Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Hum Reprod ; 29(3)2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36688722

RESUMO

Spermatogonial stem cells (SSCs) are the basis of spermatogenesis, a complex process supported by a specialized microenvironment, called the SSC niche. Postnatal development of SSCs is characterized by distinct metabolic transitions from prepubertal to adult stages. An understanding of the niche factors that regulate these maturational events is critical for the clinical application of SSCs in fertility preservation. To investigate the niche maturation events that take place during SSC maturation, we combined different '-omics' technologies. Serial single cell RNA sequencing analysis revealed changes in the transcriptomes indicative of niche maturation that was initiated at 11 years of age in humans and at 8 weeks of age in pigs, as evident by Monocle analysis of Sertoli cells and peritubular myoid cell (PMC) development in humans and Sertoli cell analysis in pigs. Morphological niche maturation was associated with lipid droplet accumulation, a characteristic that was conserved between species. Lipidomic profiling revealed an increase in triglycerides and a decrease in sphingolipids with Sertoli cell maturation in the pig model. Quantitative (phospho-) proteomics analysis detected the activation of distinct pathways with porcine Sertoli cell maturation. We show here that the main aspects of niche maturation coincide with the morphological maturation of SSCs, which is followed by their metabolic maturation. The main aspects are also conserved between the species and can be predicted by changes in the niche lipidome. Overall, this knowledge is pivotal to establishing cell/tissue-based biomarkers that could gauge stem cell maturation to facilitate laboratory techniques that allow for SSC transplantation for restoration of fertility.


Assuntos
Células de Sertoli , Nicho de Células-Tronco , Humanos , Masculino , Adulto , Animais , Suínos , Lactente , Células de Sertoli/metabolismo , Multiômica , Espermatogônias , Espermatogênese/fisiologia , Testículo/metabolismo
2.
Hum Reprod ; 37(9): 2095-2112, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-35856882

RESUMO

STUDY QUESTION: Do spermatogonia, including spermatogonial stem cells (SSCs), undergo metabolic changes during prepubertal development? SUMMARY ANSWER: Here, we show that the metabolic phenotype of prepubertal human spermatogonia is distinct from that of adult spermatogonia and that SSC development is characterized by distinct metabolic transitions from oxidative phosphorylation (OXPHOS) to anaerobic metabolism. WHAT IS KNOWN ALREADY: Maintenance of both mouse and human adult SSCs relies on glycolysis, while embryonic SSC precursors, primordial germ cells (PGCs), exhibit an elevated dependence on OXPHOS. Neonatal porcine SSC precursors reportedly initiate a transition to an adult SSC metabolic phenotype at 2 months of development. However, when and if such a metabolic transition occurs in humans is ambiguous. STUDY DESIGN, SIZE, DURATION: To address our research questions: (i) we performed a meta-analysis of publicly available and newly generated (current study) single-cell RNA sequencing (scRNA-Seq) datasets in order to establish a roadmap of SSC metabolic development from embryonic stages (embryonic week 6) to adulthood in humans (25 years of age) with a total of ten groups; (ii) in parallel, we analyzed single-cell RNA sequencing datasets of isolated pup (n = 3) and adult (n = 2) murine spermatogonia to determine whether a similar metabolic switch occurs; and (iii) we characterized the mechanisms that regulate these metabolic transitions during SSC maturation by conducting quantitative proteomic analysis using two different ages of prepubertal pig spermatogonia as a model, each with four independently collected cell populations. PARTICIPANTS/MATERIALS, SETTING, METHODS: Single testicular cells collected from 1-year, 2-year and 7-year-old human males and sorted spermatogonia isolated from 6- to 8-day (n = 3) and 4-month (n = 2) old mice were subjected to scRNA-Seq. The human sequences were individually processed and then merged with the publicly available datasets for a meta-analysis using Seurat V4 package. We then performed a pairwise differential gene expression analysis between groups of age, followed by pathways enrichment analysis using gene set enrichment analysis (cutoff of false discovery rate < 0.05). The sequences from mice were subjected to a similar workflow as described for humans. Early (1-week-old) and late (8-week-old) prepubertal pig spermatogonia were analyzed to reveal underlying cellular mechanisms of the metabolic shift using immunohistochemistry, western blot, qRT-PCR, quantitative proteomics, and culture experiments. MAIN RESULTS AND THE ROLE OF CHANCE: Human PGCs and prepubertal human spermatogonia show an enrichment of OXPHOS-associated genes, which is downregulated at the onset of puberty (P < 0.0001). Furthermore, we demonstrate that similar metabolic changes between pup and adult spermatogonia are detectable in the mouse (P < 0.0001). In humans, the metabolic transition at puberty is also preceded by a drastic change in SSC shape at 11 years of age (P < 0.0001). Using a pig model, we reveal that this metabolic shift could be regulated by an insulin growth factor-1 dependent signaling pathway via mammalian target of rapamycin and proteasome inhibition. LARGE SCALE DATA: New single-cell RNA sequencing datasets obtained from this study are freely available through NCBI GEO with accession number GSE196819. LIMITATIONS, REASONS FOR CAUTION: Human prepubertal tissue samples are scarce, which led to the investigation of a low number of samples per age. Gene enrichment analysis gives only an indication about the functional state of the cells. Due to limited numbers of prepubertal human spermatogonia, porcine spermatogonia were used for further proteomic and in vitro analyses. WIDER IMPLICATIONS OF THE FINDINGS: We show that prepubertal human spermatogonia exhibit high OXHPOS and switch to an adult-like metabolism only after 11 years of age. Prepubescent cancer survivors often suffer from infertility in adulthood. SSC transplantation could provide a powerful tool for the treatment of infertility; however, it requires high cell numbers. This work provides key insight into the dynamic metabolic requirements of human SSCs across development that would be critical in establishing ex vivo systems to support expansion and sustained function of SSCs toward clinical use. STUDY FUNDING/COMPETING INTEREST(S): This work was funded by the NIH/NICHD R01 HD091068 and NIH/ORIP R01 OD016575 to I.D. K.E.O. was supported by R01 HD100197. S.K.M. was supported by T32 HD087194 and F31 HD101323. The authors declare no conflict of interest.


Assuntos
Infertilidade , Testículo , Adulto , Animais , Pré-Escolar , Humanos , Infertilidade/metabolismo , Masculino , Mamíferos , Camundongos , Proteômica , Espermatogônias , Células-Tronco , Suínos , Testículo/metabolismo
3.
Andrology ; 8(4): 835-841, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31328437

RESUMO

BACKGROUND: Over the last ten years, three-dimensional organoid culture has garnered renewed interest, as organoids generated from primary cells or stem cells with cell associations and functions similar to organs in vivo can be a powerful tool to study tissue-specific cell-cell interactions in vitro. Very recently, a few interesting approaches have been put forth for generating testicular organoids for studying the germ cell niche microenvironment. AIM: To review different model systems that have been employed to study germ cell biology and testicular cell-cell interactions and discuss how the organoid approach can address some of the shortcomings of those systems. RESULTS AND CONCLUSION: Testicular organoids that bear architectural and functional similarities to their in vivo counterparts are a powerful model system to study cell-cell interactions in the germ cell niche. Organoids enable studying samples in humans and other large animals where in vivo experiments are not possible, allow modeling of testicular disease and malignancies and may provide a platform to design more precise therapeutic interventions.


Assuntos
Comunicação Celular , Organoides/citologia , Testículo/citologia , Animais , Técnicas de Cultura de Células , Humanos , Masculino
4.
Andrology ; 5(2): 336-346, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28160442

RESUMO

The study of spermatogenesis in the horse is challenging because of the absence of an in vitro system that is capable of reproducing efficient spermatogenesis and because of the difficulties and costs associated with performing well-controlled studies in vivo. In an attempt to develop novel methods for the study of equine spermatogenesis, we tested whether cells from enzymatically digested pre-pubertal equine testicular tissue were capable of de novo tissue formation and spermatogenesis following xenografting under the back skin of immunocompromised mice. Testes were obtained from normal pre-pubertal colts and dissociated into cell suspensions using trypsin/collagenase digestion. Resulting cell pellets, consisting of both somatic and germ cells, were injected into fascial pockets under the back skin of immunocompromised, castrated mice and maintained for between 1 and 14 months. Mice were killed and grafts were recovered and analyzed. As has been reported for testis cell suspensions from pigs, mice, cattle, and sheep, de novo formation of equine testicular tissue was observed, as evidenced by the presence of seminiferous tubules and an interstitial compartment. There was an increased likelihood of de novo testicular formation as grafting period increased. Using indirect immunofluorescence, we confirmed the presence of spermatogonia in de novo formed seminiferous tubules. However, we found no evidence of meiotic or haploid cells. These results indicate that dissociated pre-pubertal equine testis cells are capable of reorganizing into the highly specialized endocrine and spermatogenic compartments of the testis following ectopic xenografting. However, in spite of the presence of spermatogonia within the seminiferous tubules, spermatogenesis does not occur. Although this technique does allow access to the cells within the seminiferous tubule and interstitial compartments of the equine testis prior to reaggregation, the absence of spermatogenesis will limit its use as a method for the study of testicular function in the horse.


Assuntos
Morfogênese/fisiologia , Túbulos Seminíferos/crescimento & desenvolvimento , Espermatogênese/fisiologia , Espermatogônias/transplante , Testículo/citologia , Transplante Heterólogo , Animais , Cavalos , Masculino , Camundongos
5.
Andrology ; 3(3): 590-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25877677

RESUMO

To study spermatogonial stem cells the heterogeneous testicular cell population first needs to be enriched for undifferentiated spermatogonia, which contain the stem cell population. When working with non-rodent models, this step requires working with large numbers of cells. Available cell separation methods rely on differential properties of testicular cell types such as expression of specific cell surface proteins, size, density, or differential adhesion to substrates to separate germ cells from somatic cells. The objective of this study was to develop an approach that allowed germ cell enrichment while providing efficiency of handling large cell numbers. Here, we report the use of stirred suspension bioreactors (SSB) to exploit the adhesion properties of Sertoli cells to enrich cells obtained from pre-pubertal porcine testes for undifferentiated spermatogonia. We also compared the bioreactor approach with an established differential plating method and the combination of both: SSB followed by differential plating. After 66 h of culture, germ cell enrichment in SSBs provided 7.3 ± 1.0-fold (n = 9), differential plating 9.8 ± 2.4-fold (n = 6) and combination of both methods resulted in 9.1 ± 0.3-fold enrichment of germ cells from the initial germ cell population (n = 3). To document functionality of cells recovered from the bioreactor, we demonstrated that cells retained their functional ability to reassemble seminiferous tubules de novo after grafting to mouse hosts and to support spermatogenesis. These results demonstrate that the SSB allows enrichment of germ cells in a controlled and scalable environment providing an efficient method when handling large cell numbers while reducing variability owing to handling.


Assuntos
Reatores Biológicos , Túbulos Seminíferos/citologia , Células de Sertoli/citologia , Espermatogônias/citologia , Testículo/citologia , Animais , Técnicas de Cultura de Células , Células Cultivadas , Masculino , Camundongos , Espermatogênese/fisiologia , Suínos
6.
Anim Reprod ; 12(1): 93-104, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27390591

RESUMO

Genetically-modified domestic animal models are of increasing significance in biomedical research and agriculture. As authentic ES cells derived from domestic animals are not yet available, the prevailing approaches for engineering genetic modifications in those animals are pronuclear microinjection and somatic cell nuclear transfer (SCNT, also known as cloning). Both pronuclear microinjection and SCNT are inefficient, costly, and time-consuming. In animals produced by pronuclear microinjection, the exogenous transgene is usually inserted randomly into the genome, which results in highly variable expression patterns and levels in different founders. Therefore, significant efforts are required to generate and screen multiple founders to obtain animals with optimal transgene expression. For SCNT, specific genetic modifications (both gain-of-function and loss-of-function) can be engineered and carefully selected in the somatic cell nucleus before nuclear transfer. SCNT has been used to generate a variety of genetically modified animals such as goats, pigs, sheep and cattle; however, animals resulting from SCNT frequently suffer from developmental abnormalities associated with incomplete nuclear reprogramming. Other strategies to generate genetically-modified animals rely on the use of the spermatozoon as a natural vector to introduce genetic material into the female gamete. This sperm mediated DNA transfer (SMGT) combined with intracytoplasmatic sperm injection (ICSI) has relatively high efficiency and allows the insertion of large DNA fragments, which, in turn, enhance proper gene expression. An approach currently being developed to complement SCNT for producing genetically modified animals is germ cell transplantation using genetically modified male germline stem cells (GSCs). This approach relies on the ability of GSCs that are genetically modified in vitro to colonize the recipient testis and produce donor derived sperm upon transplantation. As the genetic change is introduced into the male germ line just before the onset of spermatogenesis, the time required for the production of genetically modified sperm is significantly shorter using germ cell transplantation compared to cloning or embryonic stem (ES) cell based technology. Moreover, the GSC-mediated germline modification circumvents problems associated with embryo manipulation and nuclear reprogramming. Currently, engineering targeted mutations in domestic animals using GSCs remains a challenge as GSCs from those animals are difficult to maintain in vitro for an extended period of time. Recent advances in genome editing techniques such as Zinc-Finger Nucleases (ZFNs), Transcription Activator-like Effector Nucleases (TALENs) and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPRs) greatly enhance the efficiency of engineering targeted genetic change in domestic animals as demonstrated by the generation of several gene knock-out pig and cattle models using those techniques. The potential of GSC-mediated germline modification in making targeted genetic modifications in domestic animal models will be maximized if those genome editing techniques can be applied in GSCs.

7.
Mol Reprod Dev ; 79(4): 255-61, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22231935

RESUMO

Germline stem cells (GSCs) can be used for large animal transgenesis, in which GSCs that are genetically manipulated in vitro are transplanted into a recipient testis to generate donor-derived transgenic sperm. The objectives of this study were to explore a non-viral approach for transgene delivery into goat GSCs and to investigate the efficiency of nucleofection in producing transgenic sperm. Four recipient goats received fractionated irradiation at 8 weeks of age to deplete endogenous GSCs. Germ cell transplantations were performed 8-9 weeks post-irradiation. Donor cells were collected from testes of 9-week-old goats, enriched for GSCs by Staput velocity sedimentation, and transfected by nucleofection with a transgene construct harboring the human growth hormone gene under the control of the goat beta-casein promoter (GBC) and a chicken beta-globin insulator (CBGI) sequence upstream of the promoter. For each recipient, transfected cells from 10 nucleofection reactions were pooled, mixed with non-transfected cells to a total of 1.5 × 10(8) cells in 3 ml, and transplanted into one testis (n = 4 recipients) by ultrasound-guided cannulation of the rete testis. The second testis of each recipient was removed. Semen was collected, starting at 9 months after transplantation, for a period of over a year (a total of 62 ejaculates from four recipients). Nested genomic PCR for hGH and CBGI sequences demonstrated that 31.3% ± 12.6% of ejaculates were positive for both hGH and CBGI. This study provides proof-of-concept that non-viral transfection (nucleofection) of primary goat germ cells followed by germ cell transplantation results in transgene transmission to sperm in recipient goats.


Assuntos
Animais Geneticamente Modificados/genética , Células Germinativas/transplante , Espermatozoides/fisiologia , Transplante de Células-Tronco/métodos , Transfecção/métodos , Transgenes , Animais , Caseínas/genética , Galinhas , Feminino , Genótipo , Células Germinativas/citologia , Cabras , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Regiões Promotoras Genéticas , Espermatozoides/citologia , Células-Tronco/citologia , Testículo/fisiologia , Globinas beta/genética
8.
Mol Reprod Dev ; 78(3): 202-11, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21387453

RESUMO

Embryonic stem (ES) cells with the capacity for germ line transmission have only been verified in mouse and rat. Methods for derivation, propagation, and differentiation of ES cells from domestic animals have not been fully established. Here, we describe derivation of ES cells from goat embryos. In vivo-derived embryos were cultured on goat fetal fibroblast feeders. Embryos either attached to the feeder layer or remained floating and expanded in culture. Embryos that attached showed a prominent inner cell mass (ICM) and those that remained floating formed structures resembling ICM disks surrounded by trophectodermal cells. ICM cells and embryonic disks were isolated mechanically, cultured on feeder cells in the presence of hLIF, and outgrown into ES-like colonies. Two cell lines were cultured for 25 passages and stained positive for alkaline phosphatase, POU5F1, NANOG, SOX2, SSEA-1, and SSEA-4. Embryoid bodies formed in suspension culture without hLIF. One cell line was cultured for 2 years (over 120 passages). This cell line differentiated in vitro into epithelia and neuronal cells, and could be stably transfected and selected for expression of a fluorescent marker. When cells were injected into SCID mice, teratomas were identified 5-6 weeks after transplantation. Expression of known ES cell markers, maintenance in vitro for 2 years in an undifferentiated state, differentiation in vitro, and formation of teratomas in immunodeficient mice provide evidence that the established cell line represents goat ES cells. This also is the first report of teratoma formation from large animal ES cells.


Assuntos
Separação Celular/métodos , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Cabras/embriologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Imuno-Histoquímica , Cariotipagem , Camundongos , Camundongos SCID , Teratoma/etiologia , Teratoma/patologia
10.
Reprod Fertil Dev ; 21(3): 489-97, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19261226

RESUMO

Grafting of immature mammalian testis tissue to mouse hosts can preserve the male germline. To make this approach applicable to a clinical or field situation, it is imperative that the testis tissue and/or spermatozoa harvested from grafted tissue are preserved successfully. The aim of the present study was to evaluate protocols for the preservation of testis tissue in a porcine model. Testis tissue was stored at 4 degrees C for short-term preservation or cryopreserved by slow-freezing, automated slow-freezing or vitrification for long-term storage. Preserved tissue was transplanted ectopically to mouse hosts and recovered xenografts were analysed histologically. In addition, spermatozoa were harvested from xenografts and cryopreserved. Total cell viability and germ cell viability remained high after tissue preservation. Complete spermatogenesis occurred in xenografts preserved by cooling up to 48 h, whereas spermatogenesis progressed to round spermatids in the xenografts that were frozen-thawed before grafting. Approximately 50% of spermatozoa harvested from xenografts remained viable after freezing and thawing. The in vivo developmental potential of cryopreserved tissue was reduced despite high post-thaw viability. Therefore, it is important to evaluate germ cell differentiation in vivo in addition to cell viability in vitro when optimising freezing protocols for testis tissue.


Assuntos
Suínos , Testículo/fisiologia , Testículo/transplante , Preservação de Tecido/veterinária , Animais , Sobrevivência Celular , Temperatura Baixa , Criopreservação/métodos , Criopreservação/veterinária , Temperatura Alta , Masculino , Camundongos , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Testículo/citologia , Preservação de Tecido/métodos , Transplante Heterólogo/veterinária , Transplante Heterotópico/veterinária
11.
Reprod Domest Anim ; 43 Suppl 2: 288-94, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18638137

RESUMO

Transplantation of male germ line stem cells from a donor animal to the testes of an infertile recipient was first described in 1994. Donor germ cells colonize the recipient's testis and produce donor-derived sperm, such that the recipient male can distribute the genetic material of the germ cell donor. Germ cell transplantation represents a functional reconstitution assay for male germ line stem cells and as such has vastly increased our ability to study the biology of stem cells in the testis and define phenotypes of infertility. First developed in rodents, the technique has now been used in a number of animal species, including domestic mammals, chicken and fish. There are three major applications for this technology in animals: first, to study fundamental aspects of male germ line stem cell biology and male fertility; second, to preserve the reproductive potential of genetically valuable individuals by male germ cell transplantation within or between species; third, to produce transgenic sperm by genetic manipulation of isolated germ line stem cells and subsequent transplantation. Transgenesis through the male germ line has tremendous potential in species in which embryonic stem cells are not available and somatic cell nuclear transfer has limited success. Therefore, transplantation of male germ cells is a uniquely valuable approach for the study, preservation and manipulation of male fertility in animals.


Assuntos
Células Germinativas/transplante , Infertilidade Masculina/terapia , Espermatogênese/fisiologia , Testículo/transplante , Animais , Animais Domésticos , Fertilidade/fisiologia , Masculino , Transplante Heterólogo , Transplante Homólogo
12.
Reprod Fertil Dev ; 19(6): 732-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17714627

RESUMO

The transplantation of spermatogonial stem cells between males results in a recipient animal producing spermatozoa carrying a donor's haplotype. First pioneered in rodents, this technique has now been used in several animal species. Importantly, germ cell transplantation was successful between unrelated, immuno-competent large animals, whereas efficient donor-derived spermatogenesis in rodents requires syngeneic or immuno-compromised recipients. Transplantation requires four steps: recipient preparation, donor cell isolation, transplantation and identifying donor-derived spermatozoa. There are two main applications for this technology. First, genetic manipulation of isolated germ line stem cells and subsequent transplantation will result in production of transgenic spermatozoa. Transgenesis through the male germ line has tremendous potential in species in which embryonic stem cells are not available and somatic cell nuclear transfer and reprogramming pose several problems. Second, spermatogonial stem cell transplantation within or between species offers a means of preserving the reproductive potential of genetically valuable individuals. This might have significance in the captive propagation of non-domestic animals of high conservation value. Transplantation of germ cells is a uniquely valuable approach for the study, preservation and manipulation of male fertility in mammalian species.


Assuntos
Animais Domésticos , Animais Selvagens , Cruzamento/métodos , Extinção Biológica , Óvulo/transplante , Espermatogônias/transplante , Animais , Humanos , Masculino , Camundongos , Ratos , Espermatogênese
13.
Artigo em Inglês | MEDLINE | ID: mdl-16903423

RESUMO

Transplantation of male germ line stem cells from a fertile donor to the testis of an infertile recipient restores donor-derived spermatogenesis in the recipient testis and the resulting sperm pass the donor genotype to the offspring of the recipient. Germ cell transplantation has been an invaluable tool to elucidate the biology of male germ line stem cells and their niche in the testis, develop systems to isolate and culture spermatogonial stem cells, examine defects in spermatogenesis, correct male infertility and introduce genetic changes into the male germ line. Although most widely studied in rodents, germ cell transplantation has been applied to larger mammals, including primates. Recently, ectopic grafting of testis tissue from diverse donor species, including primates, into a mouse host has opened an additional possibility to study spermatogenesis and to produce fertile sperm from immature donors. Testis xenografts are ideally suitable to study toxicants or drugs with the potential to enhance or suppress male fertility without the necessity of performing experiments in the target species. Therefore, transplantation of germ cells or xenografting of testis tissue represent powerful approaches for the study, preservation, and manipulation of male fertility.


Assuntos
Transplante de Células/métodos , Espermatogênese , Espermatozoides/citologia , Células-Tronco/citologia , Animais , Humanos , Infertilidade Masculina , Masculino , Fenótipo
14.
Soc Reprod Fertil Suppl ; 62: 331-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16866328

RESUMO

Transplantation of germ cells from fertile donor mice to the testes of infertile recipient mice results in donor-derived spermatogenesis and transmission of the donor haplotype to offspring of recipient animals. In the pig, germ cells can be transplanted to a recipient testis by ultrasound-guided cannulation of the rete testis with delivery of cells by gravity flow. It is important to note that germ cell transplantation was successful between unrelated, immuno-competent pigs, whereas transplantation in rodents requires syngeneic or immuno-compromised recipients. Efficiency of colonization of the recipient testis by donor-derived germ cells can be improved by pretreatment of the recipient animal to deplete endogenous germ cells. Genetic manipulation of isolated germ line stem cells and subsequent transplantation will result in production of transgenic sperm. Transgenesis through the male germ line has tremendous potential in species like pigs where embryonic stem cell technology is not available and current options to generate transgenic animals are inefficient. Introduction of a genetic change prior to fertilization will circumvent problems associated with manipulation of early embryos and developmental abnormalities associated with somatic cell nuclear transfer and reprogramming. Viral transduction of germ cells prior to transplantation has been used to generate transgenic rodents and has also shown early promising results in pigs. Current research is directed toward improving protocols for isolation and culture of porcine male germ cells to increase efficiency of transgene transmission and to allow for gene targeting prior to germ cell transplantation. It is expected that germ cell transplantation will then provide a viable alternate approach to generate germ line transgenic pigs.


Assuntos
Infertilidade Masculina/terapia , Espermatozoides/transplante , Sus scrofa , Animais , Animais Geneticamente Modificados , Masculino , Rede do Testículo , Transfecção/métodos , Transgenes , Transplante Heterólogo
15.
Reproduction ; 131(6): 1091-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16735548

RESUMO

Grafting of testis tissue from immature animals to immunodeficient mice results in complete spermatogenesis, albeit with varying efficiency in different species. The objectives of this study were to investigate if grafting of horse testis tissue would result in spermatogenesis, and to assess the effect of exogenous gonadotropins on xenograft development. Small fragments of testis tissue from 7 colts (2 week to 4 years of age) were grafted under the back skin of castrated male immunodeficient mice. For 2 donor animals, half of the mice were treated with gonadotropins. Xenografts were analyzed at 4 and 8 months post-transplantation. Spermatogenic differentiation following grafting ranged from no differentiation to progression through meiosis with appearance of haploid cells. Administration of exogenous gonadotropins appeared to support post-meiotic differentiation. For more mature donor testis samples where spermatogenesis had progressed into or through meiosis, after grafting an initial loss of differentiated germ cells was observed followed by a resurgence of spermatogenesis. However, if haploid cells had been present prior to grafting, spermatogenesis did not progress beyond meiotic division. In all host mice with spermatogenic differentiation in grafts, increased weight of the seminal vesicles compared to castrated mice showed that xenografts were releasing testosterone. These results indicate that horse spermatogenesis occurs in a mouse host albeit with low efficiency. In most cases, spermatogenesis arrested at meiosis. The underlying mechanisms of this spermatogenic arrest require further investigation.


Assuntos
Espermatogênese , Espermatozoides/citologia , Testículo/transplante , Animais , Gonadotropinas Equinas/farmacologia , Sobrevivência de Enxerto , Masculino , Meiose , Camundongos , Camundongos SCID , Orquiectomia , Túbulos Seminíferos/crescimento & desenvolvimento , Testículo/metabolismo , Testosterona/biossíntese , Transplante Heterólogo
16.
Reprod Fertil Dev ; 18(1-2): 13-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16478598

RESUMO

Male germ cell transplantation is a powerful approach to study the control of spermatogenesis with the ultimate goal to enhance or suppress male fertility. In livestock animals, applications can be expanded to provide an alternative method of transgenesis and an alternative means of artificial insemination (AI). The transplantation technique uses testis stem cells, harvested from the donor animal. These donor stem cells are injected into seminiferous tubules, migrate from the lumen to relocate to the basement membrane and, amazingly, they can retain the capability to produce donor sperm in their new host. Adaptation of the mouse technique for livestock is progressing, with gradual gains in efficiency. Germ cell transfer in goats has produced offspring, but not yet in cattle and pigs. In goats and pigs, the applications of germ cell transplantation are mainly in facilitating transgenic animal production. In cattle, successful male germ cell transfer could create an alternative to AI in areas where it is impractical. Large-scale culture of testis stem cells would enhance the use of elite bulls by providing a renewable source of stem cells for transfer. Although still in a developmental state, germ cell transplantation is an emerging technology with the potential to create new opportunities in livestock production.


Assuntos
Animais Domésticos , Transplante de Células/métodos , Espermatozoides/transplante , Transplante Heterólogo/métodos , Animais , Transplante de Células/tendências , Células Cultivadas , Criopreservação/métodos , Masculino , Preservação do Sêmen/métodos , Especificidade da Espécie , Espermatogônias/citologia , Espermatogônias/fisiologia , Espermatozoides/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
17.
Hum Reprod ; 21(2): 384-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16239313

RESUMO

BACKGROUND: Grafting of testicular tissue into immunodeficient mice has become an interesting and promising scientific tool for the generation of gametes and the study of testicular function. This technique might potentially be used to generate sperm from patients whose testes need to be removed or are destroyed due to therapeutic intervention or as a consequence of disease. Here we explore whether adult human testicular tissue from patients with different testicular pathologies survives as xenograft. METHODS AND RESULTS: Testis tissue from adult patients with varying degrees of spermatogenesis was grafted into two strains of immunodeficient mice (severe combined immunodeficiency, Nu/Nu). Tissue with active spermatogenesis prior to grafting largely regressed. However, testicular tissue survival was better in cases where spermatogenesis was suppressed prior to grafting and occasionally spermatogonial stem cells survived. Cases with spermatogenic disruption were not corrected by the xenografting. CONCLUSION: Superior survival of the germinal epithelium and spermatogonia when spermatogenesis was suppressed prior to grafting could provide a novel strategy for germline preservation in pre-pubertal cancer patients. This approach could also be valuable to study the early stages of human spermatogenesis.


Assuntos
Sobrevivência de Enxerto , Testículo/transplante , Transplante Heterólogo , Adulto , Animais , Coristoma/patologia , Humanos , Infertilidade Masculina/patologia , Masculino , Camundongos , Espermatogênese/fisiologia , Testículo/patologia
19.
Tissue Cell ; 33(2): 200-7, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11392673

RESUMO

Gonadotropin-releasing hormone (GnRH)-agonist or antagonist treatment supports recovery of spermatogenesis after irradiation damage in the rat and appears to be beneficial to colonization of recipient testes after spermatogonial transplantation from fertile donors to the testes of infertile recipients in rats and mice. In the present study, we quantified the effect of treatment of recipient mice with the GnRH-agonist leuprolide acetate on the extent of colonization by donor spermatogonial stem cells in the recipient testis. Testis cells from mice carrying transgenes, which produce beta-galactosidase in spermatogenic cells, were used as donor cells for transplantation to allow for quantification of donor spermatogenesis in the recipient testis by staining for enzyme activity. Donor cell colonization 3 months after transplantation was compared between recipients receiving leuprolide in different treatment protocols and untreated control mice. Two injections of leuprolide 4 weeks apart prior to transplantation with as little as 3.8 mg/kg resulted in a pronounced improvement in the number of donor-derived spermatogenic colonies as well as in the in the area of recipient seminiferous tubules occupied by donor cell spermatogenesis. Improved colonization efficiency by treatment with GnRH-agonist can make the technique of spermatogonial transplantation applicable to situations when only low numbers of donor cells are available.


Assuntos
Antineoplásicos Hormonais/farmacologia , Hormônio Liberador de Gonadotropina/agonistas , Leuprolida/farmacologia , Espermatogônias/citologia , Espermatogônias/transplante , Animais , Transplante de Células , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Espermatogênese , Transplante de Células-Tronco , Células-Tronco/citologia , Testículo/citologia
20.
Mol Reprod Dev ; 57(3): 270-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11013435

RESUMO

Donor-derived spermatogenesis after spermatogonial transplantation to recipient animals could serve as a novel approach to manipulate the male germ line in species where current methods of genetic modification are still inefficient. The objective of the present study was to investigate germ cell transplantation from boars, bulls, and stallions, which are economically important domestic animals, to mouse recipients. Donor testis cells (fresh, cryopreserved, or cultured for 1 month) were transplanted into testes of immunodeficient recipient mice in which endogenous spermatogenesis had been destroyed. Recipient testes were analyzed from 1 to > 12 months after transplantation for the presence of donor germ cells by donor-specific immunohistochemistry. Donor cells were present in most recipient testes with species-dependent differences in pattern and extent of colonization. Porcine donor germ cells formed chains and networks of round cells connected by intercellular bridges but later stages of donor-derived spermatogenesis were not observed. Transplanted bovine testis cells initially appeared similar but then developed predominantly into fibrous tissue within recipient seminiferous tubules. Few equine germ cells proliferated in mouse testes with no obvious difference between cells recovered from a scrotal or a cryptorchid donor testis. The pattern of colonization after transplantation of cultured cells did not resemble spermatogonial proliferation. These results indicate that fresh or cryopreserved germ cells from large animals can colonize the mouse testis but do not differentiate beyond the stage of spermatogonial expansion. Species-specific differences in the compatibility of large animal donors and mouse recipients were detected which cannot be predicted solely on the basis of phylogenetic distance between donor and recipient species.


Assuntos
Transplante de Células , Espermatogênese/fisiologia , Espermatozoides/fisiologia , Testículo , Animais , Animais Domésticos , Bovinos , Transplante de Células/métodos , Cavalos , Masculino , Camundongos , Camundongos Nus , Espermatozoides/citologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...