Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
2.
Nature ; 573(7773): 271-275, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31485074

RESUMO

Development is often assumed to be hardwired in the genome, but several lines of evidence indicate that it is susceptible to environmental modulation with potential long-term consequences, including in mammals1,2. The embryonic germline is of particular interest because of the potential for intergenerational epigenetic effects. The mammalian germline undergoes extensive DNA demethylation3-7 that occurs in large part by passive dilution of methylation over successive cell divisions, accompanied by active DNA demethylation by TET enzymes3,8-10. TET activity has been shown to be modulated by nutrients and metabolites, such as vitamin C11-15. Here we show that maternal vitamin C is required for proper DNA demethylation and the development of female fetal germ cells in a mouse model. Maternal vitamin C deficiency does not affect overall embryonic development but leads to reduced numbers of germ cells, delayed meiosis and reduced fecundity in adult offspring. The transcriptome of germ cells from vitamin-C-deficient embryos is remarkably similar to that of embryos carrying a null mutation in Tet1. Vitamin C deficiency leads to an aberrant DNA methylation profile that includes incomplete demethylation of key regulators of meiosis and transposable elements. These findings reveal that deficiency in vitamin C during gestation partially recapitulates loss of TET1, and provide a potential intergenerational mechanism for adjusting fecundity to environmental conditions.


Assuntos
Ácido Ascórbico/metabolismo , Metilação de DNA/fisiologia , Células Germinativas/fisiologia , Transcriptoma/fisiologia , Animais , Deficiência de Ácido Ascórbico/fisiopatologia , Contagem de Células , Proteínas de Ligação a DNA/genética , Epigenômica , Feminino , Mutação com Perda de Função , Meiose/fisiologia , Camundongos , Modelos Animais , Gravidez , Proteínas Proto-Oncogênicas/genética
4.
Cell ; 132(4): 612-30, 2008 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-18295579

RESUMO

Regulated movement of stem cells is critical for organogenesis during development and for homeostasis and repair in adulthood. Here we analyze the biological significance and molecular mechanisms underlying stem cell trafficking in the generation of the germline, and the generation and regeneration of blood and muscle. Comparison across organisms and lineages reveals remarkable conservation as well as specialization in homing and migration mechanisms used by mature leukocytes, adult and fetal stem cells, and cancer stem cells. In vivo trafficking underpins the successful therapeutic application of hematopoietic stem cells for bone-marrow transplant, and further elucidation of homing and migration pathways in other systems will enable broader application of stem cells for targeted cell therapy and drug delivery.


Assuntos
Movimento Celular , Células-Tronco/citologia , Células-Tronco Adultas/citologia , Animais , Adesão Celular , Desenvolvimento Embrionário , Humanos , Metástase Neoplásica , Neoplasias/metabolismo
5.
Development ; 146(12)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30658985

RESUMO

Epigenetic resetting in germ cells during development de-represses transposable elements (TEs). piRNAs protect fetal germ cells by targeted mRNA destruction and deposition of repressive epigenetic marks. Here, we provide the first evidence for an active piRNA pathway and TE repression in germ cells of human fetal testis. We identify pre-pachytene piRNAs with features of secondary amplification that map most abundantly to the long interspersed element type 1 (L1) family of TEs. L1-ORF1p expression is heterogeneous in fetal germ cells, peaks at mid-gestation and declines concomitantly with increases in piRNAs, nuclear localization of HIWI2 and an increase in H3K9me3. Surprisingly, the same cells with accumulation of L1-ORF1p display highest levels of HIWI2 and H3K9me3. Conversely, the earliest germ cells with high levels of L1-ORF1p express low levels of the chaperone HSP90α. We propose that a subset of germ cells resists L1 expression, whereas L1-expressing germ cells activate the repression pathway that leads to epigenetic silencing of L1 via H3K9me3.


Assuntos
Elementos de DNA Transponíveis , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , RNA Interferente Pequeno/genética , Testículo/embriologia , Animais , Proteínas Argonautas/metabolismo , Núcleo Celular/metabolismo , Análise por Conglomerados , Epigênese Genética , Perfilação da Expressão Gênica , Inativação Gênica , Proteínas de Choque Térmico HSP90/metabolismo , Xenoenxertos , Histonas/metabolismo , Homozigoto , Humanos , Masculino , Camundongos , Chaperonas Moleculares , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Análise de Célula Única , Testículo/transplante
6.
Development ; 143(24): 4749-4754, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27836961

RESUMO

Although much is known about the embryo during implantation, the architecture of the uterine environment in which the early embryo develops is not well understood. We employed confocal imaging in combination with 3D analysis to identify and quantify dynamic changes to the luminal structure of murine uterus in preparation for implantation. When applied to mouse mutants with known implantation defects, this method detected striking peri-implantation abnormalities in uterine morphology that cannot be visualized by histology. We revealed 3D organization of uterine glands and found that they undergo a stereotypical reorientation concurrent with implantation. Furthermore, we extended this technique to generate a 3D rendering of the cycling human endometrium. Analyzing the uterine and embryo structure in 3D for different genetic mutants and pathological conditions will help uncover novel molecular pathways and global structural changes that contribute to successful implantation of an embryo.


Assuntos
Blastocisto/ultraestrutura , Implantação do Embrião/fisiologia , Embrião de Mamíferos/ultraestrutura , Endométrio/ultraestrutura , Útero/ultraestrutura , Animais , Embrião de Mamíferos/diagnóstico por imagem , Endométrio/diagnóstico por imagem , Endométrio/fisiologia , Feminino , Humanos , Imageamento Tridimensional , Camundongos , Camundongos Endogâmicos C57BL , Útero/diagnóstico por imagem , Útero/fisiologia , Proteína Wnt-5a/genética
7.
Nature ; 562(7728): 497-498, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30349122
8.
Nature ; 500(7461): 222-6, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23812591

RESUMO

DNA methylation is a heritable epigenetic modification involved in gene silencing, imprinting, and the suppression of retrotransposons. Global DNA demethylation occurs in the early embryo and the germ line, and may be mediated by Tet (ten eleven translocation) enzymes, which convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Tet enzymes have been studied extensively in mouse embryonic stem (ES) cells, which are generally cultured in the absence of vitamin C, a potential cofactor for Fe(II) 2-oxoglutarate dioxygenase enzymes such as Tet enzymes. Here we report that addition of vitamin C to mouse ES cells promotes Tet activity, leading to a rapid and global increase in 5hmC. This is followed by DNA demethylation of many gene promoters and upregulation of demethylated germline genes. Tet1 binding is enriched near the transcription start site of genes affected by vitamin C treatment. Importantly, vitamin C, but not other antioxidants, enhances the activity of recombinant Tet1 in a biochemical assay, and the vitamin-C-induced changes in 5hmC and 5mC are entirely suppressed in Tet1 and Tet2 double knockout ES cells. Vitamin C has a stronger effect on regions that gain methylation in cultured ES cells compared to blastocysts, and in vivo are methylated only after implantation. In contrast, imprinted regions and intracisternal A particle retroelements, which are resistant to demethylation in the early embryo, are resistant to vitamin-C-induced DNA demethylation. Collectively, the results of this study establish vitamin C as a direct regulator of Tet activity and DNA methylation fidelity in ES cells.


Assuntos
Ácido Ascórbico/farmacologia , Metilação de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , 5-Metilcitosina/análogos & derivados , Animais , Antioxidantes/farmacologia , Blastocisto/metabolismo , Linhagem Celular , Meios de Cultura/química , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Inativação de Genes , Camundongos , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
J Cell Sci ; 129(6): 1271-82, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26826184

RESUMO

Meiotic progression requires exquisitely coordinated translation of maternal messenger (m)RNA that has accumulated during oocyte growth. A major regulator of this program is the cytoplasmic polyadenylation element binding protein 1 (CPEB1). However, the temporal pattern of translation at different meiotic stages indicates the function of additional RNA binding proteins (RBPs). Here, we report that deleted in azoospermia-like (DAZL) cooperates with CPEB1 to regulate maternal mRNA translation. Using a strategy that monitors ribosome loading onto endogenous mRNAs and a prototypic translation target, we show that ribosome loading is induced in a DAZL- and CPEB1-dependent manner, as the oocyte reenters meiosis. Depletion of the two RBPs from oocytes and mutagenesis of the 3' untranslated regions (UTRs) demonstrate that both RBPs interact with the Tex19.1 3' UTR and cooperate in translation activation of this mRNA. We observed a synergism between DAZL and cytoplasmic polyadenylation elements (CPEs) in the translation pattern of maternal mRNAs when using a genome-wide analysis. Mechanistically, the number of DAZL proteins loaded onto the mRNA and the characteristics of the CPE might define the degree of cooperation between the two RBPs in activating translation and meiotic progression.


Assuntos
Oócitos/citologia , Oócitos/metabolismo , Oogênese , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Fatores de Poliadenilação e Clivagem de mRNA/genética
10.
J Cell Sci ; 129(13): 2493-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199373

RESUMO

Mouse ovarian germ cells enter meiosis in a wave that propagates from anterior to posterior, but little is known about contribution of germ cells to initiation or propagation of meiosis. In a Ror2 mutant with diminished germ cell number and migration, we find that overall timing of meiotic initiation is delayed at the population level. We use chemotherapeutic depletion to exclude a profoundly reduced number of germ cells as a cause for meiotic delay. We rule out sex reversal or failure to specify somatic support cells as contributors to the meiotic phenotype. Instead, we find that anomalies in the distribution of germ cells as well as gonad shape in mutants contribute to aberrant initiation of meiosis. Our analysis supports a model of meiotic initiation via diffusible signal(s), excludes a role for germ cells in commencing the meiotic wave and furnishes the first phenotypic demonstration of the wave of meiotic entry. Finally, our studies underscore the importance of considering germ cell migration defects while studying meiosis to discern secondary effects resulting from positioning versus primary meiotic entry phenotypes.


Assuntos
Células Germinativas/metabolismo , Gônadas/patologia , Meiose/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Animais , Contagem de Células , Movimento Celular/genética , Forma Celular/genética , Feminino , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/patologia , Gônadas/crescimento & desenvolvimento , Camundongos , Mutação , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Ovário/patologia , Transdução de Sinais/genética
11.
Stem Cells ; 34(7): 1985-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27066911

RESUMO

The embryonic stem cell cycle (ESCC) and let-7 families of miRNAs function antagonistically in the switch between mouse embryonic stem cell self-renewal and somatic differentiation. Here, we report that the human ESCC miRNA miR-372 and let-7 act antagonistically in germline differentiation from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs). hESC and iPSC-derived primordial germ cell-like cells (PGCLCs) expressed high levels of miR-372 and conversely, somatic cells expressed high levels of let-7. Manipulation of miRNA levels by introduction of miRNA mimics or knockdown with miRNA sponges demonstrated that miR-372 promotes whereas let-7 antagonizes PGCLC differentiation. Knockdown of the individual miR-372 targets SMARCC1, MECP2, CDKN1, RBL2, RHOC, and TGFBR2 increased PGCLC production, whereas knockdown of the let-7 targets CMYC and NMYC suppressed PGCLC differentiation. These findings uncover a miR-372/let-7 axis regulating human primordial germ cell (PGC) specification. Stem Cells 2016;34:1985-1991.


Assuntos
Linhagem da Célula , Células Germinativas/citologia , Células Germinativas/metabolismo , MicroRNAs/metabolismo , Transdução de Sinais/genética , Biomarcadores/metabolismo , Humanos , MicroRNAs/genética
12.
Dev Biol ; 403(1): 69-79, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25889274

RESUMO

Quantitative analysis of tissues and organs can reveal large-scale patterning as well as the impact of perturbations and aging on biological architecture. Here we develop tools for imaging of single cells in intact organs and computational approaches to assess spatial relationships in 3D. In the mouse ovary, we use nuclear volume of the oocyte to read out quiescence or growth of oocyte-somatic cell units known as follicles. This in-ovary quantification of non-growing follicle dynamics from neonate to adult fits a mathematical function, which corroborates the model of fixed oocyte reserve. Mapping approaches show that radial organization of folliculogenesis established in the newborn ovary is preserved through adulthood. By contrast, inter-follicle clustering increases during aging with different dynamics depending on size. These broadly applicable tools can reveal high dimensional phenotypes and age-related architectural changes in other organs. In the adult mouse pancreas, we find stochastic radial organization of the islets of Langerhans but evidence for localized interactions among the smallest islets.


Assuntos
Imageamento Tridimensional/métodos , Ilhotas Pancreáticas/fisiologia , Oócitos/fisiologia , Folículo Ovariano/fisiologia , Análise de Célula Única/métodos , Envelhecimento , Algoritmos , Animais , Feminino , Ilhotas Pancreáticas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Oócitos/ultraestrutura , Folículo Ovariano/ultraestrutura
13.
Hum Mol Genet ; 22(2): 313-27, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23077213

RESUMO

KitL, via its receptor cKit, supports primordial germ cell (PGC) growth, survival, migration and reprogramming to pluripotent embryonic germ cells (EGCs). However, the signaling downstream of KitL and its regulation in PGCs remain unclear. A constitutively activating mutation, cKit(V558Δ), causes gain-of-function phenotypes in mast cells and intestines, and gastrointestinal stromal tumors (GISTs) when heterozygous. Unexpectedly, we find that PGC growth is not significantly affected in cKit(V558Δ) heterozygotes, whereas in homozygotes, increased apoptosis and inefficient migration lead to the depletion of PGCs. Through genetic studies, we reveal that this oncogenic cKit allele exhibits loss-of-function behavior in PGCs distinct from that in GIST development. Examination of downstream signaling in GISTs from cKit(V558Δ/+) mice confirmed hyperphosphorylation of AKT and ERK, but both remain unperturbed in cKit(V558Δ/+) PGCs and EGCs. In contrast, we find reduced activation of ERK1/2 and JNK1 in cKit(V558Δ) homozygous PGCs and EGCs. Inhibiting JNK, though not ERK1/2, increased apoptosis of wild-type PGCs, but did not further affect the already elevated apoptosis of cKit(V558Δ)(/V558Δ) PGCs. These results demonstrate a cell-context-dependent response to the cKit(V558Δ) mutation. We propose that AKT overload protection and JNK-mediated survival comprise PGC-specific mechanisms for regulating cKit signaling.


Assuntos
Alelos , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/metabolismo , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/metabolismo , Células Germinativas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ativação Enzimática , Feminino , Genótipo , Células Germinativas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais
14.
Dev Dyn ; 243(8): 1037-45, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24753105

RESUMO

BACKGROUND: The noncanonical Wnt receptor and tyrosine kinase Ror2 has been associated with recessive Robinow syndrome (RRS) and dominant brachydactyly type B1. The phenotypes of mouse mutants implicate Ror2 in the development of the heart, lungs, bone, and craniofacial structures, which are affected in RRS. Following a recently identified role of Ror2 in the migration of mouse primordial germ cells, we extensively characterized its expression throughout the fetal internal reproductive system and the postnatal ductal system. RESULTS: We show that Ror2 gene products are present in the germ cells and somatic cells of the testis and the ovary of both the mouse and human fetus. In reproductive tract structures, we find that Ror2 is expressed in the mesonephros, developing Wolffian and Müllerian ducts, and later in their derivatives, the epididymal epithelium and uterine epithelium. CONCLUSIONS: This study sets the stage to explore function for this tyrosine kinase receptor in novel regions of expression in the developing reproductive system in both mouse and human.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Animais , Feminino , Células Germinativas/citologia , Células da Granulosa , Humanos , Masculino , Camundongos , Ductos Paramesonéfricos/citologia , Ovário/citologia , Proteínas Proto-Oncogênicas/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Células de Sertoli/citologia , Testículo/citologia , Proteínas Wnt/genética , Proteína Wnt-5a , Ductos Mesonéfricos/citologia
15.
PLoS Genet ; 7(12): e1002428, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22216013

RESUMO

The trafficking of primordial germ cells (PGCs) across multiple embryonic structures to the nascent gonads ensures the transmission of genetic information to the next generation through the gametes, yet our understanding of the mechanisms underlying PGC migration remains incomplete. Here we identify a role for the receptor tyrosine kinase-like protein Ror2 in PGC development. In a Ror2 mouse mutant we isolated in a genetic screen, PGC migration and survival are dysregulated, resulting in a diminished number of PGCs in the embryonic gonad. A similar phenotype in Wnt5a mutants suggests that Wnt5a acts as a ligand to Ror2 in PGCs, although we do not find evidence that WNT5A functions as a PGC chemoattractant. We show that cultured PGCs undergo polarization, elongation, and reorientation in response to the chemotactic factor SCF (secreted KitL), whereas Ror2 PGCs are deficient in these SCF-induced responses. In the embryo, migratory PGCs exhibit a similar elongated geometry, whereas their counterparts in Ror2 mutants are round. The protein distribution of ROR2 within PGCs is asymmetric, both in vitro and in vivo; however, this asymmetry is lost in Ror2 mutants. Together these results indicate that Ror2 acts autonomously to permit the polarized response of PGCs to KitL. We propose a model by which Wnt5a potentiates PGC chemotaxis toward secreted KitL by redistribution of Ror2 within the cell.


Assuntos
Polaridade Celular/genética , Células Germinativas/fisiologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Fator de Células-Tronco/genética , Proteínas Wnt/genética , Animais , Movimento Celular/genética , Células Cultivadas , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Fenótipo , Fator de Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt-5a
16.
Elife ; 122024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38226689

RESUMO

While physiologic stress has long been known to impair mammalian reproductive capacity through hormonal dysregulation, mounting evidence now suggests that stress experienced prior to or during gestation may also negatively impact the health of future offspring. Rodent models of gestational physiologic stress can induce neurologic and behavioral changes that persist for up to three generations, suggesting that stress signals can induce lasting epigenetic changes in the germline. Treatment with glucocorticoid stress hormones is sufficient to recapitulate the transgenerational changes seen in physiologic stress models. These hormones are known to bind and activate the glucocorticoid receptor (GR), a ligand-inducible transcription factor, thus implicating GR-mediated signaling as a potential contributor to the transgenerational inheritance of stress-induced phenotypes. Here, we demonstrate dynamic spatiotemporal regulation of GR expression in the mouse germline, showing expression in the fetal oocyte as well as the perinatal and adult spermatogonia. Functionally, we find that fetal oocytes are intrinsically buffered against changes in GR signaling, as neither genetic deletion of GR nor GR agonism with dexamethasone altered the transcriptional landscape or the progression of fetal oocytes through meiosis. In contrast, our studies revealed that the male germline is susceptible to glucocorticoid-mediated signaling, specifically by regulating RNA splicing within the spermatogonia, although this does not abrogate fertility. Together, our work suggests a sexually dimorphic function for GR in the germline, and represents an important step towards understanding the mechanisms by which stress can modulate the transmission of genetic information through the germline.


Assuntos
Glucocorticoides , Receptores de Glucocorticoides , Gravidez , Masculino , Feminino , Camundongos , Animais , Glucocorticoides/farmacologia , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição , Células Germinativas/metabolismo , Oócitos/metabolismo , Mamíferos/metabolismo
17.
Dev Cell ; 59(8): 1010-1027.e8, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38569549

RESUMO

Ten-eleven translocation (TET) enzymes iteratively oxidize 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC), 5-formylcytosine, and 5-carboxylcytosine to facilitate active genome demethylation. Whether these bases are required to promote replication-coupled dilution or activate base excision repair during mammalian germline reprogramming remains unresolved due to the inability to decouple TET activities. Here, we generated two mouse lines expressing catalytically inactive TET1 (Tet1-HxD) and TET1 that stalls oxidation at 5hmC (Tet1-V). Tet1 knockout and catalytic mutant primordial germ cells (PGCs) fail to erase methylation at select imprinting control regions and promoters of meiosis-associated genes, validating the requirement for the iterative oxidation of 5mC for complete germline reprogramming. TET1V and TET1HxD rescue most hypermethylation of Tet1-/- sperm, suggesting the role of TET1 beyond its oxidative capability. We additionally identify a broader class of hypermethylated regions in Tet1 mutant mouse sperm that depend on TET oxidation for reprogramming. Our study demonstrates the link between TET1-mediated germline reprogramming and sperm methylome patterning.


Assuntos
5-Metilcitosina , 5-Metilcitosina/análogos & derivados , Metilação de DNA , Proteínas de Ligação a DNA , Impressão Genômica , Oxirredução , Proteínas Proto-Oncogênicas , Espermatozoides , Animais , Masculino , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Espermatozoides/metabolismo , 5-Metilcitosina/metabolismo , Reprogramação Celular/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL
18.
Mol Ther Methods Clin Dev ; 32(2): 101263, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38827250

RESUMO

Prenatal somatic cell gene therapy (PSCGT) could potentially treat severe, early-onset genetic disorders such as spinal muscular atrophy (SMA) or muscular dystrophy. Given the approval of adeno-associated virus serotype 9 (AAV9) vectors in infants with SMA by the U.S. Food and Drug Administration, we tested the safety and biodistribution of AAV9-GFP (clinical-grade and dose) in fetal lambs to understand safety and efficacy after umbilical vein or intracranial injection on embryonic day 75 (E75) . Umbilical vein injection led to widespread biodistribution of vector genomes in all examined lamb tissues and in maternal uteruses at harvest (E96 or E140; term = E150). There was robust GFP expression in brain, spinal cord, dorsal root ganglia (DRGs), without DRG toxicity and excellent transduction of diaphragm and quadriceps muscles. However, we found evidence of systemic toxicity (fetal growth restriction) and maternal exposure to the viral vector (transient elevation of total bilirubin and a trend toward elevation in anti-AAV9 antibodies). There were no antibodies against GFP in ewes or lambs. Analysis of fetal gonads demonstrated GFP expression in female (but not male) germ cells, with low levels of integration-specific reads, without integration in select proto-oncogenes. These results suggest potential therapeutic benefit of AAV9 PSCGT for neuromuscular disorders, but warrant caution for exposure of female germ cells.

19.
bioRxiv ; 2023 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-37425891

RESUMO

While physiologic stress has long been known to impair mammalian reproductive capacity through hormonal dysregulation, mounting evidence now suggests that stress experienced prior to or during gestation may also negatively impact the health of future offspring. Rodent models of gestational physiologic stress can induce neurologic and behavioral changes that persist for up to three generations, suggesting that stress signals can induce lasting epigenetic changes in the germline. Treatment with glucocorticoid stress hormones is sufficient to recapitulate the transgenerational changes seen in physiologic stress models. These hormones are known to bind and activate the glucocorticoid receptor (GR), a ligand-inducible transcription factor, thus implicating GR-mediated signaling as a potential contributor to the transgenerational inheritance of stress-induced phenotypes. Here we demonstrate dynamic spatiotemporal regulation of GR expression in the mouse germline, showing expression in the fetal oocyte as well as the perinatal and adult spermatogonia. Functionally, we find that fetal oocytes are intrinsically buffered against changes in GR signaling, as neither genetic deletion of GR nor GR agonism with dexamethasone altered the transcriptional landscape or the progression of fetal oocytes through meiosis. In contrast, our studies revealed that the male germline is susceptible to glucocorticoid-mediated signaling, specifically by regulating RNA splicing within the spermatogonia, although this does not abrogate fertility. Together, our work suggests a sexually dimorphic function for GR in the germline, and represents an important step towards understanding the mechanisms by which stress can modulate the transmission of genetic information through the germline.

20.
Methods Mol Biol ; 2677: 203-219, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37464244

RESUMO

Recent advances in tissue clearing methodologies have enabled three-dimensional (3D) visualization of the ovary and, consequently, in-depth exploration of the dynamic changes occurring at the single-cell level. Here we describe methods for whole-mount immunofluorescence, clearing, imaging, and analysis of whole ovarian tissue in 3D throughout murine development and aging.


Assuntos
Imageamento Tridimensional , Ovário , Feminino , Camundongos , Animais , Imageamento Tridimensional/métodos , Imunofluorescência , Envelhecimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA