Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
J Med Primatol ; 45(3): 105-11, 2016 06.
Article in English | MEDLINE | ID: mdl-26987570

ABSTRACT

BACKGROUND: Hypothalamic kisspeptin-kisspeptin receptor signalling in primates ensures the successful progression into puberty during development and maintenance of reproductive capacity during adulthood. Human testis has been shown to express high-to-moderate levels of kisspeptin and kisspeptin receptor gene expression. In this study, we aimed at characterizing the localization of kisspeptin and kisspeptin receptor in adult primate testis tissue. METHODS: Immunocytochemistry was performed on paraffin-embedded testicular sections from adult rhesus monkeys and from common marmoset monkeys. RESULTS: Kisspeptin receptor was detected in Sertoli cells in the periphery of the seminiferous tubules in adult testes of both species. In contrast, kisspeptin was not localized in the seminiferous epithelium and was detected only in the interstitial compartment of the adult rhesus monkey testis. CONCLUSION: Kisspeptin receptor and kisspeptin are localized in the testis of Old World and New World primates.


Subject(s)
Callithrix/metabolism , Immunohistochemistry , Kisspeptins/metabolism , Macaca mulatta/metabolism , Receptors, G-Protein-Coupled/metabolism , Testis/chemistry , Animals , Male , Seminiferous Tubules/chemistry , Sertoli Cells/chemistry , Species Specificity
2.
Theriogenology ; 84(9): 1620-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410833

ABSTRACT

A common feature of assisted reproductive techniques such as IVF or intracytoplasmic sperm injection is the IVC of oocytes or preimplantation embryos in artificial culture media. The IVC conditions are selected to mimic the environment of the female genital tract. We have shown that murine preimplantation embryos respond to different culture media with changes in developmental rates, cellular lineage composition, and gene expression patterns. In this study, we explored whether apoptosis is responsible for cell loss in mouse preimplantation embryos after exposure to different human culture media. We examined total embryonic cell count as well as the pattern of protein expression for caspase-9 (intrinsic pathway), caspase-8 (extrinsic pathway), and the executioner caspase-3 via immunohistochemical staining. Total cell counts decline in embryos cultured either in innovative sequential medium 1 and Blast Assist (Origio) or human tubal fluid and MultiBlast (Irvine Scientific) when compared to KSOM(aa). Few cells were caspase-9 and -3 positive in all experimental groups. Staining for caspase-8 was almost undetectable. We conclude that embryonic cell loss is not associated with higher rates of intrinsic apoptotic cell loss. Our results suggest that the culture medium-dependent decline in total cell count and the developmental restriction in embryos cultured in innovative sequential medium 1/Blast Assist and human tubal fluid/MultiBlast are related to processes affecting cell proliferation.


Subject(s)
Apoptosis/drug effects , Blastocyst/physiology , Culture Media , Embryo Culture Techniques/methods , Animals , Embryo Transfer , Female , Male , Mice
3.
PLoS One ; 10(2): e0117839, 2015.
Article in English | MEDLINE | ID: mdl-25692788

ABSTRACT

Establishment and maintenance of the correct epigenetic code is essential for a plethora of physiological pathways and disturbed epigenetic patterns can provoke severe consequences, e.g. tumour formation. In recent years, epigenetic drugs altering the epigenome of tumours actively have been developed for anti-cancer therapies. However, such drugs could potentially also affect other physiological pathways and systems in which intact epigenetic patterns are essential. Amongst those, male fertility is one of the most prominent. Consequently, we addressed possible direct effects of two epigenetic drugs, decitabine and vorinostat, on both, the male germ line and fertility. In addition, we checked for putative transgenerational epigenetic effects on the germ line of subsequent generations (F1-F3). Parental adult male C57Bl/6 mice were treated with either decitabine or vorinostat and analysed as well as three subsequent untreated generations derived from these males. Treatment directly affected several reproductive parameters as testis (decitabine & vorinostat) and epididymis weight, size of accessory sex glands (vorinostat), the height of the seminiferous epithelium and sperm concentration and morphology (decitabine). Furthermore, after decitabine administration, DNA methylation of a number of loci was altered in sperm. However, when analysing fertility of treated mice (fertilisation, litter size and sex ratio), no major effect of the selected epigenetic drugs on male fertility was detected. In subsequent generations (F1-F3 generations) only subtle changes on reproductive organs, sperm parameters and DNA methylation but no overall effect on fertility was observed. Consequently, in mice, decitabine and vorinostat neither affected male fertility per se nor caused marked transgenerational effects. We therefore suggest that both drugs do not induce major adverse effects-in terms of male fertility and transgenerational epigenetic inheritance-when used in anti-cancer-therapies.


Subject(s)
Azacitidine/analogs & derivatives , Fertility/drug effects , Hydroxamic Acids/adverse effects , Animals , Azacitidine/adverse effects , DNA Methylation/drug effects , Decitabine , Endocrine Disruptors/adverse effects , Epididymis/drug effects , Epididymis/metabolism , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Spermatozoa/drug effects , Spermatozoa/metabolism , Testis/drug effects , Testis/metabolism , Vorinostat
4.
PLoS One ; 9(12): e112598, 2014.
Article in English | MEDLINE | ID: mdl-25460567

ABSTRACT

In mice the chemokine Cxcl12 and its receptor Cxcr4 participate in maintenance of the spermatogonial population during postnatal development. More complexity arises since Cxcl12 also binds to the non-classical/atypical chemokine receptor Cxcr7. We explored the expression pattern of Cxcl12, Cxcr4 and Cxcr7 during postnatal development in mouse testes and investigated the response of Cxcl12, Cxcr4, Cxcr7 and SSC-niche associated factors to busulfan-induced germ cell depletion and subsequent recovery by RNA expression analysis and localization of the proteins. In neonatal testes transcript levels of Cxcl12, Cxcr4 and Cxcr7 were relatively low and protein expression of Cxcr7 was restricted to gonocytes and spermatogonia. During development, RNA expression of Cxcl12 remained stable but that of Cxcr4 and Cxcr7 increased. Cxcr7 was expressed in germ cells located at the basement membrane of the seminiferous tubules. In adult testes, transcript levels of Cxcl12 were highest while the localization of Cxcr7 did not change. Following germ cell depletion, a significantly increased expression of Cxcl12 and a decreased expression of Cxcr7 were observed. Germ cells repopulating the seminiferous tubules were immunopositive for Cxcr7. We conclude that Cxcr7 expression to be restricted to premeiotic germ cells throughout postnatal testicular development and during testicular recovery. Hence, the spermatogonial population may not only be simply controlled by interaction of Cxcl12 with Cxcr4 but may also involve Cxcr7 as an important player.


Subject(s)
Chemokine CXCL12/metabolism , Gene Expression Regulation , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Spermatogenesis/physiology , Testis/metabolism , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Gene Expression Profiling , Immunohistochemistry , Male , Mice , Microscopy, Fluorescence , Organ Size/drug effects , Spermatogonia/physiology , Testis/drug effects
5.
Mol Hum Reprod ; 20(12): 1169-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25323971

ABSTRACT

Spermatogenesis is a long and complex process that, despite the shared overall goal of producing the male gamete, displays striking amounts of interspecific diversity. In this review, we argue that sperm competition has been an important selection pressure acting on multiple aspects of spermatogenesis, causing variation in the number and morphology of sperm produced, and in the molecular and cellular processes by which this happens. We begin by reviewing the basic biology of spermatogenesis in some of the main animal model systems to illustrate this diversity, and then ask to what extent this variation arises from the evolutionary forces acting on spermatogenesis, most notably sperm competition. We explore five specific aspects of spermatogenesis from an evolutionary perspective, namely: (i) interspecific diversity in the number and morphology of sperm produced; (ii) the testicular organizations and stem cell systems used to produce them; (iii) the large number and high evolutionary rate of genes underpinning spermatogenesis; (iv) the repression of transcription during spermiogenesis and its link to the potential for haploid selection; and (v) the phenomenon of selection acting at the level of the germline. Overall we conclude that adopting an evolutionary perspective can shed light on many otherwise opaque features of spermatogenesis, and help to explain the diversity of ways in which males of different species perform this fundamentally important process.


Subject(s)
Biological Evolution , Reproduction , Sexual Behavior, Animal , Sperm-Ovum Interactions , Spermatogenesis , Spermatozoa/physiology , Testis/physiology , Animals , Cell Shape , Female , Fertilization , Gene Expression Regulation, Developmental , Humans , Male , Selection, Genetic , Species Specificity , Sperm Count , Transcription, Genetic
6.
Semin Cell Dev Biol ; 29: 2-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24685618

ABSTRACT

This review describes the regulation of spermatogenesis taking into consideration the hypothalamic-pituitary gonadal axis, the male reproductive organs and the endocrine and paracrine factors involved in the control of sperm production and the release of androgens. Instead of detailed descriptions of many hormones and growth factors, we attempt to provide an integrative and evolutionary view by comparing different species and considering their specific needs for successful male reproduction. The review focuses on species specific differences in the structural organization of spermatogenesis and indicates that the crucial regulatory mechanisms controlling sperm output are targeted toward differentiating spermatogonia when they initiate clonal expansion. We argue that the further differentiation of germ cells is following a highly coordinated and strictly predetermined morphogenetic cascade widely independent of hormonal control. We propose a hypothetical "ancient" model. Spermatogenesis and steroidogenesis are controlled by a master switch (GnRH pulse generator) under whose control two separate feedback systems provide independent control of androgen (LH-testosterone) and sperm production (FSH-inhibin). This scenario offers high flexibility and has seen uncountable adaptions to optimize the specific needs of different species. Models for the hormonal regulation in hamsters, laboratory rodents and primates are presented to illustrate the species specific diversity.


Subject(s)
Spermatogenesis/physiology , Spermatogonia/cytology , Spermatozoa/cytology , Testis/physiology , Animals , Cell Differentiation , Cell Proliferation , Cricetinae , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Inhibins/biosynthesis , Luteinizing Hormone/metabolism , Macaca , Male , Mice , Rats , Testosterone/biosynthesis
7.
Tissue Cell ; 46(1): 86-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24411714

ABSTRACT

Testicular organogenesis in vitro requires an environment allowing a reassembly of testicular cell types. Previous in vitro studies using male murine germ cells cultured in a defined three-dimensional environment demonstrated tubulogenesis and differentiation into spermatozoa. Combining scaffolds as artificial culture substrates with testicular cell culture, we analysed the colonization of collagen sponges by rat testicular cells focusing on cell survival and reassembly of tubule-like-structures in vitro. Isolated testicular cells obtained from juvenile Sprague Dawley and eGFP transgenic rats were cultured on collagen sponges (DMEM high glucose+Glutamax, 35°C, 5% CO2 with or without gonadotropins). Live cell imaging revealed the colonization of cells across the entire scaffold for up to 35 days. After two days, histology showed cell clusters attached to the collagen fibres and displaying signs of tubulogenesis. Clusters consisted mainly of Sertoli and peritubular cells which surrounded some undifferentiated spermatogonia. Flow cytometry confirmed lack of differentiation as no haploid cells were detected. Leydig cell activity was detected by a rise of testosterone after gonadotropin stimulation. Our approach provides a novel method which is in particular suitable to follow the somatic testicular cells in vitro an issue of growing importance for the analysis of germ line independent failure of spermatogenesis.


Subject(s)
Leydig Cells/cytology , Spermatogenesis/physiology , Spermatogonia/cytology , Testis/cytology , Testosterone/metabolism , Animals , Animals, Newborn , Cells, Cultured , Male , Organogenesis/physiology , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology , Testis/metabolism
8.
Hum Reprod ; 29(2): 220-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24324026

ABSTRACT

STUDY QUESTION: Is post-implantation embryonic development after blastocyst transfer affected by exposure to different assisted reproduction technology (ART) culture media? SUMMARY ANSWER: Fetal development and placental histology of ART embryos cultured in vitro in different ART media was not impaired compared with embryos grown in vivo. WHAT IS KNOWN ALREADY: The application of different in vitro culture (IVC) media for human ART has an effect on birthweight of newborns. In the mouse model, differences in blastocyst formation were reported after culture in different ART media. Moreover, abnormalities in the liver and heart have been detected as a result of suboptimal IVC conditions. STUDY DESIGN, SIZE, DURATION: Fertilized oocytes from inbred and outbred breeding schemes were retrieved and either immediately transferred to foster mothers or incubated in control or human ART culture media up to the blastocyst stage prior to transfer. Placental and fetal anatomy and particularly bone development were evaluated. PARTICIPANTS/MATERIALS, SETTING, METHODS: B6C3F1 female mice were used as oocyte donors after ovulation induction. C57Bl/6 and CD1 males were used for mating and CD1 females as foster mothers for embryo transfer. Fertilized oocytes were recovered from mated females and incubated in sequential human ART media (ISM1/ISM2 and HTF/Multiblast), in control media [KSOM(aa) and Whitten's medium] or grown in utero without IVC (zygote control). As in vivo, control B6C3F1 females were superovulated and left untreated. Fetuses and placentae were isolated by Caesarean section and analysed at 18.5 days post-coitum (dpc) for placenta composition and at 15.5 dpc for body weight, crown-rump length (CRL), fetal organ development, morphological development, total bone length and extent of bone ossification. MAIN RESULTS AND THE ROLE OF CHANCE: No major differences in the number of implantation sites or in histological appearance of the placentae were detected. CRL of KSOM(aa) fetuses was higher compared with zygote control and Whitten's medium. Histological analysis of tissue sections revealed no gross morphological differences compared with the in vitro groups or in vivo controls. Furthermore, no changes in skeletal development and degree of ossification were observed. However, fibula and tibia of ISM1/ISM2 fetuses were longer than the respective ones from in vivo fetuses. LIMITATIONS, REASONS FOR CAUTION: Findings in the mouse embryo and fetus may not be fully transferable to humans. In addition to skeletal development and placentation, there may be other parameters, e.g. on the molecular level which respond to IVC in ART media. Some comparisons have limited statistical power. WIDER IMPLICATIONS OF THE FINDINGS: Our data suggest that once implantation is achieved, subsequent post-implantation development unfolds normally, resulting in healthy fetuses. With mouse models, we gather information for the safety of human ART culture media. Our mouse study is reassuring for the safety of ART conditions on human embryonic development, given the lack of bold detrimental effects observed in the mouse model. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Deutsche Forschungsgemeinschaft (BO 2540/4-1 and SCHL 394/9-1) and by the Nederlandse Organisatie voor Wetenschappelijk Onderzoek (S.L.G.); Bilateral grant NWO-DFG 63-258. None of the authors has any conflict of interest to declare. TRIAL REGISTRATION NUMBER: Not applicable.


Subject(s)
Culture Media/chemistry , Embryo Culture Techniques , Embryo Implantation , Embryo Transfer/methods , Reproductive Techniques, Assisted/instrumentation , Animals , Blastocyst/cytology , Bone and Bones/embryology , Cartilage/embryology , Female , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Oocytes/cytology , Pregnancy , Pregnancy, Animal
9.
Reproduction ; 147(3): 253-64, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24298047

ABSTRACT

Klinefelter's syndrome is a male sex-chromosomal disorder (47,XXY), causing hypogonadism, cognitive and metabolic deficits. The majority of patients are infertile due to complete germ cell loss after puberty. As the depletion occurs during development, the possibilities to study the underlying causes in humans are limited. In this study, we used the 41,XX(Y*) mouse model to characterise the germ line postnatally. We examined marker expression of testicular cells focusing on the spermatogonial stem cells (SSCs) and found that the number of germ cells was approximately reduced fivefold at day 1pp in the 41,XX(Y*) mice, indicating the loss to start prenatally. Concurrently, immunohistochemical SSC markers LIN28A and PGP9.5 also showed decreased expression on day 1pp in the 41,XX(Y*) mice (48.5 and 38.9% of all germ cells were positive), which dropped to 7.8 and 7.3% on 3dpp, and were no longer detectable on days 5 and 10pp respectively. The differences in PCNA-positive proliferating cells in XY* and XX(Y*) mice dramatically increased towards day 10pp. The mRNA expression of the germ cell markers Lin28a (Lin28), Pou5f1 (Oct4), Utf1, Ddx4 (Vasa), Dazl, and Fapb1 (Sycp3) was reduced and the Lin28a regulating miRNAs were deregulated in the 41,XX(Y*) mice. We suggest a model for the course of germ cell loss starting during the intrauterine period. Neonatally, SSC marker expression by the already lowered number of spermatogonia is reduced and continues fading during the first postnatal week, indicating the surviving cells of the SSC population to be disturbed in their stem cell characteristics. Subsequently, the entire germ line is then generally lost when entering meiosis.


Subject(s)
Germ Cells/pathology , Klinefelter Syndrome/pathology , RNA-Binding Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Count , Disease Models, Animal , Gene Expression Regulation, Developmental , Klinefelter Syndrome/genetics , Klinefelter Syndrome/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Proteins/genetics , Sexual Maturation/genetics , Spermatogonia/pathology , Testis/metabolism , Testis/pathology , Y Chromosome/genetics
10.
Metallomics ; 5(10): 1440-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934158

ABSTRACT

A novel quantification approach for tissue imaging using laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) based on tissue embedding in cold-curing resins (Technovit 7100) is presented. With respect to massive side effects on cisplatin, the platinum distribution at different time intervals after cisplatin treatment of mice was determined quantitatively in different tissues including cochlea, testis and kidney. For this purpose, cold-curing resin blocks spiked with different amounts of platinum acetyl acetonate prior to curing were ablated after sectioning at 5 µm thickness and were analysed using ICP-MS after microwave digestion. High spatial resolution and limits of detection in the low ppb range (8 µg kg(-1)) were achieved using a simple and efficient sample preparation. External calibration using the Technovit 7100 standards proved to yield precise and reproducible quantification results. The distribution and retention behaviour of cisplatin in the organs was investigated using the new calibration method.


Subject(s)
Diagnostic Imaging/methods , Laser Therapy , Organ Specificity , Platinum/metabolism , Polymers/chemistry , Spectrophotometry, Atomic , Animals , Calibration , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
11.
Cell Tissue Res ; 354(2): 623-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23873629

ABSTRACT

TCam-2 cells are the main in vitro model for investigations into seminomatous tumors. However, despite their widespread use, questions remain regarding the cells' homogeneity and consequently how representative they are of seminomas. We assess the TCam-2 cell line using routine and novel authentication methods to determine its homogeneity, identify any cellular sub-populations and resolve whether any changes could be due to generational differentiation. TCam-2, embryonal carcinoma cells (2102EP) and breast cancer cell (MCF7) lines were assessed using qRT-PCR, immunocytochemistry, flow cytometry and short tandem repeat analyses. Raman maps of individual cells (minimum of 10) and single scan spectra from 200 cells per culture were obtained. TCam-2s displayed the characteristic marker gene expression pattern for seminoma, were uniform in size and granularity and short tandem repeat analysis showed no contamination. However, based only on physical parameters, flowcytometry was unable to differentiate between TCam-2 and 2102EPs. Raman maps of TCam-2s comprised three equally distributed, distinct spectral patterns displaying large intercellular single spectral variation. All other cells showed little variation. Principal component, cluster and local spectral angle analyses indicated that the TCam-2s contained two different types of cells, one of which comprised two subgroups and was similar to some 2102EP cells. Protein expression corroborated the presence of different cells and generational differences. The detailed characterization provided by the Raman spectra, augmented by the routine methods, provide substantiation to the long-held suspicion that TCam-2 are not homogeneous but comprise differing cell populations, one of which may be embryonal carcinoma in origin.


Subject(s)
Seminoma/diagnosis , Spectrum Analysis, Raman/methods , Testicular Neoplasms/diagnosis , Breast Neoplasms/chemistry , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Male , Seminoma/chemistry , Seminoma/pathology , Testicular Neoplasms/chemistry , Testicular Neoplasms/pathology
12.
Reproduction ; 145(4): 439-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23431271

ABSTRACT

Marmosets are used as preclinical model in reproductive research. In contrast to other primates, they display short gestation times rendering this species valid for exploration of effects on fertility. However, their peculiar endocrine regulation differs from a those of macaques and humans. We subjected male marmosets to previously clinically tested hormonal regimens that are known to effectively suppress spermatogenesis. Beside a control group, seven groups (each n=6) were investigated for different periods of up to 42 months: regimen I, (four groups) received testosterone undecanoate (TU) and norethisterone enanthate (NETE); regimen II, (two groups) received TU and NETE followed by NETE only; and regimen III, (one group) received NETE only. Testicular volume, cell ploidy and histology, endocrine changes and fertility were monitored weekly. TU and NETE and initial TU and NETE treatment followed by NETE failed to suppress spermatogenesis and fertility. Testicular volumes dropped, although spermatogenesis was only mildly affected; however, testicular cellular composition remained stable. Serum testosterone dropped when NETE was given alone but the animals remained fertile. Compared with controls, no significant changes were observed in sperm motility and fertility. Administration of TU and NETE affected testicular function only mildly, indicating that the regulatory role of chorionic gonadotrophin and testosterone on spermatogenesis is obviously limited and testicular function is maintained, although the endocrine axis is affected by the treatment. In conclusion, marmosets showed a different response to regimens of male contraception from macaques or men and have to be considered as a problematic model for preclinical trials of male hormonal contraception.


Subject(s)
Antispermatogenic Agents/administration & dosage , Callithrix/blood , Fertility/drug effects , Norethindrone/analogs & derivatives , Testosterone/analogs & derivatives , Animals , Body Weight/drug effects , Chorionic Gonadotropin/metabolism , Epididymis/drug effects , Male , Models, Animal , Norethindrone/administration & dosage , Organ Size , Pituitary Gland/metabolism , Ploidies , Sperm Motility , Testis/drug effects , Testis/metabolism , Testosterone/administration & dosage , Testosterone/blood
13.
Spermatogenesis ; 2(4): 245-252, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23248765

ABSTRACT

Many previous studies have aimed at spermatogenesis of male murine germ cells in vitro, but no efficient system has been established yet that covers the entire process of mammalian spermatogenesis in a culture dish permanently. In this review, we report on the requirements of spermatogenesis and the current state of different culture methods using testicular tissue fragments, single cell suspensions or three-dimensional culture environments.

14.
Cancer Res ; 72(20): 5174-8, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22902414

ABSTRACT

Boys faced with future sterility as a result of the need of a sterilizing cancer therapy might avoid this fate by engraftment of cryopreserved immature testicular tissue after therapy is completed. Efforts to address this important survivorship issue have been encouraged by reports of the long-term survival and proliferation of human spermatogonia after xenotransplant of cryopreserved immature testicular tissue into immunocompromised murine hosts. However, spermatogenic arrest at the pachytene spermatocyte stage that occurs in this situation has been associated with a failure in sperm production. In this study, we used a prepubescent simian model to address the possibility that testicular tissue engraftment is insufficiently supported in the model to allow suitable maturation of germ cells. Briefly, we carried out autologous orthotopic grafting of cryopreserved testicular tissue from four prepubescent monkeys and one pubescent rhesus monkey after testicular irradiation and castration of the host animal. Five months after implantation of scrotal grafts, we determined that 3% to 7% of the autografts could be recovered with spermatogenesis proceeding through spermatozoa formation in 13% to 17% of the seminiferous tubules formed in the grafts. In contrast, Sertoli cell-only tubules were detected in parallel xenografts transplanted into immunocompromised mice. Our results show that cryopreservation of testicular tissue from prepubescent primates can maintain the fully functional capacity of spermatogonia to produce sperm, but that host conditions are critical for spermatogenic maturation. Furthermore, our results establish an initial perspective on the quantity of cryopreserved material needed to ensure success in preserving fertility through testicular tissue grafts.


Subject(s)
Antineoplastic Agents/toxicity , Cryopreservation , Fertility/drug effects , Sexual Maturation , Testis/transplantation , Animals , Macaca mulatta , Male
15.
Cells Tissues Organs ; 196(6): 543-54, 2012.
Article in English | MEDLINE | ID: mdl-22739409

ABSTRACT

The marmoset monkey is a valuable model in reproductive medicine. While previous studies have evaluated germ cell dynamics in the postnatal marmoset, the features of testicular somatic cells remain largely unknown. Therefore, the aim of this study was to establish marmoset-specific markers for Sertoli and peritubular cells (PTCs) and to compare protocols for the enrichment and culture of testicular cell types. Immunohistochemistry of Sertoli and PTC-specific markers - anti-müllerian hormone (AMH), vimentin (VIM), α-smooth muscle actin (SMA) - was performed and corresponding RNA expression profiles were established by quantitative PCR analysis (SOX9,AMH, FSHR,VIM, and SMA). For these analyses, testicular tissue from newborn (n = 4), 8-week-old (n = 4) and adult (n = 3) marmoset monkeys was used. Protocols for the enrichment and culture of testicular cell fractions from the 8-week-old marmoset monkeys (n = 3) were evaluated and cells were analyzed using germ cell- and somatic cell-specific markers. The expression of AMH, VIM and SMA reflects the proportion and differentiation status of Sertoli and PTCs at the RNA and the protein levels. While applied protocols did not support the propagation of germ cells in vitro, our analyses revealed that PTCs maintain their proliferative potential and constitute the dominant cell type after short- and long-term culture. Expression of functionally meaningful testicular somatic markers is similar in the human and the marmoset monkey, indicating that this primate can indeed be used as model for human testicular development. The PTC culture system established in this study will facilitate the identification of factors influencing male sex differentiation and spermatogenesis.


Subject(s)
Callithrix/anatomy & histology , Germ Cells/cytology , Sertoli Cells/cytology , Testis/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Humans , Immunohistochemistry , Male , Testis/embryology
16.
Theriogenology ; 77(2): 299-310, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21958640

ABSTRACT

The loss of genetic diversity due to premature death of valuable individuals is a significant problem in animal conservation programs, including endangered felids. Testis tissue xenografting has emerged as a system to obtain spermatozoa from dead immature animals, however protocols to store this tissue before xenografting are still lacking. This study focused on testis tissue cryopreservation and storage from the domestic cat (Felis catus) classified as "pre-pubertal" and "pubertal" according to spermatogenesis development. Grafts from testis tissue cryopreserved with DMSO 1.4M, recovered after 10 weeks xenografting, presented seminiferous tubules with no germ cells. On the contrary, testis tissue from pre-pubertal animals preserved in ice-cold medium for 2 to 5 days presented no loss of viability or spermatogenic potential, while the number of grafts of pubertal cat testis tissue with germ cells after 10 weeks of xenografting decreased with increasing storage time. Nevertheless, even grafts from pre-pubertal cat testis tissue presented lower anti-DDX4 and anti-BOULE staining (proteins necessary for the meiosis completion), when compared with adult cat testis. Finally, a strong correlation found between testis weight and xenograft outcome may help choose good candidates for xenografting.


Subject(s)
Cats , Endangered Species , Spermatogenesis , Testis/transplantation , Tissue Preservation/veterinary , Transplantation, Heterologous/veterinary , Animals , Cold Temperature , Cryopreservation/methods , Cryopreservation/veterinary , Felidae , Male , Mice , Mice, Nude , Organ Size , Sexual Maturation , Testis/anatomy & histology , Testis/growth & development , Time Factors , Tissue Preservation/methods , Treatment Outcome
17.
Hum Reprod ; 26(8): 1945-54, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21613315

ABSTRACT

BACKGROUND: Although infertility is a serious concern in survivors of pediatric cancers, little is known about the influence of the degree of sexual maturation at the time of irradiation on spermatogenic recovery after treatment. Thus, we address this question in a non-human primate model, the rhesus monkey (Macaca mulatta). METHODS: Two pubertal (testis size 3 and 6.5 ml, no sperm in ejaculate) and four prepubertal (testis size 1 ml, no sperm in ejaculate) macaques were submitted to a single fraction of testicular irradiation (10 Gy). Unilateral autologous transfer of cryopreserved testis cells was performed 2 months after irradiation. Testicular volume, histology and semen parameters were analyzed to assess irradiation effects and testicular recovery. RESULTS: Irradiation provoked acute testis involution only in the two pubertal monkeys. Subsequently, testis sizes recovered and sperm was present in the ejaculates. Longitudinal outgrowth of seminiferous tubules continued, and, in testes without autologous cell transfer, 4-22% of tubular cross sections showed spermatogenesis 2 years after irradiation. In contrast, the four prepubertal monkeys showed neither a detectable involution as direct response to irradiation, nor a detectable growth of seminiferous tubules later. However, two of these animals showed spermarche 2 years after irradiation, and 8-12% of tubules presented spermatogenesis. One prepubertally irradiated monkey presented fast growth of one testis after cell transfer, and showed spermarche 1 year after irradiation. The infused testis had spermatogenesis in 70% of the tubules. The contralateral testis remained smaller. CONCLUSION: We conclude that irradiation before puberty has a severe detrimental effect on outgrowth of seminiferous tubules. But, within the seminiferous epithelium, spermatogenetic recovery occurs at a low rate with no detectable relation to the maturity of the epithelium at irradiation. We also show that autologous testis cell transplantation can enhance spermatogenesis, but only in isolated cases.


Subject(s)
Germ Cells/transplantation , Seminiferous Tubules/growth & development , Spermatogenesis/radiation effects , Testis/radiation effects , Animals , Macaca mulatta , Male , Puberty , Seminiferous Tubules/radiation effects , Sexual Maturation , Spermatogenesis/physiology , Testis/anatomy & histology , Testis/physiology
18.
Best Pract Res Clin Endocrinol Metab ; 25(2): 287-302, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21397199

ABSTRACT

The testis has been shown to be highly susceptible to the toxic effects of cancer therapy at all stages of life. Young cancer survivors are approximately half as likely as their siblings to sire a pregnancy. Radiation therapy to the testes and high cumulative dose of alkylating agents are the major factors decreasing the probability of fertility. This review aims to present an overview of the current state of knowledge in mechanisms how human spermatogonia proliferate and differentiate and how cancer therapy affects germ cells, what are the options for fertility preservation and what are the clinical risks and limitations related to such procedures. This area of research is discussed in the context of the potential future options that may become available for preserving fertility in male cancer patients.


Subject(s)
Antineoplastic Agents/adverse effects , Fertility/drug effects , Fertility/radiation effects , Neoplasms/drug therapy , Neoplasms/radiotherapy , Testis/physiology , Animals , Antineoplastic Agents, Alkylating/toxicity , Carboplatin/toxicity , Cell Differentiation , Child , Cisplatin/toxicity , Cryopreservation , Cyclophosphamide/toxicity , Germ Cells/transplantation , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infertility, Male/prevention & control , Leydig Cells/radiation effects , Male , Neoplasm Seeding , Puberty , Semen Preservation , Spermatogonia/drug effects , Spermatogonia/physiology , Spermatogonia/radiation effects , Testis/drug effects , Testis/radiation effects , Testis/transplantation
19.
Endocrinology ; 151(8): 4018-23, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20555023

ABSTRACT

The hypothalamic-pituitary-gonadal (HPG) axis is involved in both the regulation of growth of the developing testis and in controlling spermatogenic and steroidogenic activity in the adult testis. Here, we develop a novel testicular xenografting model to examine to which degree testicular growth and function are controlled by intra- and extratesticular factors. Two or eight halves of neonatal Djungarian hamster testes were implanted into intact, hemicastrated, or castrated nude mouse recipients, and the development of the grafts under reduced or increased competition of testicular tissue was monitored and analyzed. We hypothesized that the outgrowth of the testicular grafts is influenced by the total amount of testicular tissue present in a host and that less testicular tissue in a host would result in more extended outgrowth of the grafts. Our results reveal that the hypothesis is wrong, because implanted hamster testis tissue irrespectively of the grafting condition grows to a similar size revealing an intrinsic mechanism for testicular growth. In contrast, similar size of seminal vesicle as bio-indicator of androgen levels in all hosts revealed that the steroidogenic activity is independent from the mass of testicular tissue and that steroid levels are extrinsically regulated by the recipient's HPG axis. We propose that the model of testicular xenografting provides highly valuable options to explore testicular growth and endocrine regulation of the HPG axis.


Subject(s)
Models, Theoretical , Testicular Hormones/physiology , Testis/anatomy & histology , Testis/growth & development , Testis/transplantation , Animals , Animals, Newborn , Body Weight , Cells, Cultured , Cricetinae , Endocrine System/physiology , Male , Mice , Mice, Nude , Orchiectomy , Organ Size/physiology , Phodopus , Testicular Hormones/metabolism , Testis/physiology , Transplantation, Heterologous
20.
Biol Reprod ; 82(5): 888-95, 2010 May.
Article in English | MEDLINE | ID: mdl-20107205

ABSTRACT

Immature testicular tissue of a wide variety of mammalian species continues growth and maturation when ectopically grafted under the dorsal skin of adult nude mouse recipients. Tissues from most donor species fully mature, exhibiting complete spermatogenesis within months. The connection to the recipient's vascular system is mandatory for graft development, and failure of vascularization leads to necrosis in the grafted tissue. In the present study, we analyze to what extent 1) the xenografted immature donor tissue and 2) the recipient's cells and tissues contribute to the functional recovery of a "testicular xenograft." We address whether recipient cells migrate into the testicular parenchyma and whether the circulatory connection between the donor testicular tissue and the recipient is established by ingrowing host or outgrowing donor blood vessels. Although this issue has been repeatedly discussed in previous xenografting studies, so far it has not been possible to unequivocally distinguish between donor and recipient tissues and thus to identify the mechanisms by which the circulatory connection is established. To facilitate the distinction of donor and recipient tissues, herein we used immature green fluorescent protein-positive rat testes as donor tissues and adult nude mice as graft recipients. At the time of graft recovery, donor tissues could be easily identified by the GFP expression in these tissues, allowing us to distinguish donor- and recipient-derived blood vessels. We conclude that the circulatory connection between graft and host is established by a combination of outgrowing small capillaries from the donor tissue and formation of larger vessels by the host, which connect the graft to subcutaneous blood vessels.


Subject(s)
Graft Survival/physiology , Neovascularization, Physiologic/physiology , Spermatogenesis/physiology , Testis/growth & development , Animals , Cell Movement , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Nude , Rats , Rats, Transgenic , Recovery of Function , Species Specificity , Testis/blood supply , Testis/metabolism , Testis/transplantation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...