Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Biol Reprod ; 108(2): 316-323, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36383400

RESUMO

Zebra finch is a unique model for behavioral, neural, and genomic studies of vocal learning. Several transgenic zebra finches have been produced, although the germline transmission efficiencies are reportedly low. Recently, there have been attempts to produce germline chimeras using primordial germ cells (PGCs). However, this has been hampered by difficulties associated with the manipulation of the small eggs and the fact that the zebra finch is an altricial species that requires parental care after birth, unlike precocial chickens. Consequently, it is difficult to transplant PGCs into embryos and maintain the chimeras. Here, we developed a busulfan-mediated system for transplantation of PGCs into adult testes, to produce germline chimeras with an improved germline transmission capacity. We established microsomal glutathione-S-transferase II (MGSTII)-overexpressing PGCs that are resistant to busulfan, which induces germ cell-specific cytotoxicity, and transplanted them into testes rendered temporarily infertile by busulfan. The recipients were given a second dose of busulfan to deplete endogenous germ cells and enrich the transplanted cells, and donor cell-derived spermatogenesis was accomplished. This method requires fewer recipients due to higher survival rates, and there is no need to wait for maturation of the founders, which is required when transplanting PGCs into embryos. These results are expected to improve transgenic zebra finch production.


Assuntos
Galinhas , Aves Canoras , Masculino , Animais , Galinhas/genética , Testículo , Bussulfano/farmacologia , Células Germinativas/transplante , Animais Geneticamente Modificados
2.
Biol Reprod ; 103(6): 1289-1299, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32940693

RESUMO

In animals, spermatogonial transplantation in sterile adult males is widely developed; however, despite its utility, ovarian germ cell transplantation is not well developed. We previously showed that the interspecific hybrid offspring of sciaenid was a suitable model for germ cell transplantation studies as they have germ cell-less gonads. However, all these gonads have testis-like characteristics. Here, we tested whether triploidization in hybrid embryos could result in germ cell-less ovary development. Gonadal structure dimorphism and sex-specific gene expression patterns were examined in 6-month-old triploid hybrids (3nHybs). Thirty-one percent of 3nHybs had germ cell-less gonads with an ovarian cavity. cyp19a1a and foxl2, ovarian differentiation-related genes, were expressed in these gonads, whereas dmrt1 and vasa were not expressed, suggesting ovary-like germ cell-less gonad development. Some (26%) 3nHybs had testis-like germ cell-less gonads. Ovarian germ cells collected from homozygous green fluorescent protein (GFP) transgenic blue drum (BD) (Nibea mitsukurii) were transplanted into 6-month-old 3nHybs gonads via the urogenital papilla or oviduct. After 9 months, the recipients were crossed with wild type BD. Among the six 3nHyb recipients that survived, one female and one male produced fertile eggs and motile sperm carrying gfp-specific DNA sequences. Progeny tests revealed that all F1 offspring possessed gfp-specific DNA sequences, suggesting that these recipients produced only donor-derived eggs or sperm. Histological observation confirmed donor-derived gametogenesis in the 3nHyb recipients' gonads. Overall, triploidization reduces male-biased sex differentiation in germ cell-less gonads. We report, for the first time, donor-derived egg production in an animal via direct ovarian germ cell transplantation into a germ cell-less ovary.


Assuntos
Peixes/genética , Peixes/fisiologia , Células Germinativas/transplante , Gônadas/citologia , Triploidia , Animais , Animais Geneticamente Modificados , Aromatase/genética , Aromatase/metabolismo , Temperatura Baixa , RNA Helicases DEAD-box , Embrião não Mamífero , Feminino , Proteína Forkhead Box L2/genética , Proteína Forkhead Box L2/metabolismo , Regulação da Expressão Gênica , Masculino , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Reprod Fertil Dev ; 32(9): 807-821, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32527372

RESUMO

Fish populations continue to decline globally, signalling the need for new initiatives to conserve endangered species. Over the past two decades, with advances in our understanding of fish germ line biology, new exsitu management strategies for fish genetics and reproduction have focused on the use of germ line cells. The development of germ cell transplantation techniques for the purposes of propagating fish species, most commonly farmed species such as salmonids, has been gaining interest among conservation scientists as a means of regenerating endangered species. Previously, exsitu conservation methods in fish have been restricted to the cryopreservation of gametes or maintaining captive breeding colonies, both of which face significant challenges that have restricted their widespread implementation. However, advances in germ cell transplantation techniques have made its application in endangered species tangible. Using this approach, it is possible to preserve the genetics of fish species at any stage in their reproductive cycle regardless of sexual maturity or the limitations of brief annual spawning periods. Combining cryopreservation and germ cell transplantation will greatly expand our ability to preserve functional genetic samples from threatened species, to secure fish biodiversity and to produce new individuals to enhance or restore native populations.


Assuntos
Aquicultura , Criopreservação/veterinária , Espécies em Perigo de Extinção , Peixes/fisiologia , Células Germinativas/transplante , Reprodução , Técnicas de Reprodução Assistida/veterinária , Animais , Feminino , Peixes/genética , Masculino , Densidade Demográfica
4.
Gen Comp Endocrinol ; 295: 113525, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32502497

RESUMO

We aim to establish a small-bodied surrogate broodstock, such as mackerel, which produces functional bluefin tuna gametes by spermatogonial transplantation. When reproductively fertile fish are used as recipients, endogenous gametogenesis outcompetes donor-derived gametogenesis, and recipient fish predominantly produce their gametes. In this study, we assessed fertility of hybrid mackerel, Scomber australasicus × S. japonicus, and its suitability as a recipient for transplantation of bluefin tuna germ cells. Hybrid mackerel were produced by artificially inseminating S. australasicus eggs with S. japonicus spermatozoa. Cellular DNA content and PCR analyses revealed that F1 offspring were diploid carrying both paternal and maternal genomes. Surprisingly, histological observations found no germ cells in hybrid mackerel gonads at 120 days post-hatch (dph), although they were present in the gonad of 30- and 60-dph hybrid mackerel. The frequency of germ cell-less fish was 100% at 120-dph, 63.1% at 1-year-old, and 81.8% at 2-year-old. We also confirmed a lack of expression of germ cell marker (DEAD-box helicase 4, ddx4) in the germ cell-less gonads of hybrid mackerel. By contrast, expression of Sertoli cell marker (gonadal soma-derived growth factor, gsdf) and of Leydig cell marker (steroid 11-beta-hydroxlase, cyp11b1) were clearly detected in hybrid mackerel gonads. Together these results showed that most of the hybrid gonads were germ cell-less sterile, but still possessed supporting cells and steroidogenic cells, both of which are indispensable for nursing donor-derived germ cells. To determine whether hybrid gonads could attract and incorporate donor bluefin tuna germ cells, testicular cells labeled with PKH26 fluorescent dye were intraperitoneally transplanted. Fluorescence observation of hybrid recipients at 14 days post-transplantation revealed that donor cells had been incorporated into the recipient's gonads. This suggests that hybrid mackerel show significant promise for use as a recipient to produce bluefin tuna gametes.


Assuntos
Cruzamentos Genéticos , Células Germinativas/citologia , Células Germinativas/transplante , Gônadas/metabolismo , Hibridização Genética , Infertilidade/genética , Atum/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Reprodução , Testículo/citologia , Testículo/metabolismo , Testículo/transplante
5.
Biol Reprod ; 101(1): 200-207, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30980659

RESUMO

Successful derivation and cultivation of primordial germ cells (PGCs) opened the way to efficient transgenesis and genome editing in the chicken. Furthermore, implantation of male PGCs from non-chicken galliform species into the chicken embryos resulted in cross-species germline chimeras and viable offspring. We have recently improved the PGC technology by demonstrating that chicken male PGCs transplanted into the testes of adult cockerel recipients mature into functional sperms. However, the availability of this orthotopic transplantation for cross-species transfer remains to be explored. Here we tested the capacity of genetically distant male PGCs to mature in the microenvironment of adult testes. We derived PGCs from the Chinese black-bone Silkie and transplanted them into infertile White Leghorn cockerels. Within 15-18 weeks after transplantation, we observed restoration of spermatogenesis in recipient cockerels and production of healthy progeny derived from the transplanted PGCs. Our findings also indicate the possibility of cross-species orthotopic transplantation of PGCs. Thus, our results might contribute to the preservation of endangered avian species and maintaining the genetic variability of the domestic chicken.


Assuntos
Galinhas , Quimera/genética , Conservação dos Recursos Naturais , Células Germinativas/transplante , Espermatozoides/citologia , Animais , Cruzamento/métodos , Células Cultivadas , Embrião de Galinha , Galinhas/classificação , Galinhas/genética , Conservação dos Recursos Naturais/métodos , Cruzamentos Genéticos , Espécies em Perigo de Extinção , Preservação da Fertilidade/métodos , Preservação da Fertilidade/veterinária , Masculino , Espermatogênese/fisiologia , Espermatozoides/transplante , Testículo/citologia , Transplante Heterólogo/veterinária
6.
Biol Reprod ; 100(4): 1108-1117, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30544188

RESUMO

Many bitterling species are facing extinction because of habitat destruction. Since cryopreservation of fish eggs is still not available to date due to their large size and high yolk content, long-term and stable storage of bitterling genetic resources is currently not possible. We recently discovered that cryopreservation of early-stage germ cells is possible in several fish species and that functional gametes derived from the frozen materials can be produced through their transplantation to embryonic recipients. However, bitterlings have uniquely shaped eggs and their embryos are extremely fragile, making it difficult to perform germ cell transplantation. Therefore, as a first step, we conducted intra-species spermatogonial transplantation using recessive albino Chinese rosy bitterling as donors and wild-type Chinese rosy bitterling as recipients to develop a system to convert freezable early-stage germ cells into functional gametes, particularly eggs. Approximately 3000 testicular cells were transplanted into the peritoneal cavity of 4-day-old germ cell-less recipient embryos produced by dead end (dnd)-knockdown. At 6 months, ten male recipients and nine female recipients produced gametes. Mating studies with the opposite sex of recessive albino control fish revealed that six males and three females produced only albino offspring, suggesting that these recipients' endogenous germ cells were completely removed by dnd-knockdown and they produced only donor-derived gametes. Thus, we successfully established a germ cell transplantation system in an iconic endangered teleost, bitterling. The technology established in this study can be directly applied to produce functional gametes of endangered bitterlings using cryopreserved donor cells.


Assuntos
Cyprinidae/fisiologia , Espermatogônias/transplante , Doadores de Tecidos , Animais , Animais Geneticamente Modificados , Transplante de Células , Conservação dos Recursos Naturais/métodos , Criopreservação/métodos , Criopreservação/veterinária , Cyprinidae/genética , Cyprinidae/crescimento & desenvolvimento , Espécies em Perigo de Extinção , Feminino , Fertilização in vitro/veterinária , Técnicas de Silenciamento de Genes , Células Germinativas/transplante , Masculino , Proteínas de Ligação a RNA/genética , Preservação do Sêmen/veterinária , Transplante Homólogo/veterinária
7.
Mol Reprod Dev ; 86(12): 1810-1821, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31544311

RESUMO

In the fish germ cell transplantation system, only type A spermatogonia (ASGs) and oogonia are known to be incorporated into the recipient genital ridges, where they undergo gametogenesis. Therefore, high colonization efficiency can be achieved by enriching undifferentiated germ cells out of whole testicular cells. In this study, we used magnetic-activated cell sorting (MACS) for enriching undifferentiated germ cells of rainbow trout using a monoclonal antibody that recognizes a specific antigen located on the germ cell membrane. We screened the antibodies to be used for MACS by performing immunohistochemistry on rainbow trout gonads. Two antibodies, nos. 172 and 189, showed strong signals for ASGs and oogonia. Next, we performed MACS with antibody no. 172 using gonadal cells isolated from vasa-gfp rainbow trout showing GFP in undifferentiated germ cells. We found that GFP-positive cells are highly enriched in antibody no. 172-positive fractions. Finally, to examine the transplantability of MACS-enriched cells, we intraperitoneally transplanted sorted or unsorted cells into recipient larvae. We observed that transplantability of sorted cells, particularly ovarian cells, were significantly higher than that of unsorted cells. Therefore, MACS with antibody no. 172 could enrich ASGs and oogonia and become a powerful tool to improve transplantation efficiency in salmonids.


Assuntos
Animais Geneticamente Modificados , Anticorpos Monoclonais/química , Células Germinativas , Separação Imunomagnética , Oncorhynchus mykiss , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Feminino , Células Germinativas/citologia , Células Germinativas/transplante , Masculino , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo
8.
Reprod Fertil Dev ; 31(3): 538-546, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30309435

RESUMO

The object of this study was to investigate if testis germ cell transplantation (TGCT) into a heterologous recipient would result in donor-origin spermatogenesis in the dromedary camel. First, we investigated a workable protocol for TGCT in camels, including donor cell isolation, enrichment by density gradient centrifugation (Percoll and Bovicoll), rete testis injection and microsatellite detection of donor and recipient genotypes. Second, the effects of three doses of Dolichos biflorus agglutinin (DBA), a glycoprotein that specifically binds to gonocytes or Type A spermatogonia, on testis germ cell depletion were investigated by direct injection into the rete testis of a male camel. Seven recipients were prepared with DBA treatment, two males were castrated at 4 weeks for depletion assessment and the remaining five received donor cells 4-6 weeks after treatment. On average, ~17 million cells were isolated per gram of testis tissue, with 19.5±1.9% DBA-positive (DBA+) cells. Percoll centrifugation yielded a 1.5-fold increase in DBA+ cells while Bovicoll centrifugation produced a 2.5-fold increase from the input cells of 18.6±2.1% DBA+ cells. Semen was collected from the recipients 13-20 weeks after transfer and the presence of donor DNA in the samples was determined using microsatellite markers. In two of the five recipients, all semen samples were shown to be positive for donor-derived cells. These results demonstrate for the first time that: (1) heterologous testicular germ cell transplantation in camels is feasible and the recipients are able to produce spermatozoa of donor origin and (2) DBA can be used effectively to deplete endogenous stem cells.


Assuntos
Transplante de Células/veterinária , Células Germinativas/transplante , Espermatogênese/fisiologia , Testículo/efeitos dos fármacos , Transplante Heterólogo/veterinária , Animais , Camelus , Transplante de Células/métodos , Genótipo , Masculino , Lectinas de Plantas/farmacologia , Espermatogônias/citologia , Espermatogônias/efeitos dos fármacos , Testículo/citologia , Transplante Heterólogo/métodos
9.
J Assist Reprod Genet ; 36(6): 1041-1048, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31127476

RESUMO

PURPOSE: To heighten awareness of the potential legal and financial burdens faced by those providing cryopreservation storage services of embryos and gametes in light of recent lawsuits involving inadvertent thawing of specimens. METHODS: Case law review of US legal databases and courthouse dockets with a focus on lawsuits against reproductive endocrinologists and cryostorage facilities offering cryopreservation. Emphasis was placed on court decisions, awarded damages, and legal and media coverage related to cryostorage failure events. RESULTS: Lawsuits pertaining to two notable ongoing cases of cryostorage failure that occurred at fertility clinics in the US in 2018 were reviewed. Media coverage of these events and plaintiff and defense attorney strategies were evaluated. Legal documents from previous, similar cryostorage failures were also reviewed. Common claims in cryostorage system failures include breach of contract and negligent handling of property. Facilities offering cryostorage services are vulnerable to significant burden, legally and financially, if they are to experience a storage system failure. CONCLUSION: Providing cryostorage services is not without significant financial risk. Inadvertent thawing of specimens can lead to high damage awards against cryostorage facilities and those individuals linked to a cryostorage failure event. Because monetary damages can surpass insurance policy limits, those providing cryostorage services should be aware of plaintiff attorney strategies, common legal defenses, and basic asset protection principles to safeguard themselves if ever faced with these situations. Facilities should also carry out regular maintenance and safety checks on equipment and alarm structures to deter such events.


Assuntos
Criopreservação , Imperícia/legislação & jurisprudência , Técnicas de Reprodução Assistida/legislação & jurisprudência , Manejo de Espécimes , Feminino , Células Germinativas/transplante , Humanos , Masculino , Técnicas de Reprodução Assistida/economia
10.
J Cell Physiol ; 233(11): 8441-8449, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29870061

RESUMO

Stem cells (SCs) are classes of undifferentiated biological cells existing only at the embryonic, fetal, and adult stages that can divide to produce specialized cell types during fetal development and remain in our bodies throughout life. The progression of regenerative and reproductive medicine owes the advancement of respective in vitro and in vivo biological science on the stem cell nature under appropriate conditions. The SCs are promising therapeutic tools to treat currently of infertility because of wide sources and high potency to differentiate. Nevertheless, no effective remedies are available to deal with severe infertility due to congenital or gonadotoxic stem cell deficiency in prepubertal childhood. Some recent solutions have been developed to address the severe fertility problems, including in vitro formation of germ cells from stem cells, induction of pluripotency from somatic cells, and production of patient-specific pluripotent stem cells. There is a possibility of fertility restoration using the in vitro formation of germ cells from somatic cells. Accordingly, the present review aimed at studying the literature published on the medical application of stem cells in reproductive concerns.


Assuntos
Azoospermia/terapia , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Infertilidade Masculina/terapia , Adolescente , Adulto , Azoospermia/patologia , Diferenciação Celular/genética , Células-Tronco Embrionárias/transplante , Células Germinativas/citologia , Células Germinativas/patologia , Células Germinativas/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/transplante , Infertilidade Masculina/patologia , Masculino , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia , Neoplasias Testiculares/terapia , Adulto Jovem
11.
Hum Reprod ; 33(2): 188-195, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29315416

RESUMO

Recently, significant progress has been made in ART for the treatment of male infertility. However, current ART has failed to help infertile patients with non-obstructive azoospermia, unless donor sperm is used. In fact, most couples wish to have their own genetically related child. Human induced pluripotent stem cells (hiPSCs) can be generated from patients' somatic cells and in vitro derivation of functional germ cells from patient-specific iPSCs may provide new therapeutic strategies for infertile couples. The overall developmental dynamics of human primordial germ cells are similar to that in mice, but accumulating evidence suggests that there are crucial differences between human and mouse PGC specification. Unlike mouse iPSCs (miPSCs) in naive state, hiPSCs exhibit a primed pluripotency which possess less potential for the germ cell fate. Based on research in mice, male germ cells at different stages have been derived from hiPSCs with different protocols, including spontaneous differentiation, overexpression of germ cell regulators, addition of cytokines, co-culture with gonadal cells in vitro and xeno-transplantation. The aim of this review is to summarize the current advances in derivation of male germ cells from hiPSCs and raise the perspectives of hiPSCs in medical application for male infertility, as well as in basic research for male germ cell development.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Infertilidade Masculina/terapia , Animais , Diferenciação Celular , Edição de Genes , Células Germinativas/citologia , Células Germinativas/transplante , Humanos , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Masculino , Camundongos , Modelos Biológicos , Técnicas de Reprodução Assistida/tendências , Especificidade da Espécie , Espermatozoides/citologia
12.
Indian J Med Res ; 148(Suppl): S38-S49, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30964080

RESUMO

A large proportion of patients who survive cancer are rendered infertile as an unwanted side effect of oncotherapy. Currently accepted approaches for fertility preservation involve banking eggs/sperm/embryos or ovarian/testicular tissue before oncotherapy for future use. Such approaches are invasive, expensive, technically challenging and depend on assisted reproductive technologies (ART). Establishing a gonadal tissue bank (for cancer patients) is also fraught with ethical, legal and safety issues. Most importantly, patients who find it difficult to meet expenses towards cancer treatment will find it difficult to meet expenses towards gonadal tissue banking and ART to achieve parenthood later on. In this review an alternative strategy to regenerate non-functional gonads in cancer survivors by targeting endogenous stem cells that survive oncotherapy is discussed. A novel population of pluripotent stem cells termed very small embryonic-like stem cells (VSELs), developmentally equivalent to late migratory primordial germ cells, exists in adult gonads and survives oncotherapy due to their quiescent nature. However, the stem-cell niche gets compromised by oncotherapy. Transplanting niche cells (Sertoli or mesenchymal cells) can regenerate the non-functional gonads. This approach is safe, has resulted in the birth of fertile offspring in mice and could restore gonadal function early in life to support proper growth and later serve as a source of gametes. This newly emerging understanding on stem cells biology can obviate the need to bank gonadal tissue and fertility may also be restored in existing cancer survivors who were earlier deprived of gonadal tissue banking before oncotherapy.


Assuntos
Células-Tronco Embrionárias/transplante , Células Germinativas/crescimento & desenvolvimento , Infertilidade/genética , Ovário/transplante , Adulto , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Feminino , Preservação da Fertilidade/métodos , Células Germinativas/transplante , Gônadas/crescimento & desenvolvimento , Gônadas/transplante , Humanos , Infertilidade/patologia , Infertilidade/terapia , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Ovário/crescimento & desenvolvimento , Regeneração/genética , Espermatogênese/genética , Nicho de Células-Tronco , Testículo/crescimento & desenvolvimento
13.
Fish Physiol Biochem ; 44(6): 1469-1485, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29707740

RESUMO

This review discusses the new biotechnological tools that are arising and promising for conservation and enhancement of fish production, mainly regarding the endangered and the most economically important species. Two main techniques, in particular, are available to avoid extinction of endangered fish species and to improve the production of commercial species. Germ cell transplantation technology includes a number of approaches that have been studied, such as the transplantation of embryo-to-embryo blastomere, embryo-to-embryo differentiated PGC, larvae to larvae and embryo differentiated PGC, transplantation of spermatogonia from adult to larvae or between adults, and oogonia transplantation. However, the success of germ cell transplantation relies on the prior sterilization of fish, which can be performed at different stages of fish species development by means of several protocols that have been tested in order to achieve the best approach to produce a sterile fish. Among them, fish hybridization and triploidization, germline gene knockdown, hyperthermia, and chemical treatment deserve attention based on important results achieved thus far. This review currently used technologies and knowledge about surrogate technology and fish sterilization, discussing the stronger and the weaker points of each approach.


Assuntos
Peixes/fisiologia , Células Germinativas/citologia , Células Germinativas/transplante , Técnicas de Reprodução Assistida/veterinária , Animais , Biotecnologia , Conservação dos Recursos Naturais , Reprodução
14.
Fish Physiol Biochem ; 44(6): 1487-1498, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29756177

RESUMO

Interspecific transplantation of germ cells from the brown trout Salmo trutta m. fario and the European grayling Thymallus thymallus into rainbow trout Oncorhynchus mykiss recipients was carried out in order to improve current practices in conservation of genetic resources of endangered salmonid species in the Balkan Peninsula. Current conservation methods mainly include in situ efforts such as the maintenance of purebred individuals in isolated streams and restocking with purebred fingerlings; however, additional ex situ strategies such as surrogate production are needed. Steps required for transplantation such as isolation of high number of viable germ cells and fluorescent labeling of germ cells which are to be transplanted have been optimized. Isolated and labeled brown trout and grayling germ cells were intraperitoneally transplanted into 3 to 5 days post hatch rainbow trout larvae. Survival of the injected larvae was comparable to the controls. Sixty days after transplantation, fluorescently labeled donor cells were detected within the recipient gonads indicating successful incorporation of germ cells (brown trout spermatogonia and oogonia-27%; grayling spermatogonia-28%; grayling oogonia-23%). PCR amplification of donor mtDNA CR fragments within the recipient gonads additionally corroborated the success of incorporation. Overall, the transplantation method demonstrated in this study presents the first step and a possible onset of the application of the germ cell transplantation technology in conservation and revitalization of genetic resources of endangered and endemic species or populations of salmonid fish and thus give rise to new or improved management strategies for such species.


Assuntos
Transplante de Células/veterinária , Embrião não Mamífero/citologia , Células Germinativas/citologia , Células Germinativas/transplante , Oncorhynchus mykiss/embriologia , Salmonidae/embriologia , Transplante Heterólogo/veterinária , Animais , Península Balcânica , Diferenciação Celular , Transplante de Células/métodos , Conservação dos Recursos Naturais , Embrião não Mamífero/fisiologia , Desenvolvimento Embrionário , Oncorhynchus mykiss/genética , Salmonidae/classificação , Salmonidae/genética
15.
Biol Reprod ; 96(3): 686-693, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28339605

RESUMO

Cloning (somatic cell nuclear transfer) in avian species has proven unachievable due to the physical structure of the avian oocyte. Here, the sexual differentiation of primordial germ cells with genetic sex ZZ (ZZ PGCs) was investigated in female germline chimeric chicken hosts with the aim to produce uniparental offspring. ZZ PGCs were expanded in culture and transplanted into the same and opposite sex chicken embryos which were partially sterilized using irradiation. All tested chimeric roosters (ZZ/ZZ) showed germline transmission with transmission rates of 3.2%-91.4%. Unexpectedly, functional oogenesis of chicken ZZ PGCs was found in three chimeric hens, resulting in a transmission rate of 2.3%-27.8%. Matings were conducted between the germline chimeras (ZZ/ZZ and ZZ/ZW) which derived from the same ZZ PGCs line. Paternal uniparental chicken offspring were obtained with a transmission rate up to 28.4% and as expected, all uniparental offspring were phenotypic male (ZZ). Genotype analysis of uniparental offsprings was performed using 13 microsatellite markers. The genotype profile showed that uniparental offspring were 100% genetically identical to the donor ZZ PGC line, shared 69.2%-88.5% identity with the donor bird. Homozygosity of the tested birds varied from 61.5% to 84.6%, which was higher than the donor bird (38.5%). These results demonstrate that male avian ZZ PGCs can differentiate into functional ova in an ovary, and uniparental avian clones are possible. This technology suggests novel approaches for generating genetically similar flocks of birds and for the conservation of avian genetic resources.


Assuntos
Células Germinativas/transplante , Oogênese , Quimera por Radiação , Animais , Embrião de Galinha , Galinhas , Feminino , Células Germinativas/fisiologia , Masculino
16.
Mol Reprod Dev ; 84(10): 1100-1111, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28731265

RESUMO

Germ cell transplantation is a promising technology for the propagation of endangered or valuable fishes. In this technique, sterile male and female recipient fish are injected with donor germ cells so they can produce viable gametes derived from the donor cells. The dead end (dnd) gene is involved in the migration of primordial germ cells; therefore, dnd-knockout zebrafish are expected to be germ-cell-free, making them suitable recipients for germ cell transplantation. dnd mutants were produced by microinjecting 2 nl of 10 ng/µl cRNAs encoding zinc finger nucleases against dnd into the blastodisc of zebrafish embryos before the cell- cleavage stage. One of the resulting founder males was mated with a wild-type female, and produced heterozygous mutants in the F1 generation. Mating of these F1 mutants produced an F2 generation with approximately 25% of the clutch being homozygous mutant (dnd-knockout) male, and lacking germ cells (as confirmed by expression analyses of vasa). The resulting dnd-knockout zebrafish males were tested for suitability as germ cell transplantation recipients by intraperitoneal transplantation of testicular cells prepared from vasa-gfp zebrafish. GFP-positive germ cells incorporated into the germ-cell-free gonads of the dnd-knockout recipients matured into functional sperm. Progeny tests revealed that the sperm from these dnd-knockout recipients were derived entirely from donor cells. Thus, we demonstrated that homozygous dnd mutants became germ-cell-free males that are able to nurse donor-derived germ cells.


Assuntos
Transplante de Células/métodos , Células Germinativas/transplante , Modelos Animais , Proteínas de Ligação a RNA/genética , Transplantados , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Células-Tronco Germinativas Adultas/citologia , Células-Tronco Germinativas Adultas/transplante , Animais , Animais Geneticamente Modificados , Embrião não Mamífero , Técnicas de Inativação de Genes/métodos , Masculino , Peixe-Zebra/embriologia , Peixe-Zebra/genética
17.
Biochem Biophys Res Commun ; 478(3): 1478-83, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27581197

RESUMO

Cryopreservation of fish sperm offers the practical applications in the selective breeding and biodiversity conservation. However, because of the lack of cryopreservation methods for fish eggs and embryos, maternally inherited cytoplasmic compartments cannot be successfully preserved. We previously developed an alternative method to derive functional eggs and sperm from cryopreserved whole testis by transplanting testicular cells into female and male recipients. However, if target fish had ovaries, the previous method employing male-derived germ cells would be ineffective. Here, we aimed to generate functional gametes from cryopreserved whole ovaries by transplanting ovarian germ cells into peritoneal cavity of sterile hatchlings. Cryopreservation conditions for rainbow trout ovaries (1.0 M DMSO, 0.1 M trehalose, and 10% egg yolk) were optimized by testing several different cryoprotective agents. Ovarian germ cells from thawed ovaries were intraperitoneally transplanted into allogeneic triploid hatchlings. Transplanted germ cells migrated toward and were incorporated into recipient gonads, where they underwent gametogenesis. Transplantation efficiency of ovarian germ cells remained stable after cryopreservation period up to 1185 days. Although all triploid recipients that did not undergo transplantation were functionally sterile, 5 of 25 female recipients and 7 of 25 male recipients reached sexual maturity at 2.5 years post-transplantation. Inseminating the resultant eggs and sperm generated viable offspring displaying the donor characteristics of orange body color, green fluorescence, and chromosome numbers. This method is thus a breakthrough tool for the conservation of endangered fish species that are crucial to cryopreserve the genetic resources of female fish.


Assuntos
Criopreservação , Células Germinativas/transplante , Oncorhynchus mykiss/embriologia , Ovário/citologia , Ovário/fisiologia , Animais , Sobrevivência Celular , Feminino , Congelamento , Injeções Intraperitoneais , Masculino , Triploidia
18.
Reprod Fertil Dev ; 28(5): 628-39, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25482458

RESUMO

When derived from chicken embryos, avian primordial germ cells (PGCs) have been reported to keep their germline-specific properties and proliferative potential even after long-term culture and genetic modifications. Few teams to date have reported such long-term expansion and engineering without differentiation of primary avian PGCs' cultures. We have developed original and robust methods that allow more than 1 year culture, expansion and cryobanking of primary cultures of PGCs without obvious effects on their biological properties, including their ability to colonise the genital ridges. Overall, 38% of embryonic samples gave rise to PGCs lines derived from three commercial layers and two Belgian endangered breeds. The lines kept their proliferative potential and their characteristic PGCs phenotype after 20 months in culture, whether or not interrupted by a cryopreservation step. All the resulting lines appeared devoid of female cells, although initially pooled from male and female embryos. Labelled PGCs from 12 long-term cultured lines colonised the genital ridges of recipient embryos. Thus, this procedure allows derivation, long-term expansion and cryobanking of primary cultures of PGCs without obvious changes to their original characteristics, providing an alternative access to applications in avian biotechnology and preservation of genetic resources.


Assuntos
Movimento Celular , Proliferação de Células , Galinhas/fisiologia , Criopreservação/veterinária , Espécies em Perigo de Extinção , Células Germinativas/fisiologia , Gônadas/embriologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Células Cultivadas , Embrião de Galinha , Galinhas/genética , Feminino , Células Germinativas/metabolismo , Células Germinativas/transplante , Masculino , Fenótipo , Análise para Determinação do Sexo/veterinária , Fatores de Tempo
19.
J Reprod Dev ; 62(5): 431-437, 2016 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-27210834

RESUMO

The majority of poultry genetic resources are maintained in situ in living populations. However, in situ conservation of poultry genetic resources always carries the risk of loss owing to pathogen outbreaks, genetic problems, breeding cessation, or natural disasters. Cryobanking of germplasm in birds has been limited to the use of semen, preventing conservation of the W chromosome and mitochondrial DNA. A further challenge is posed by the structure of avian eggs, which restricts the cryopreservation of ova and fertilized embryos, a technique widely used for mammalian species. By using a unique biological property and accessibility of avian primordial germ cells (PGCs), precursor cells for gametes, which temporally circulate in the vasculature during early development, an avian PGC transplantation technique has been established. To date, several techniques for PGC manipulation including purification, cryopreservation, depletion, and long-term culture have been developed in chickens. PGC transplantation combined with recent advanced PGC manipulation techniques have enabled ex situ conservation of poultry genetic resources in their complete form. Here, the updated technologies for avian PGC manipulation are introduced, and then the concept of a poultry PGC-bank is proposed by considering the biological properties of avian PGCs.


Assuntos
Galinhas/genética , Criopreservação/veterinária , Animais , Cruzamento , DNA Mitocondrial/genética , Embrião não Mamífero , Feminino , Células Germinativas/citologia , Células Germinativas/transplante , Óvulo/citologia , Aves Domésticas/genética , Cromossomos Sexuais/genética , Temperatura
20.
Reproduction ; 149(2): 179-87, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25550524

RESUMO

In most animals, primordial germ cells (PGCs) originate from an extragonadal region and migrate across the embryo to the gonads, where they differentiate and function. During their migration, PGCs move passively by morphogenetic movement of the embryo or move actively through signaling molecules. To uncover the underlying mechanism of first-phase PGC migration toward the germinal crescent in chickens, we investigated the spatial and temporal action of PGCs during primitive streak formation. Exogenously transplanted PGCs migrated toward the anterior region of the embryo and the embryonic gonads when they were transplanted into the subgerminal cavity, but not into the posterior marginal zone, in Eyal-Giladi and Kochav stage X embryos. These results indicate that for passive migration toward the anterior region the initial location of PGCs should be the central region. Notably, although PGCs and DF-1 cells migrated passively toward the anterior region, only PGCs migrated to the germinal crescent, where endogenous PGCs mainly reside, by active movement. In a live-imaging experiment with green fluorescence protein-expressing transgenic embryos, exogenous PGCs demonstrated markedly faster migration when they reached the anterior one-third of the embryo, while somatic cells showed epiblast movement with constant speed. Also, migrating PGCs exhibited successive contraction and expansion indicating their active migration. Our results suggest that chicken PGCs use sequential passive and active forces to migrate toward the germinal crescent.


Assuntos
Movimento Celular , Embrião de Galinha/citologia , Células Germinativas/fisiologia , Animais , Embrião de Galinha/crescimento & desenvolvimento , Células Germinativas/transplante , Gônadas/citologia , Gônadas/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA