Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
BMC Biol ; 20(1): 109, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35550124

RESUMO

BACKGROUND: Ovarian follicles, which are the basic units of female reproduction, are composed of oocytes and surrounding somatic (pre) granulosa cells (GCs). A recent study revealed that signaling in somatic preGCs controlled the activation (initial recruitment) of follicles in the adult ovaries, but it is also known that there are two waves of follicle with age-related heterogeneity in their developmental dynamics in mammals. Although this heterogeneity was proposed to be crucial for female reproduction, our understanding of how it arises and its significance is still elusive. RESULTS: In the current study, by deleting the key secreted factor KIT ligand from preGCs and analyzing the follicle cell developmental dynamics, we revealed distinct patterns of activation and growth associated with the two waves of follicles in mouse ovary. Our results confirmed that activation of adult wave follicles is initiated by somatic preGCs and dependent on the KIT ligand. By contrast, activation of first wave follicles, which are awakened from germ cells before follicle formation, can occur in the absence of preGC-secreted KIT ligand in postnatal ovaries and appears to be oocyte-initiated. We also found that the asynchronous activity of phosphatidylinositol 3 kinases (PI3K) signaling and meiotic process in embryonic germ cells lead to the follicle heterogeneity in postnatal ovaries. In addition, we supplied evidence that the time sequence of embryonic germ cell development and its related first wave follicle growth are correlated to the time of puberty onset in females. CONCLUSION: Taken together, our study provides evidence that asynchronous development of embryonic oocytes leads to the heterogeneity of postnatal ovarian follicle activation and development, and affects the timing of onset of puberty in females.


Assuntos
Células Germinativas Embrionárias , Fosfatidilinositol 3-Quinases , Animais , Feminino , Mamíferos , Camundongos , Oócitos/fisiologia , Oogênese , Folículo Ovariano , Maturidade Sexual , Fator de Células-Tronco
2.
Nat Commun ; 13(1): 711, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35132083

RESUMO

In many species including humans, aging reduces female fertility. Intriguingly, some animals preserve fertility longer under specific environmental conditions. For example, at low temperature and short day-length, Drosophila melanogaster enters a state called adult reproductive diapause. As in other stressful conditions, ovarian development arrests at the yolk uptake checkpoint; however, mechanisms underlying fertility preservation and post-diapause recovery are largely unknown. Here, we report that diapause causes more complete arrest than other stresses yet preserves greater recovery potential. During dormancy, germline stem cells (GSCs) incur DNA damage, activate p53 and Chk2, and divide less. Despite reduced niche signaling, germline precursor cells do not differentiate. GSCs adopt an atypical, suspended state connected to their daughters. Post-diapause recovery of niche signaling and resumption of division contribute to restoring GSCs. Mimicking one feature of quiescence, reduced juvenile hormone production, enhanced GSC longevity in non-diapausing flies. Thus, diapause mechanisms provide approaches to GSC longevity enhancement.


Assuntos
Diapausa de Inseto/fisiologia , Células Germinativas Embrionárias/fisiologia , Animais , Diferenciação Celular , Senescência Celular , Quinase do Ponto de Checagem 2/metabolismo , Dano ao DNA , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Células Germinativas Embrionárias/citologia , Feminino , Fertilidade , Hormônios Juvenis/metabolismo , Ovário/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Nicho de Células-Tronco , Proteína Supressora de Tumor p53/metabolismo
4.
Gene ; 794: 145760, 2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34116118

RESUMO

BMP4 is the critical gene of primordial germ cell formation in mammal, however, the mechanism of PGCs formation in chicken still unknown. In this research, we compared the evolution relationship of different species. Although the protein sequence is highly conservative between mouse, human and chicken, promotors vary among avian and mammal species. Therefore, it is easily to predict that there would be different regulation mechanism of Bmp4 expression in chicken. Here, we elucidate the function of chicken Bmp4 during PGCs formation. In vivo, Bmp4 can promote PGCs development and migration, and increase the expression of key genes (Cvh, c-kit, cxcr4, etc.). Whereas, the expression of these genes will decrease after knocking out Bmp4. After over-expression and knockout Bmp4 in vitro, we found that overexpression of Bmp4 could promote the formation of embryoid bodies (EB) and up-regulate the key genes of PGCs formation and migration, while knockout Bmp4 could inhibit the formation of embryoid bodies and decrease the expression of related genes. Flow and indirect immunofluorescence also indicated the same result. These all results proved that chicken Bmp4 could also promote the formation of PGCs. Furthermore, dual-luciferase activity detection showed that the promotor activity of Bmp4 was positively regulated by transcription factor Zeb1. Overexpression of Zeb1 can also increase the mRNA and protein expression of Bmp4. At the same time, DNA methylation inhibited Bmp4 transcription and histone methylation was able to promote its transcription. In conclusion, this study established that chicken Bmp4 can promote the formation of chicken PGCs. This gene is regulated by DNA, histone methylation and transcription factor Zeb1. These results lay a theoretical foundation for exploring the function and molecular mechanism of Bmp4 in the process of PGCs formation.


Assuntos
Proteína Morfogenética Óssea 4/genética , Células Germinativas Embrionárias/citologia , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Animais , Proteína Morfogenética Óssea 4/metabolismo , Movimento Celular , Células Cultivadas , Galinhas , Metilação de DNA , Corpos Embrioides/metabolismo , Células Germinativas Embrionárias/metabolismo , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes
5.
Development ; 148(9)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33912935

RESUMO

In response to signals from the embryonic testis, the germ cell intrinsic factor NANOS2 coordinates a transcriptional program necessary for the differentiation of pluripotent-like primordial germ cells toward a unipotent spermatogonial stem cell fate. Emerging evidence indicates that genetic risk factors contribute to testicular germ cell tumor initiation by disrupting sex-specific differentiation. Here, using the 129.MOLF-Chr19 mouse model of testicular teratomas and a NANOS2 reporter allele, we report that the developmental phenotypes required for tumorigenesis, including failure to enter mitotic arrest, retention of pluripotency and delayed sex-specific differentiation, were exclusive to a subpopulation of germ cells failing to express NANOS2. Single-cell RNA sequencing revealed that embryonic day 15.5 NANOS2-deficient germ cells and embryonal carcinoma cells developed a transcriptional profile enriched for MYC signaling, NODAL signaling and primed pluripotency. Moreover, lineage-tracing experiments demonstrated that embryonal carcinoma cells arose exclusively from germ cells failing to express NANOS2. Our results indicate that NANOS2 is the nexus through which several genetic risk factors influence tumor susceptibility. We propose that, in the absence of sex specification, signals native to the developing testis drive germ cell transformation.


Assuntos
Diferenciação Celular , Neoplasias Embrionárias de Células Germinativas , Diferenciação Sexual , Neoplasias Testiculares , Animais , Diferenciação Celular/genética , Proliferação de Células , Células-Tronco de Carcinoma Embrionário/metabolismo , Células Germinativas Embrionárias , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos , Proteínas de Ligação a RNA , Transdução de Sinais , Espermatogônias/metabolismo , Teratoma
6.
Nat Commun ; 12(1): 2285, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863891

RESUMO

During Drosophila embryonic development, cell death eliminates 30% of the primordial germ cells (PGCs). Inhibiting apoptosis does not prevent PGC death, suggesting a divergence from the conventional apoptotic program. Here, we demonstrate that PGCs normally activate an intrinsic alternative cell death (ACD) pathway mediated by DNase II release from lysosomes, leading to nuclear translocation and subsequent DNA double-strand breaks (DSBs). DSBs activate the DNA damage-sensing enzyme, Poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) and the ATR/Chk1 branch of the DNA damage response. PARP-1 and DNase II engage in a positive feedback amplification loop mediated by the release of PAR polymers from the nucleus and the nuclear accumulation of DNase II in an AIF- and CypA-dependent manner, ultimately resulting in PGC death. Given the anatomical and molecular similarities with an ACD pathway called parthanatos, these findings reveal a parthanatos-like cell death pathway active during Drosophila development.


Assuntos
Drosophila/efeitos dos fármacos , Desenvolvimento Embrionário/fisiologia , Células Germinativas Embrionárias/fisiologia , Endodesoxirribonucleases/metabolismo , Parthanatos/fisiologia , Animais , Animais Geneticamente Modificados , Núcleo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Drosophila/citologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrião não Mamífero/citologia , Células Germinativas Embrionárias/citologia , Endodesoxirribonucleases/genética , Retroalimentação Fisiológica , Feminino , Lisossomos/metabolismo , Masculino , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo
7.
Cell Rep ; 34(9): 108799, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33657369

RESUMO

The Tre1 G-protein coupled receptor (GPCR) was discovered to be required for Drosophila germ cell (GC) coalescence almost two decades ago, yet the molecular events both upstream and downstream of Tre1 activation remain poorly understood. To gain insight into these events, we describe a bona fide null allele and both untagged and tagged versions of Tre1. We find that the primary defect with complete Tre1 loss is the failure of GCs to properly navigate, with GC mis-migration occurring from early stages. We find that Tre1 localizes with F-actin at the migration front, along with PI(4,5)P2; dPIP5K, an enzyme that generates PI(4,5)P2; and dWIP, a protein that binds activated Wiskott-Aldrich syndrome protein (WASP), which stimulates F-actin polymerization. We show that Tre1 is required for polarized accumulation of F-actin, PI(4,5)P2, and dPIP5K. Smoothened also localizes with F-actin at the migration front, and Hh, through Smo, increases levels of Tre1 at the plasma membrane and Tre1's association with dPIP5K.


Assuntos
Citoesqueleto de Actina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células Germinativas Embrionárias/metabolismo , Proteínas Hedgehog/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Citoesqueleto de Actina/genética , Animais , Animais Geneticamente Modificados , Movimento Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Proteínas Hedgehog/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Fatores de Tempo , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
8.
Clin Epigenetics ; 13(1): 28, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33541399

RESUMO

BACKGROUND: Patients suffering from the BCR-ABL1-negative myeloproliferative disease prefibrotic primary myelofibrosis (pre-PMF) have a certain risk for progression to myelofibrosis. Accurate risk estimation for this fibrotic progression is of prognostic importance and clinically relevant. Commonly applied risk scores are based on clinical, cytogenetic, and genetic data but do not include epigenetic modifications. Therefore, we evaluated the assessment of genome-wide DNA methylation patterns for their ability to predict fibrotic progression in PMF patients. RESULTS: For this purpose, the DNA methylation profile was analyzed genome-wide in a training set of 22 bone marrow trephines from patients with either fibrotic progression (n = 12) or stable disease over several years (n = 10) using the 850 k EPIC array from Illumina. The DNA methylation classifier constructed from this data set was validated in an independently measured test set of additional 11 bone marrow trephines (7 with stable disease, 4 with fibrotic progress). Hierarchical clustering of methylation ß-values and linear discriminant classification yielded very good discrimination between both patient groups. By gene ontology analysis, the most differentially methylated CpG sites are primarily associated with genes involved in cell-cell and cell-matrix interactions. CONCLUSIONS: In conclusion, we could show that genome-wide DNA methylation profiling of bone marrow trephines is feasible under routine diagnostic conditions and, more importantly, is able to predict fibrotic progression in pre-fibrotic primary myelofibrosis with high accuracy.


Assuntos
Impressões Digitais de DNA/métodos , Fibrose/genética , Estudo de Associação Genômica Ampla/métodos , Mielofibrose Primária/genética , Experimentação Animal , Medula Óssea/metabolismo , Competição entre as Células/genética , Técnicas de Reprogramação Celular/métodos , Ilhas de CpG/genética , Metilação de DNA , Progressão da Doença , Células Germinativas Embrionárias/metabolismo , Epigenômica/métodos , Feminino , Fibrose/patologia , Proteínas de Fusão bcr-abl/genética , Ontologia Genética , Humanos , Masculino , Valor Preditivo dos Testes , Mielofibrose Primária/patologia , Prognóstico , Fatores de Risco
9.
Methods Mol Biol ; 2214: 59-73, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32944903

RESUMO

Primordial germ cells (PGCs) are the embryonic precursors of the gametes. Despite decades of research, in vitro culture of PGCs remains a major challenge and has previously relied on undefined components such as serum and feeders. Notably, PGCs cultured for extended periods do not maintain their lineage identity but instead undergo conversion to form pluripotent stem cell lines called embryonic germ (EG) cells in response to LIF/STAT3 signaling. Here we report both established and new methodologies to derive EG cells, in a range of different conditions. We show that basic fibroblast growth factor is not required for EG cell conversion. We detail the steps taken in our laboratory to systematically remove complex components and establish a fully defined protocol that allows efficient conversion of isolated PGCs to pluripotent EG cells. In addition, we demonstrate that PGCs can adhere and proliferate in culture without the support of feeder cells or serum. This may well suggest novel approaches to establishing short-term culture of PGCs in defined conditions.


Assuntos
Técnicas de Cultura de Células/métodos , Células Germinativas Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Animais , Adesão Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Camundongos
10.
Methods Mol Biol ; 2214: 75-89, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32944904

RESUMO

The specification and development of germ cells to gametes is a unique process, which is of great biological and clinical relevance. In mammals, the founding cells of the germline are primordial germ cells (PGCs), which arise during early embryogenesis. The low number of PGCs within the developing embryo limits the study of these cells in model organisms. The generation of PGC-like cells (PGCLCs) from murine pluripotent stem cells reconstitutes the earliest stages of germ cell development and mitigates the technical constraints of studying this developmental process in vivo. Here, we describe the technical details of the PGCLC specification approach and illustrate adaptations designed to improve compatibility with methods such as chromatin immunoprecipitation by increasing the yield of PGCLC generation.


Assuntos
Células Germinativas Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular , Embrião de Mamíferos/citologia , Camundongos
11.
Methods Mol Biol ; 2214: 91-108, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32944905

RESUMO

Knockout CRISPR screening enables the unbiased discovery of genes with a functional role in almost any cellular or molecular process of interest. The approach couples a genome-scale library of guide RNA (gRNA), the Cas9 endonuclease, and a faithful phenotypic read-out to systematically identify candidate genes via their loss-of-function effect. Here we provide a detailed description of the CRISPR screen protocol and outline how to apply it to decipher the gene networks that underlie developmental cell fate decisions. As a paradigm we use the in vitro model of cell state transition(s) from naive pluripotency to primordial germ cell (PGC) fate, exploiting the Stella-GFP:Esg1-tdTomato (SGET) mouse ESC line. The principles in this protocol can be readily adapted to characterize lineage regulators for other cell fate models and/or for other species.


Assuntos
Sistemas CRISPR-Cas , Células Germinativas Embrionárias/citologia , Células-Tronco Embrionárias Murinas/citologia , Animais , Diferenciação Celular , Linhagem Celular , Células Germinativas Embrionárias/metabolismo , Redes Reguladoras de Genes , Células HEK293 , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , RNA Guia de Cinetoplastídeos/genética , Transdução Genética
12.
Methods Mol Biol ; 2214: 253-264, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32944915

RESUMO

Cleavage under targets and release using nuclease (CUT&RUN) allows the chromatin profiling of proteins of interest for which specific antibodies are available. Because it is performed on intact chromatin in situ, CUT&RUN provides exceptional signal over background, making it an ideal choice for chromatin profiling on primary cells available at limited numbers. Here, we describe its application to the profiling of histone post-translational modifications in germ cells isolated from mouse embryos from 12.5 up to 18.5 days postfertilization. This approach can be applied to as low as 100 isolated germ cells, allowing the generation of multiple genome-wide profiles from the cells obtained from a single embryo.


Assuntos
Cromatina/genética , Células Germinativas Embrionárias/metabolismo , Código das Histonas , Camundongos/genética , Animais , Separação Celular/métodos , Células Cultivadas , Células Germinativas Embrionárias/citologia , Biblioteca Gênica , Camundongos/embriologia , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional
13.
Nat Commun ; 11(1): 5656, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168808

RESUMO

Establishment of spermatogonia throughout the fetal and postnatal period is essential for production of spermatozoa and male fertility. Here, we establish a protocol for in vitro reconstitution of human prospermatogonial specification whereby human primordial germ cell (PGC)-like cells differentiated from human induced pluripotent stem cells are further induced into M-prospermatogonia-like cells and T1 prospermatogonia-like cells (T1LCs) using long-term cultured xenogeneic reconstituted testes. Single cell RNA-sequencing is used to delineate the lineage trajectory leading to T1LCs, which closely resemble human T1-prospermatogonia in vivo and exhibit gene expression related to spermatogenesis and diminished proliferation, a hallmark of quiescent T1 prospermatogonia. Notably, this system enables us to visualize the dynamic and stage-specific regulation of transposable elements during human prospermatogonial specification. Together, our findings pave the way for understanding and reconstructing human male germline development in vitro.


Assuntos
Células Germinativas Embrionárias/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Espermatogênese/genética , Espermatogênese/fisiologia , Animais , Diferenciação Celular , Epigenômica , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Elementos Reguladores de Transcrição , Análise de Sequência de RNA , Espermatogônias/citologia , Espermatozoides , Testículo/citologia , Transcriptoma
14.
Genes Dev ; 34(23-24): 1637-1649, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33184219

RESUMO

Germ cells specified during fetal development form the foundation of the mammalian germline. These primordial germ cells (PGCs) undergo rapid proliferation, yet the germline is highly refractory to mutation accumulation compared with somatic cells. Importantly, while the presence of endogenous or exogenous DNA damage has the potential to impact PGCs, there is little known about how these cells respond to stressors. To better understand the DNA damage response (DDR) in these cells, we exposed pregnant mice to ionizing radiation (IR) at specific gestational time points and assessed the DDR in PGCs. Our results show that PGCs prior to sex determination lack a G1 cell cycle checkpoint. Additionally, the response to IR-induced DNA damage differs between female and male PGCs post-sex determination. IR of female PGCs caused uncoupling of germ cell differentiation and meiotic initiation, while male PGCs exhibited repression of piRNA metabolism and transposon derepression. We also used whole-genome single-cell DNA sequencing to reveal that genetic rescue of DNA repair-deficient germ cells (Fancm-/- ) leads to increased mutation incidence and biases. Importantly, our work uncovers novel insights into how PGCs exposed to DNA damage can become developmentally defective, leaving only those genetically fit cells to establish the adult germline.


Assuntos
Dano ao DNA , DNA/efeitos da radiação , Células Germinativas Embrionárias/efeitos da radiação , Células Germinativas/efeitos da radiação , Mutação/genética , Radiação Ionizante , Animais , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Diferenciação Celular/efeitos da radiação , Elementos de DNA Transponíveis/efeitos da radiação , Células Germinativas Embrionárias/citologia , Feminino , Masculino , Meiose/genética , Meiose/efeitos da radiação , Camundongos , Oócitos/citologia , Oócitos/efeitos da radiação , Gravidez , RNA Interferente Pequeno/metabolismo , Fatores Sexuais
15.
Biochem Biophys Res Commun ; 533(4): 938-944, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33008598

RESUMO

Arginine methylation is one of the most important post-translational modifications which is catalyzed by protein arginine methyltransferases (PRMTs). Previous studies have demonstrated that Prmt5 plays important role in germ cell development. Prmt7 is the only family member responsible for mono-methylation of arginine residue. However, whether Prmt7 is also involved in germ cell development remains unclear. In this study, we find that PRMT7 is abundantly expressed in the male germ cells during embryonic stage (from E10.5). Depletion of Prmt7 results in the defect of germ cell proliferation during embryonic stage and the number of primordial germ cells is significantly reduced in Prmt7-/- mice at E11.5. We also find that the size of testes is reduced in Prmt7-/- mice at P5 with reduced germ cell number and the diameter of seminiferous tubules. Further study reveals that the expression of BMPs and TGF-ß singling pathway is significantly changed in germ cells of Prmt7-/- mice at E12.5. However, no defect of testes development is observed in adult Prmt7-/flox; Mvh-Cre mice. Collectively, this study demonstrates that Prmt7 plays roles in male germ cell proliferation during embryonic stages and it is not required for germ cell development postnatally.


Assuntos
Células Germinativas Embrionárias/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Células Germinativas Embrionárias/citologia , Epigênese Genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteína-Arginina N-Metiltransferases/deficiência , Proteína-Arginina N-Metiltransferases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Testículo/citologia , Testículo/embriologia , Fator de Crescimento Transformador beta/genética
16.
In Vitro Cell Dev Biol Anim ; 56(9): 808-815, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33029688

RESUMO

Homologous feeder culture system can efficiently promote the proliferation of embryonic germ (EG) cells or embryonic stem (ES) cells while avoiding contamination by exogenous proteins and pathogens. In this study, we compared the potency of using homologous porcine embryonic fibroblasts (PEFs), gonadal stromal cells (GSCs), porcine adipose-derived stem cells (PASCs), or porcine amniotic fluid stem (PAFS) cells as feeder cells for porcine EG growth, with the commonly used mouse embryonic fibroblasts (MEFs). We compared the feeder cell growth rates; secretion of growth factors including stem cell factor (SCF), basic fibroblast growth factor (bFGF), and leukemia inhibitory factor (LIF); the effects of growth factors on porcine PGC growth; and EG growth rates when individual cells were used as feeders. Our results showed that feeder cells secreted limited amounts of growth factors, and supplementation of growth factors can significantly improve the formation of EG colonies and number of passages (P < 0.05). GSC and PEF were more suitable for EG growth because of their faster growth rate and their support on EG growth. In conclusion, this study identified novel homologous cells that can be used for EG production.


Assuntos
Técnicas de Cultura de Células/métodos , Células Germinativas Embrionárias/citologia , Animais , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/citologia , Células Alimentadoras/citologia , Feminino , Fibroblastos/citologia , Camundongos , Suínos
17.
Infect Genet Evol ; 85: 104521, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32882433

RESUMO

The recent pandemic of Zika virus (ZIKV) infections highlight the urgent need for the development of a safe and efficacious ZIKV vaccine. We previously demonstrated that robust humoral and cellular immunity was elicited in BALB/c mice by ZIKV DNA vaccine encoding the precursor membrane (prM) and envelope (E) proteins while the protective efficacies were not evaluated against ZIKV challenge. To further explore the protective immunity elicited by various targets of ZIKV, we constructed a novel DNA-based vaccine expressing nonstructural protein 1 (NS1), named as VRC-NS1, and evaluated and compared immune responses and protective efficacies of three ZIKV DNA vaccine candidates (VRC-prME, VRC-NS1, and VRC-prME+VRC-NS1) using an A129 (Ifnar-/-) murine challenge model. The results showed that each of DNA vaccine candidates induced robust antigen-specific humoral immunity and conferred protection against ZIKV-SMGC-1 with two doses (20 µg per dose) of homologous intramuscularly (i.m.) immunizations via in vivo electroporation. All DNA vaccine candidates induced significant protection against infection-associated weight loss in addition to preventing viral replication in blood, brain and spleen tissue following in vivo viral challenge. Notably, NS1-based DNA vaccination alone was capable of conferring mouse protective immunity to reduce viremia and viral burden in tissues against ZIKV challenge, even though it did not induce neutralizing antibodies. These data demonstrated that VRC-NS1 and VRC-prME are highly promising vaccine candidates for ZIKV control. Furthermore, our results highlight an alternative strategy (DNA vaccine based on non-neutralizing antigen NS1) for designing novel flaviviral vaccines (including for ZIKV) and provide a foundation for the development of a safe and effective NS1-based vaccine against ZIKV infection.


Assuntos
Células Cultivadas/efeitos dos fármacos , Imunidade Ativa/efeitos dos fármacos , Imunidade Ativa/genética , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Células Germinativas Embrionárias/efeitos dos fármacos , Feminino , Variação Genética , Genótipo , Humanos , Rim/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Vacinação , Células Vero/efeitos dos fármacos , Proteínas não Estruturais Virais/genética , Vacinas Virais/genética , Vacinas Virais/imunologia
18.
Nat Commun ; 11(1): 4477, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32901019

RESUMO

Individual cells detach from cohesive ensembles during development and can inappropriately separate in disease. Although much is known about how cells separate from epithelia, it remains unclear how cells disperse from clusters lacking apical-basal polarity, a hallmark of advanced epithelial cancers. Here, using live imaging of the developmental migration program of Drosophila primordial germ cells (PGCs), we show that cluster dispersal is accomplished by stabilizing and orienting migratory forces. PGCs utilize a G protein coupled receptor (GPCR), Tre1, to guide front-back migratory polarity radially from the cluster toward the endoderm. Posteriorly positioned myosin-dependent contractile forces pull on cell-cell contacts until cells release. Tre1 mutant cells migrate randomly with transient enrichment of the force machinery but fail to separate, indicating a temporal contractile force threshold for detachment. E-cadherin is retained on the cell surface during cell separation and augmenting cell-cell adhesion does not impede detachment. Notably, coordinated migration improves cluster dispersal efficiency by stabilizing cell-cell interfaces and facilitating symmetric pulling. We demonstrate that guidance of inherent migratory forces is sufficient to disperse cell clusters under physiological settings and present a paradigm for how such events could occur across development and disease.


Assuntos
Drosophila melanogaster/embriologia , Células Germinativas Embrionárias/fisiologia , Animais , Animais Geneticamente Modificados , Fenômenos Biomecânicos , Padronização Corporal/fisiologia , Caderinas/metabolismo , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/fisiologia , Células Germinativas Embrionárias/citologia , Microscopia de Fluorescência por Excitação Multifotônica , Miosina Tipo II/metabolismo , Transdução de Sinais , Análise de Célula Única , Proteínas rho de Ligação ao GTP/metabolismo
19.
Genesis ; 58(8): e23388, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32776392

RESUMO

PR domain zinc finger protein 14 (PRDM14) plays an essential role in the development of primordial germ cells (PGCs) in mice. However, its functions in avian species remain unclear. In the present study, we used CRISPR/Cas9 to edit the PRDM14 locus in chickens in order to demonstrate its importance in development. The eGFP gene was introduced into the PRDM14 locus of cultured chicken PGCs to knockout PRDM14 and label PGCs. Chimeric chickens were established by a direct injection of eGFP knocked-in (gene-trapped) PGCs into the blood vessels of Hamburger-Hamilton stages (HH-stages) 13-16 chicken embryos. Gene-trapped chickens were established by crossing a chimeric chicken with a wild-type hen with very high efficiency. Heterozygous gene-trapped chickens grew normally and SSEA-1-positive cells expressed eGFP during HH-stages 13-30. These results indicated the specific expression of eGFP within circulating PGCs and gonadal PGCs. At the blastodermal stage, the ratio of homozygous gene-trapped embryos obtained by crossing heterozygous gene-trapped roosters and hens was almost normal; however, all embryos died soon afterward, suggesting the important roles of PRDM14 in chicken early development.


Assuntos
Células Germinativas Embrionárias/metabolismo , Marcação de Genes/métodos , Proteínas de Fluorescência Verde/genética , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Células Cultivadas , Embrião de Galinha , Proteínas de Fluorescência Verde/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transgenes
20.
Biol Reprod ; 103(4): 717-735, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32627815

RESUMO

Regulatory mechanisms of germline differentiation have generally been explained via the function of signaling pathways, transcription factors, and epigenetic regulation; however, little is known regarding proteomic and metabolomic regulation and their contribution to germ cell development. Here, we conducted integrated proteomic and metabolomic analyses of fetal germ cells in mice on embryonic day (E)13.5 and E18.5 and demonstrate sex- and developmental stage-dependent changes in these processes. In male germ cells, RNA processing, translation, oxidative phosphorylation, and nucleotide synthesis are dominant in E13.5 and then decline until E18.5, which corresponds to the prolonged cell division and more enhanced hyper-transcription/translation in male primordial germ cells and their subsequent repression. Tricarboxylic acid cycle and one-carbon pathway are consistently upregulated in fetal male germ cells, suggesting their involvement in epigenetic changes preceding in males. Increased protein stability and oxidative phosphorylation during female germ cell differentiation suggests an upregulation of aerobic energy metabolism, which likely contributes to the proteostasis required for oocyte maturation in subsequent stages. The features elucidated in this study shed light on the unrevealed mechanisms of germ cell development.


Assuntos
Diferenciação Celular/fisiologia , Células Germinativas Embrionárias/fisiologia , Metabolômica , Proteômica , Animais , DNA/genética , DNA/metabolismo , Metilação de DNA , Embrião de Mamíferos/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Estudo de Associação Genômica Ampla , Masculino , Camundongos , Camundongos Transgênicos , Diferenciação Sexual , Fatores Sexuais , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...