ cell maturation and generate mouse models. Several studies demonstrated that CRISPR/Cas9 alters the genome of cultured male mouse germline stem cells delivered by electroporation of plasmids. Recently, we showed proof-of-principle that gene knockout can be effectively targeted in mouse germline stem cells by lipofecting Cas9:gRNA ribonucleoproteins. In this protocol, we describe a simple, fast, and cheap workflow for gene editing via the lipofection of non-integrative ribonucleoproteins in murine male germline stem cells.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Ratones , Animales , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Ribonucleoproteínas/metabolismo , ARN Guía de Sistemas CRISPR-Cas , Células Germinativas/metabolismo
7.
Methods Mol Biol ; 2770: 135-149, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38351452

RESUMEN

Testes have a complex architecture that is compartmentalized into seminiferous tubules with a diameter of approximatively 200 µm in which the germ cells differentiate, surrounded by a basement membrane and interstitium. 3D bioprinting might be used to recreate the compartmentalized testicular architecture in vitro. Directed by a software program, pneumatic microextrusion printers can deposit 3D layers of hydrogel-encapsulated interstitial cells in a controlled manner by applying pressure. Once macroporous-shaped scaffolds resembling seminiferous tubules have been bioprinted with interstitial cells, the epithelial cell fraction can be seeded in the macropores to resemble the in vivo testicular architecture. Moreover, macropores can serve as a delimitation for all testicular cells to reorganize and improve the supply of nutrients to cells through the 3D constructs.


Asunto(s)
Bioimpresión , Espermatogénesis , Masculino , Animales , Ratones , Testículo , Túbulos Seminíferos , Andamios del Tejido , Células Intersticiales del Testículo , Hidrogeles , Ingeniería de Tejidos , Impresión Tridimensional
8.
Reprod Biomed Online ; 26(3): 286-98, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23352099

RESUMEN

In a previous study, meiotic activity was observed in human intratesticular xenografts from peripubertal patients. However, full spermatogenesis could not be established. The present study aimed to evaluate whether the administration of recombinant human FSH could improve the spermatogonial survival and the establishment of full spermatogenesis in intratesticular human xenografts. Human testicular tissue was obtained from six boys (aged 2.5-12.5years). The testicular biopsy was fragmented and one fragment of 1.5-3.0mm(3) was transplanted to the testis of immunodeficient nude mice. Transplanted mice were assigned to different experimental groups to enable evaluation of the effects of FSH administration and freezing. The structural integrity of the seminiferous tubules, the spermatogonial survival and the presence of differentiated cells were evaluated by histology and immunohistochemistry. Freezing or administration of FSH did not influence tubule integrity and germ cell survival in human xenografts. Meiotic germ cells were observed in the xenografts. More tubules containing only Sertoli cells were observed in frozen-thawed grafts, and more tubules with meiotic cells were present in fresh grafts. There was no clear influence of FSH treatment on meiotic differentiation. Administration of FSH did not improve the establishment of full spermatogenesis after intratesticular tissue grafting.


Asunto(s)
Hormona Folículo Estimulante Humana/farmacología , Espermatogénesis/efectos de los fármacos , Espermatogonias/efectos de los fármacos , Testículo/trasplante , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Preescolar , Criopreservación , Humanos , Masculino , Meiosis/efectos de los fármacos , Ratones , Ratones Desnudos , Pubertad , Proteínas Recombinantes/farmacología , Espermatogonias/citología , Trasplante Heterólogo
9.
Front Toxicol ; 5: 1189303, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37265663

RESUMEN

Current test strategies to identify thyroid hormone (TH) system disruptors are inadequate for conducting robust chemical risk assessment required for regulation. The tests rely heavily on histopathological changes in rodent thyroid glands or measuring changes in systemic TH levels, but they lack specific new approach methodologies (NAMs) that can adequately detect TH-mediated effects. Such alternative test methods are needed to infer a causal relationship between molecular initiating events and adverse outcomes such as perturbed brain development. Although some NAMs that are relevant for TH system disruption are available-and are currently in the process of regulatory validation-there is still a need to develop more extensive alternative test batteries to cover the range of potential key events along the causal pathway between initial chemical disruption and adverse outcomes in humans. This project, funded under the Partnership for the Assessment of Risk from Chemicals (PARC) initiative, aims to facilitate the development of NAMs that are specific for TH system disruption by characterizing in vivo mechanisms of action that can be targeted by in embryo/in vitro/in silico/in chemico testing strategies. We will develop and improve human-relevant in vitro test systems to capture effects on important areas of the TH system. Furthermore, we will elaborate on important species differences in TH system disruption by incorporating non-mammalian vertebrate test species alongside classical laboratory rat species and human-derived in vitro assays.

10.
Front Cell Dev Biol ; 10: 891173, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35774227

RESUMEN

Gene editing in male germline stem (GS) cells is a potent tool to study spermatogenesis and to create transgenic mice. Various engineered nucleases already demonstrated the ability to modify the genome of GS cells. However, current systems are limited by technical complexity diminishing application options. To establish an easier method to mediate gene editing, we tested the lipofection of site-specific Cas9:gRNA ribonucleoprotein (RNP) complexes to knockout the enhanced green fluorescent protein (Egfp) in mouse EGFP-GS cells via non-homologous end joining. To monitor whether gene conversion through homology-directed repair events occurred, single-stranded oligodeoxynucleotides were co-lipofected to deliver a Bfp donor sequence. Results showed Egfp knockout in up to 22% of GS cells, which retained their undifferentiated status following transfection, while only less than 0.7% EGFP to BFP conversion was detected in gated GS cells. These data show that CRISPR/Cas9 RNP-based lipofection is a promising system to simply and effectively knock out genes in mouse GS cells. Understanding the genes involved in spermatogenesis could expand therapeutic opportunities for men suffering from infertility.

11.
Front Physiol ; 12: 757565, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35002756

RESUMEN

Short-term germ cell survival and central tissue degeneration limit organoid cultures. Here, testicular organoids (TOs) were generated from two different mouse strains in 3D printed one-layer scaffolds (1LS) at the air-medium interface displaying tubule-like structures and Leydig cell functionality supporting long-term survival and differentiation of germ cells to the meiotic phase. Chimeric TOs, consisting of a mixture of primary testicular cells and EGFP+ germline stem (GS) cells, were cultured in two-layer scaffolds (2LSs) for better entrapment. They showed an improved spheroidal morphology consisting of one intact tubule-like structure and surrounding interstitium, representing the functional unit of a testis. However, GS cells did not survive long-term culture. Consequently, further optimization of the culture medium is required to enhance the maintenance and differentiation of germ cells. The opportunities TOs offer to manipulate somatic and germ cells are essential for the study of male infertility and the search for potential therapies.

12.
Andrology ; 8(4): 879-891, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31823507

RESUMEN

BACKGROUND: The testicular organoid concept has recently been introduced in tissue engineering to refer to testicular cell organizations modeling testicular architecture and function. The testicular organoid approach gives control over which and how cells reaggregate, which is not possible in organotypic cultures, thereby extending the applicability of in-vitro spermatogenesis (IVS) systems. However, it remains unclear which culture method and medium allow reassociation of testicular cells into a functional testicular surrogate in-vitro. OBJECTIVE: The aim of this paper is to review the different strategies that have been used in an attempt to create testicular organoids and generate spermatozoa. We want to provide an up-to-date list on culture methodologies and media compositions that have been used and determine their role in regulating tubulogenesis and differentiation of testicular cells. SEARCH METHOD: A literature search was conducted in PubMed, Web of Science, and Scopus to select studies reporting the reorganization of testicular cell suspensions in-vitro, using the keywords: three-dimensional culture, in-vitro spermatogenesis, testicular organoid, testicular scaffold, and tubulogenesis. Papers published before the August 1, 2019, were selected. OUTCOME: Only a limited number of studies have concentrated on recreating the testicular architecture in-vitro. While some advances have been made in the testicular organoid research in terms of cellular reorganization, none of the described culture systems is adequate for the reproduction of both the testicular architecture and IVS. CONCLUSION: Further improvements in culture methodology and medium composition have to be made before being able to provide both testicular tubulogenesis and spermatogenesis in-vitro.


Asunto(s)
Técnicas de Cultivo de Célula , Organoides/citología , Espermatogénesis/fisiología , Testículo/citología , Animales , Diferenciación Celular/fisiología , Humanos , Masculino , Espermatozoides/citología
13.
Methods Mol Biol ; 1576: 283-290, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-28674937

RESUMEN

Organoid systems take advantage of the self-organizing capabilities of cells to create diverse multi-cellular tissue surrogates that constitute a powerful novel class of biological models. Clearly, the formation of a testicular organoid (TO) in which human spermatogenesis can proceed from a single-cell suspension would exert a tremendous impact on research and development, clinical treatment of infertility, and screening of potential drugs and toxic agents. Recently, we showed that primary adult and pubertal human testicular cells auto-assembled in TOs either with or without the support of a natural testis scaffold. These mini-tissues harboured both the spermatogonial stem cells and their important niche cells, which retained certain specific functions during long-term culture. As such, human TOs might advance the development of a system allowing human in vitro spermatogenesis. Here we describe the methodology to make scaffold-based and scaffold-free TOs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/citología , Células de Sertoli/citología , Espermatogonias/citología , Testículo/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Adulto , Diferenciación Celular , Células Cultivadas , Humanos , Masculino , Espermatogénesis
14.
Biofabrication ; 11(3): 035011, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30921781

RESUMEN

In vitro spermatogenesis (IVS) has already been successfully achieved in rodents by organotypic and soft matrix culture systems. However, the former does not allow single cell input, and the latter presents as a simple thick layer in which all cells are embedded. We explored a new culture system using a mouse model by employing an alginate-based hydrogel and 3D bioprinting, to control scaffold design and cell deposition. We produced testicular constructs consisting of printed cell-free scaffolds (CFS) with prepubertal testicular cells (TC) in their easy-to-access macropores. Here, the pores represented the only cell compartment (TC/CFS). Double-cell compartment testicular constructs were achieved by culturing magnetic-activated cell sorting-enriched epithelial cells in the pores of interstitial cell-laden scaffolds (CD49f+/CLS). Cell spheres formed in the pores in the weeks following cell seeding on both CFS and CLS. Although restoration of the tubular architecture was not observed, patches of post-meiotic cells including elongated spermatids were found in 66% of TC/CFS. Differentiation up to the level of round spermatids and elongated spermatids was observed in all and 33% of CD49f+/CLS constructs, respectively. Organ culture served as the reference method for IVS, with complete spermatogenesis identified in 80% of cultivated prepubertal tissue fragments. So far, this is the first report applying a 3D bioprinting approach for IVS. Further optimization of the scaffold design and seeding parameters might be permissive for tubular architecture recreation and thereby increase the efficiency of IVS in printed testicular constructs. While it remains to be tested whether the gametes generated on the alginate-based scaffolds can support embryogenesis following IVF, this IVS approach might be useful for (patho)physiological studies and drug-screening applications.


Asunto(s)
Alginatos/farmacología , Impresión Tridimensional , Espermatogénesis , Andamios del Tejido/química , Animales , Diferenciación Celular/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Espermatogénesis/efectos de los fármacos , Espermatozoides/citología , Espermatozoides/efectos de los fármacos , Testículo/fisiología , Ingeniería de Tejidos
15.
Methods Mol Biol ; 1577: 121-127, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28456952

RESUMEN

Biological scaffolds composed of extracellular matrix (ECM) are typically derived by processes that involve decellularization of tissues or organs. Here we describe a simple and robust methodology for the preparation of decellularized testicular matrix (DTM) scaffolds with minimal damage to the native three-dimensional structure and tissue-specific ECM components. Such DTM scaffolds can help to gain a better insight into the molecular composition and function of testicular ECM and to develop new tissue engineering approaches to treat various types of male fertility disorders.


Asunto(s)
Matriz Extracelular/química , Testículo/química , Testículo/citología , Andamios del Tejido/química , Diseño de Equipo , Matriz Extracelular/ultraestructura , Proteínas de la Matriz Extracelular/análisis , Humanos , Masculino , Microtomía/instrumentación , Microtomía/métodos , Espermatogénesis , Testículo/ultraestructura , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
16.
Methods Mol Biol ; 1748: 287-294, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29453579

RESUMEN

Tissue cryopreservation uses very low temperatures to preserve structurally intact living cells in their natural microenvironment. Cell survival is strongly influenced by the biophysical effects of ice during both the freezing and the subsequent thawing. These effects can be controlled by optimizing the fragment size, type of cryoprotectant, and cooling rate. The challenge is to determine cryopreservation parameters that suit all cell types present in the tissue. Here we describe a quick and convenient protocol for the cryopreservation of testicular tissue using an isopropyl-insulated freezing device, which was validated in both a mouse and a human model.


Asunto(s)
Alquenos/química , Criopreservación/métodos , Crioprotectores/química , Congelación , Testículo/citología , Animales , Supervivencia Celular , Frío , Humanos , Masculino , Ratones , Modelos Animales , Transición de Fase
17.
Stem Cell Res Ther ; 9(1): 317, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30463610

RESUMEN

BACKGROUND: Spermatogonial stem cell transplantation (SSCT) could become a fertility restoration tool for childhood cancer survivors. However, since in mice, the colonization efficiency of transplanted spermatogonial stem cells (SSCs) is only 12%, the efficiency of the procedure needs to be improved before clinical implementation is possible. Co-transplantation of mesenchymal stem cells (MSCs) might increase colonization efficiency of SSCs by restoring the SSC niche after gonadotoxic treatment. METHODS: A mouse model for long-term infertility was developed and used to transplant SSCs (SSCT, n = 10), MSCs (MSCT, n = 10), a combination of SSCs and MSCs (MS-SSCT, n = 10), or a combination of SSCs and TGFß1-treated MSCs (MSi-SSCT, n = 10). RESULTS: The best model for transplantation was obtained after intraperitoneal injection of busulfan (40 mg/kg body weight) at 4 weeks followed by CdCl2 (2 mg/kg body weight) at 8 weeks of age and transplantation at 11 weeks of age. Three months after transplantation, spermatogenesis resumed with a significantly better tubular fertility index (TFI) in all transplanted groups compared to non-transplanted controls (P < 0.001). TFI after MSi-SSCT (83.3 ± 19.5%) was significantly higher compared to MS-SSCT (71.5 ± 21.7%, P = 0.036) but did not differ statistically compared to SSCT (78.2 ± 12.5%). In contrast, TFI after MSCT (50.2 ± 22.5%) was significantly lower compared to SSCT (P < 0.001). Interestingly, donor-derived TFI was found to be significantly improved after MSi-SSCT (18.8 ± 8.0%) compared to SSCT (1.9 ± 1.1%; P < 0.001), MSCT (0.0 ± 0.0%; P < 0.001), and MS-SSCT (3.4 ± 1.9%; P < 0.001). While analyses showed that both native and TGFß1-treated MSCs maintained characteristics of MSCs, the latter showed less migratory characteristics and was not detected in other organs. CONCLUSION: Co-transplanting SSCs and TGFß1-treated MSCs significantly improves the recovery of endogenous SSCs and increases the homing efficiency of transplanted SSCs. This procedure could become an efficient method to treat infertility in a clinical setup, once the safety of the technique has been proven.


Asunto(s)
Células Madre Germinales Adultas/trasplante , Infertilidad Masculina/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Busulfano/administración & dosificación , Cloruro de Cadmio/administración & dosificación , Supervivencia Celular , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/administración & dosificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Espermatogénesis , Factor de Crecimiento Transformador beta/administración & dosificación
18.
Stem Cell Reports ; 8(1): 30-38, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28017656

RESUMEN

So far, successful de novo formation of testicular tissue followed by complete spermatogenesis in vitro has been achieved only in rodents. Our findings reveal that primary human testicular cells are able to self-organize into human testicular organoids (TOs), i.e., multi-cellular tissue surrogates, either with or without support of a biological scaffold. Despite lacking testis-specific topography, these mini-tissues harbored spermatogonia and their important niche cells, which retained specific functionalities during long-term culture. These observations indicate the posibility of in vitro re-engineering of a human testicular microenvironment from primary cells. Human TOs might help in the development of a biomimetic testicular model that would exert a tremendous impact on research and development, clinical treatment of infertility, and screening in connection with drug discovery and toxicology.


Asunto(s)
Organogénesis , Organoides , Espermatogénesis , Testículo/citología , Testículo/embriología , Biomarcadores , Técnicas de Cultivo de Célula , Células Cultivadas , Citocinas/metabolismo , Expresión Génica , Hormonas/metabolismo , Humanos , Masculino , Nicho de Células Madre , Andamios del Tejido
19.
Fertil Steril ; 104(5): 1244-52.e1-4, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26260199

RESUMEN

OBJECTIVE: To assess whether testicular cell dynamics are altered during long-term culture after testicular tissue cryopreservation. DESIGN: Experimental basic science study. SETTING: Reproductive biology laboratory. PATIENT(S): Testicular tissue with normal spermatogenesis was obtained from six donors. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Detection and comparison of testicular cells from fresh and frozen tissues during long-term culture. RESULT(S): Human testicular cells derived from fresh (n = 3) and cryopreserved (n = 3) tissues were cultured for 2 months and analyzed with quantitative reverse-transcription polymerase chain reaction and immunofluorescence. Spermatogonia including spermatogonial stem cells (SSCs) were reliably detected by combining VASA, a germ cell marker, with UCHL1, a marker expressed by spermatogonia. The established markers STAR, ACTA2, and SOX9 were used to analyze the presence of Leydig cells, peritubular myoid cells, and Sertoli cells, respectively. No obvious differences were found between the cultures initiated from fresh or cryopreserved tissues. Single or small groups of SSCs (VASA(+)/UCHL1(+)) were detected in considerable amounts up to 1 month of culture, but infrequently after 2 months. SSCs were found attached to the feeder monolayer, which expressed markers for Sertoli cells, Leydig cells, and peritubular myoid cells. In addition, VASA(-)/UCHL1(+) cells, most likely originating from the interstitium, also contributed to this monolayer. Apart from Sertoli cells, all somatic cell types could be detected throughout the culture period. CONCLUSION(S): Testicular tissue can be cryopreserved before long-term culture without modifying its outcome, which encourages implementation of testicular tissue banking for fertility preservation. However, because of the limited numbers of SSCs available after 2 months, further exploration and optimization of the culture system is needed.


Asunto(s)
Criopreservación , Preservación de la Fertilidad/métodos , Células Intersticiales del Testículo/fisiología , Células de Sertoli/fisiología , Espermatogénesis , Espermatogonias/fisiología , Testículo/fisiología , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Células Nutrientes , Regulación de la Expresión Génica , Genotipo , Humanos , Células Intersticiales del Testículo/metabolismo , Masculino , Fenotipo , Reproducibilidad de los Resultados , Células de Sertoli/metabolismo , Espermatogénesis/genética , Espermatogonias/metabolismo , Testículo/citología , Testículo/metabolismo , Factores de Tiempo
20.
Fertil Steril ; 97(5): 1152-7.e1-2, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22369773

RESUMEN

OBJECTIVE: To investigate whether solid-surface vitrification (SSV) is an effective cryopreservation strategy regarding the integrity and function of prepubertal mouse testicular tissue. DESIGN: Prospective experimental study. SETTING: Academic research unit. ANIMAL(S): Mice. INTERVENTION(S): Testicular tissue from 5- to 10-day-old GFP(+) mice was cryopreserved with the use of a conventional uncontrolled slow freezing (USF) technique and SSV before intratesticular grafting in busulfan-treated GFP(-) mice. MAIN OUTCOME MEASURE(S): Ultrastructural cryoinjury to spermatogonial stem cells (SSCs) and somatic cells was assessed by electron microscopy. Tubular structure was evaluated by histology, and graft survival and spermatogenic recovery by immunohistochemistry. RESULT(S): The tubular morphology and the proportion of ultrastructural cryodamage were similar between vitrified and slow-frozen testicular fragments. Allografting of tissue after both USF and SSV resulted in a recovery of spermatogenesis similar to fresh samples. CONCLUSION(S): SSV resulted in success rates similar to USF in maintaining testicular cell ultrastructure, tubular morphology, and tissue function. These data provide further evidence that vitrification, being an inexpensive and simple technique, can be considered as an alternative for cryopreservation of prepubertal testicular tissue.


Asunto(s)
Criopreservación , Preservación de la Fertilidad/métodos , Infertilidad Masculina/cirugía , Preservación de Órganos , Testículo/trasplante , Animales , Busulfano , Modelos Animales de Enfermedad , Preservación de la Fertilidad/efectos adversos , Supervivencia de Injerto , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Infertilidad Masculina/inducido químicamente , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Preservación de Órganos/efectos adversos , Recuperación de la Función , Desarrollo Sexual , Espermatogénesis , Testículo/lesiones , Testículo/metabolismo , Testículo/fisiopatología , Testículo/ultraestructura , Factores de Tiempo , Vitrificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA