Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Hum Genet ; 102(5): 845-857, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29706347

RESUMEN

Loss of expression of ACTN3, due to homozygosity of the common null polymorphism (p.Arg577X), is underrepresented in elite sprint/power athletes and has been associated with reduced muscle mass and strength in humans and mice. To investigate ACTN3 gene dosage in performance and whether expression could enhance muscle force, we performed meta-analysis and expression studies. Our general meta-analysis using a Bayesian random effects model in elite sprint/power athlete cohorts demonstrated a consistent homozygous-group effect across studies (per allele OR = 1.4, 95% CI 1.3-1.6) but substantial heterogeneity in heterozygotes. In mouse muscle, rAAV-mediated gene transfer overexpressed and rescued α-actinin-3 expression. Contrary to expectation, in vivo "doping" of ACTN3 at low to moderate doses demonstrated an absence of any change in function. At high doses, ACTN3 is toxic and detrimental to force generation, to demonstrate gene doping with supposedly performance-enhancing isoforms of sarcomeric proteins can be detrimental for muscle function. Restoration of α-actinin-3 did not enhance muscle mass but highlighted the primary role of α-actinin-3 in modulating muscle metabolism with altered fatiguability. This is the first study to express a Z-disk protein in healthy skeletal muscle and measure the in vivo effect. The sensitive balance of the sarcomeric proteins and muscle function has relevant implications in areas of gene doping in performance and therapy for neuromuscular disease.


Asunto(s)
Actinina/genética , Músculo Esquelético/fisiología , Anaerobiosis , Animales , Animales Recién Nacidos , Atletas , Calcineurina/metabolismo , Dependovirus/metabolismo , Regulación hacia Abajo/genética , Estudio de Asociación del Genoma Completo , Heterocigoto , Homocigoto , Humanos , Ratones Endogámicos C57BL , Fatiga Muscular , Fibras Musculares Esqueléticas/metabolismo , Tamaño de los Órganos , Oxidación-Reducción
2.
Dev Biol ; 389(2): 160-72, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24576538

RESUMEN

DMRT1 encodes a conserved transcription factor with an essential role in gonadal function. In the chicken, DMRT1 in located on the Z sex chromosome and is currently the best candidate master regulator of avian gonadal sex differentiation. We previously showed that knockdown of DMRT1 expression during the period of sexual differentiation induces feminisation of male embryonic chicken gonads. This gene is therefore necessary for proper testis development in the chicken. However, whether it is sufficient to induce testicular differentiation has remained unresolved. We show here that over-expression of DMRT1 induces male pathway genes and antagonises the female pathway in embryonic chicken gonads. Ectopic DMRT1 expression in female gonads induces localised SOX9 and AMH expression. It also induces expression of the recently identified Z-linked male factor, Hemogen (HEMGN). Masculinised gonads show evidence of cord-like structures and retarded female-type cortical development. Furthermore, expression of the critical feminising enzyme, aromatase, is reduced in the presence of over-expressed DMRT1. These data indicate that DMRT1 is an essential sex-linked regulator of gonadal differentiation in avians, and that it likely acts via a dosage mechanism established through the lack of global Z dosage compensation in birds.


Asunto(s)
Gónadas/embriología , Gónadas/metabolismo , Procesos de Determinación del Sexo , Factores de Transcripción/metabolismo , Animales , Hormona Antimülleriana/metabolismo , Aromatasa/genética , Aromatasa/metabolismo , Embrión de Pollo , Electroporación , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Factor de Transcripción SOX9/metabolismo , Testículo/embriología , Testículo/metabolismo , Factores de Tiempo
3.
Nature ; 461(7261): 267-71, 2009 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-19710650

RESUMEN

Sex in birds is chromosomally based, as in mammals, but the sex chromosomes are different and the mechanism of avian sex determination has been a long-standing mystery. In the chicken and all other birds, the homogametic sex is male (ZZ) and the heterogametic sex is female (ZW). Two hypotheses have been proposed for the mechanism of avian sex determination. The W (female) chromosome may carry a dominant-acting ovary determinant. Alternatively, the dosage of a Z-linked gene may mediate sex determination, two doses being required for male development (ZZ). A strong candidate avian sex-determinant under the dosage hypothesis is the conserved Z-linked gene, DMRT1 (doublesex and mab-3-related transcription factor 1). Here we used RNA interference (RNAi) to knock down DMRT1 in early chicken embryos. Reduction of DMRT1 protein expression in ovo leads to feminization of the embryonic gonads in genetically male (ZZ) embryos. Affected males show partial sex reversal, characterized by feminization of the gonads. The feminized left gonad shows female-like histology, disorganized testis cords and a decline in the testicular marker, SOX9. The ovarian marker, aromatase, is ectopically activated. The feminized right gonad shows a more variable loss of DMRT1 and ectopic aromatase activation, suggesting differential sensitivity to DMRT1 between left and right gonads. Germ cells also show a female pattern of distribution in the feminized male gonads. These results indicate that DMRT1 is required for testis determination in the chicken. Our data support the Z dosage hypothesis for avian sex determination.


Asunto(s)
Pollos/genética , Pollos/fisiología , Caracteres Sexuales , Cromosomas Sexuales/genética , Procesos de Determinación del Sexo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Biomarcadores/análisis , Línea Celular , Embrión de Pollo , Trastornos del Desarrollo Sexual , Regulación hacia Abajo , Femenino , Dosificación de Gen/genética , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Modelos Genéticos , Ovario/embriología , Ovario/metabolismo , Interferencia de ARN , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/deficiencia
4.
Dev Biol ; 366(2): 317-26, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22546690

RESUMEN

MHM is a chicken Z chromosome-linked locus that is methylated and transcriptionally silent in male cells, but is hypomethylated and transcribed into a long non-coding RNA in female cells. MHM has been implicated in both localised dosage compensation and sex determination in the chicken embryo, but direct evidence is lacking. We investigated the potential role of MHM in chicken embryonic development, using expression analysis and retroviral-mediated mis-expression. At embryonic stages, MHM is only expressed in females. Northern blotting showed that both sense and antisense strands of the MHM locus are transcribed, with the sense strand being more abundant. Whole mount in situ hybridization confirmed that the sense RNA is present in developing female embryos, notably in gonads, limbs, heart, branchial arch and brain. Within these cells, the MHM RNA is localized to the nucleus. The antisense transcript is lowly expressed and has a cytoplasmic localization in cells. Mis-expression of MHM sense and antisense sequences results in overgrowth of tissues in which transcripts are predominantly expressed. This includes altered asymmetric ovarian development in females. In males, MHM mis-expression impairs gonadal expression of the testis gene, DMRT1. Both MHM sense and antisense mis-expression cause brain abnormalities, while MHM sense causes an increase in male-biased embryo mortality. These results indicate that MHM has a role in chicken normal embryonic development, including gonadal sex differentiation.


Asunto(s)
Gónadas/embriología , ARN Largo no Codificante/fisiología , Animales , Núcleo Celular , Embrión de Pollo , Compensación de Dosificación (Genética) , Embrión no Mamífero/embriología , Embrión no Mamífero/fisiología
5.
Biol Reprod ; 85(1): 22-30, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21389341

RESUMEN

Tissue-specific patterns of microRNA (miRNA) expression contribute to organogenesis during embryonic development. Using the embryonic chicken gonads as a model for vertebrate gonadogenesis, we previously reported that miRNAs are expressed in a sexually dimorphic manner during gonadal sex differentiation. Being male biased, we hypothesised that up-regulation of microRNA 202* (MIR202*) is characteristic of testicular differentiation. To address this hypothesis, we used estrogen modulation to induce gonadal sex reversal in embryonic chicken gonads and analyzed changes in MIR202* expression. In ovo injection of estradiol-17beta at Embryonic Day 4.5 (E4.5) caused feminization of male gonads at E9.5 and reduced MIR202* expression to female levels. Female gonads treated at E3.5 with an aromatase inhibitor, which blocks estrogen synthesis, were masculinized by E9.5, and MIR202* expression was increased. Reduced MIR202* expression correlated with reduced expression of the testis-associated genes DMRT1 and SOX9, and up-regulation of ovary-associated genes FOXL2 and CYP19A1 (aromatase). Increased MIR202* expression correlated with down-regulation of FOXL2 and aromatase and up-regulation of DMRT1 and SOX9. These results confirm that up-regulation of MIR202* coincides with testicular differentiation in embryonic chicken gonads.


Asunto(s)
Estrógenos/biosíntesis , Gónadas/metabolismo , MicroARNs/metabolismo , Diferenciación Sexual , Animales , Embrión de Pollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Regulación hacia Arriba
6.
Sci Adv ; 7(27)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34215586

RESUMEN

Homozygosity for the common ACTN3 null polymorphism (ACTN3 577X) results in α-actinin-3 deficiency in ~20% of humans worldwide and is linked to reduced sprint and power performance in both elite athletes and the general population. α-Actinin-3 deficiency is also associated with reduced muscle mass, increased risk of sarcopenia, and altered muscle wasting response induced by denervation and immobilization. Here, we show that α-actinin-3 plays a key role in the regulation of protein synthesis and breakdown signaling in skeletal muscle and influences muscle mass from early postnatal development. We also show that α-actinin-3 deficiency reduces the atrophic and anti-inflammatory response to the glucocorticoid dexamethasone in muscle and protects against dexamethasone-induced muscle wasting in female but not male mice. The effects of α-actinin-3 deficiency on muscle mass regulation and response to muscle wasting provide an additional mechanistic explanation for the positive selection of the ACTN3 577X allele in recent human history.

7.
Differentiation ; 77(5): 473-82, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19395148

RESUMEN

Functional genomics in avian models has lagged behind that of mammals, and the production of transgenic birds has proven to be challenging and time-consuming. All current methods rely upon breeding chimeric birds through at least one generation. Here, we report a rapid method for the ubiquitous expression of GFP in chicken embryos in a single generation (G-0), using the avian retroviral vector, Replication-Competent Avian sarcoma-leukosis virus, with a Splice acceptor, Bryan RSV Pol (RCASBP). High-titre RCASBP retrovirus carrying eGFP was injected into unincubated (stage X) blastoderms in ovo. This resulted in stable and widespread expression of eGFP throughout development in a very high proportion of embryos. Transgenic tissues were identified by fluorescence and immunohistochemistry. These results indicate that chicken blastodermal cells are permissive for infection by the RCASBP virus. This system represents a rapid and efficient method of producing global gene expression in the chicken embryo. The method can be used to generate avian cells with a stable genetic marker, or to induce global expression of a gene of choice. Interestingly, in day 8.5 embryos, somatic cells the embryonic gonads were predominantly GFP positive but primordial germ cells were GFP negative, indicating viral silencing in the embryonic germline. This dichotomy in the gonads allows the isolation or enrichment of the germ cells through negative selection during embryonic stages. This transgenic chicken model is of value in developmental studies, and for the isolation and study of avian primordial germ cells.


Asunto(s)
Pollos/genética , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Retroviridae/genética , Animales , Animales Modificados Genéticamente , Blastodermo/virología , Embrión de Pollo , Embrión no Mamífero
8.
BMC Dev Biol ; 8: 85, 2008 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-18799012

RESUMEN

BACKGROUND: Meiosis in higher vertebrates shows a dramatic sexual dimorphism: germ cells enter meiosis and arrest at prophase I during embryogenesis in females, whereas in males they enter mitotic arrest during embryogenesis and enter meiosis only after birth. Here we report the molecular analysis of meiosis onset in the chicken model and provide evidence for conserved regulation by retinoic acid. RESULTS: Meiosis in the chicken embryo is initiated late in embryogenesis (day 15.5), relative to gonadal sex differentiation (from day 6). Meiotic germ cells are first detectable only in female gonads from day 15.5, correlating with the expression of the meiosis marker, SCP3. Gonads isolated from day 10.5 female embryos and grown in serum-free medium could still initiate meiosis at day 16.5, suggesting that this process is controlled by an endogenous clock in the germ cells themselves, and/or that germ cells are already committed to meiosis at the time of explantation. Early commitment is supported by the analysis of chicken STRA8, a pre-meiotic marker shown to be essential for meiosis in mouse. Chicken STRA8 is expressed female-specifically from embryonic day 12.5, preceding morphological evidence of meiosis at day 15.5. Previous studies have shown that, in the mouse embryo, female-specific induction of STRA8 and meiosis are triggered by retinoic acid. A comprehensive analysis of genes regulating retinoic acid metabolism in chicken embryos reveals dynamic expression in the gonads. In particular, the retinoic acid-synthesising enzyme, RALDH2, is expressed in the left ovarian cortex at the time of STRA8 up-regulation, prior to meiosis. CONCLUSION: This study presents the first molecular analysis of meiosis onset in an avian embryo. Although aspects of avian meiosis differ from that of mammals, a role for retinoic acid may be conserved.


Asunto(s)
Pollos/genética , Meiosis/fisiología , Tretinoina/fisiología , Aldehído Oxidorreductasas/biosíntesis , Aldehído Oxidorreductasas/genética , Animales , Embrión de Pollo , Pollos/metabolismo , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Meiosis/genética , Ratones , Técnicas de Cultivo de Órganos , Ácido Retinoico 4-Hidroxilasa , Tretinoina/metabolismo , Regulación hacia Arriba/fisiología
9.
BMC Dev Biol ; 8: 72, 2008 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-18651984

RESUMEN

BACKGROUND: R-Spondin1 (Rspo1) is a novel regulator of the Wnt/beta-catenin signalling pathway. Loss-of-function mutations in human RSPO1 cause testicular differentiation in 46, XX females, pointing to a role in ovarian development. Here we report the cloning and comparative expression analysis of R-SPONDIN1 orthologues in the mouse, chicken and red-eared slider turtle, three species with different sex-determining mechanisms. Evidence is presented that this gene is an ancient component of the vertebrate ovary-determining pathway. RESULTS: Gonadal RSPO1 gene expression is female up-regulated in the embryonic gonads in each species at the onset of sexual differentiation. In the mouse gonad, Rspo1 mRNA is expressed in the somatic cell lineage at the time of ovarian differentiation (E12.5-E15.5), with little expression in germ cells. However, the protein is localised in the cytoplasm and at the cell surface of both somatic (pre-follicular) and germ cells. In the chicken embryo, RSPO1 expression becomes elevated in females at the time of ovarian differentiation, coinciding with female-specific activation of the FOXL2 gene and estrogen synthesis. RSPO1 protein in chicken is localised in the outer cortical zone of the developing ovary, the site of primordial follicle formation and germ cell differentiation. Inhibition of estrogen synthesis with a specific aromatase inhibitor results in a decline in chicken RSPO1 expression, indicating that RSPO1 is influenced by estrogen. In the red-eared slider turtle, which exhibits temperature-dependent sex determination, up-regulation of RSPO1 occurs during the temperature-sensitive period, when gonadal development is responsive to temperature. Accordingly, RSPO1 expression is temperature-responsive, and is down-regulated in embryos shifted from female- to male-producing incubation temperatures. CONCLUSION: These results indicate that RSPO1 is up-regulated in the embryonic gonads of female vertebrates with different sex-determining mechanisms. In all instances, RSPO1 is expressed in the incipient ovary. These findings suggest that R-SPONDIN1 is an ancient, conserved part of the vertebrate ovary-determining pathway.


Asunto(s)
Ovario/embriología , Procesos de Determinación del Sexo , Trombospondinas/genética , Tortugas/embriología , Secuencia de Aminoácidos , Animales , Inhibidores de la Aromatasa/farmacología , Embrión de Pollo , Clonación Molecular , Embrión de Mamíferos/embriología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Fadrozol/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Ovario/citología , Ovario/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Temperatura , Proteínas Wnt/genética , Proteína Wnt4
10.
Methods Mol Biol ; 1650: 177-190, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28809021

RESUMEN

One of the advantages of the avian embryo as an experimental model is its in ovo development and hence accessibility for genetic manipulation. Electroporation has been used extensively in the past to study gene function in chicken and quail embryos . Readily accessible tissues such as the neural tube, somites, and limb bud, in particular, have been targeted. However, more inaccessible tissues, such as the embryonic urogenital system , have proven more challenging to study. Here, we describe the use of in ovo electroporation of TOL2 vectors or RCASBP avian viral vectors for the rapid functional analysis of genes involved in avian sex determination and urogenital development . In the context of the developing urogenital system , these vectors have inherent advantages and disadvantages, which will be considered here. Either vector can both be used for mis-expressing a gene and for targeting endogenous gene knockdown via expression of short hairpin RNAs (shRNAs). Both of these vectors integrate into the genome and are hence spread throughout developing tissues. Going forward, electroporation could be combined with CRISPR/Cas9 technology for targeted genome editing in the avian urogenital system .


Asunto(s)
Embrión de Pollo , Pollos/genética , Electroporación/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos , Sistema Urogenital/fisiología , Animales , Elementos Transponibles de ADN , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen
11.
Endocrinology ; 158(10): 3684-3695, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977603

RESUMEN

Although it is well established that exogenous androgens have anabolic effects on skeletal muscle mass in humans and mice, data from muscle-specific androgen receptor (AR) knockout (ARKO) mice indicate that myocytic expression of the AR is dispensable for hind-limb muscle mass accrual in males. To identify possible indirect actions of androgens via the AR in neurons to regulate muscle, we generated neuron-ARKO mice in which the dominant DNA binding-dependent actions of the AR are deleted in neurons of the cortex, forebrain, hypothalamus, and olfactory bulb. Serum testosterone and luteinizing hormone levels were elevated twofold in neuron-ARKO males compared with wild-type littermates due to disruption of negative feedback to the hypothalamic-pituitary-gonadal axis. Despite this increase in serum testosterone levels, which was expected to increase muscle mass, the mass of the mixed-fiber gastrocnemius (Gast) and the fast-twitch fiber extensor digitorum longus hind-limb muscles was decreased by 10% in neuron-ARKOs at 12 weeks of age, whereas muscle strength and fatigue of the Gast were unaffected. The mass of the soleus muscle, however, which consists of a high proportion of slow-twitch fibers, was unaffected in neuron-ARKOs, demonstrating a stimulatory action of androgens via the AR in neurons to increase the mass of fast-twitch hind-limb muscles. Furthermore, neuron-ARKOs displayed reductions in voluntary and involuntary physical activity by up to 60%. These data provide evidence for a role of androgens via the AR in neurons to positively regulate fast-twitch hind-limb muscle mass and physical activity in male mice.


Asunto(s)
Encéfalo/metabolismo , Actividad Motora/genética , Músculo Esquelético/anatomía & histología , Neuronas/metabolismo , Condicionamiento Físico Animal , Receptores Androgénicos/genética , Andrógenos , Animales , Western Blotting , Retroalimentación Fisiológica , Genotipo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Noqueados , Fatiga Muscular/genética , Fibras Musculares Esqueléticas , Fuerza Muscular/genética , Tamaño de los Órganos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testosterona/metabolismo
12.
Nat Commun ; 8(1): 132, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28743862

RESUMEN

The ratites are a distinctive clade of flightless birds, typified by the emu and ostrich that have acquired a range of unique anatomical characteristics since diverging from basal Aves at least 100 million years ago. The emu possesses a vestigial wing with a single digit and greatly reduced forelimb musculature. However, the embryological basis of wing reduction and other anatomical changes associated with loss of flight are unclear. Here we report a previously unknown co-option of the cardiac transcription factor Nkx2.5 to the forelimb in the emu embryo, but not in ostrich, or chicken and zebra finch, which have fully developed wings. Nkx2.5 is expressed in emu limb bud mesenchyme and maturing wing muscle, and mis-expression of Nkx2.5 throughout the limb bud in chick results in wing reductions. We propose that Nkx2.5 functions to inhibit early limb bud expansion and later muscle growth during development of the vestigial emu wing.The transcription factor Nkx2.5 is essential for heart development. Here, the authors identify a previously unknown expression domain for Nkx2.5 in the emu wing and explore its role in diminished wing bud development in the flightless emu, compared with three other birds that have functional wings.


Asunto(s)
Proteínas Aviares/genética , Proteína Homeótica Nkx-2.5/genética , Factores de Transcripción/genética , Alas de Animales/metabolismo , Animales , Proteínas Aviares/metabolismo , Dromaiidae , Miembro Anterior/embriología , Miembro Anterior/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Músculo Esquelético/embriología , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alas de Animales/embriología
13.
Evodevo ; 7: 26, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28031782

RESUMEN

BACKGROUND: The forelimb of the flightless emu is a vestigial structure, with greatly reduced wing elements and digit loss. To explore the molecular and cellular mechanisms associated with the evolution of vestigial wings and loss of flight in the emu, key limb patterning genes were examined in developing embryos. METHODS: Limb development was compared in emu versus chicken embryos. Immunostaining for cell proliferation markers was used to analyze growth of the emu forelimb and hindlimb buds. Expression patterns of limb patterning genes were studied, using whole-mount in situ hybridization (for mRNA localization) and RNA-seq (for mRNA expression levels). RESULTS: The forelimb of the emu embryo showed heterochronic development compared to that in the chicken, with the forelimb bud being retarded in its development. Early outgrowth of the emu forelimb bud is characterized by a lower level of cell proliferation compared the hindlimb bud, as assessed by PH3 immunostaining. In contrast, there were no obvious differences in apoptosis in forelimb versus hindlimb buds (cleaved caspase 3 staining). Most key patterning genes were expressed in emu forelimb buds similarly to that observed in the chicken, but with smaller expression domains. However, expression of Sonic Hedgehog (Shh) mRNA, which is central to anterior-posterior axis development, was delayed in the emu forelimb bud relative to other patterning genes. Regulators of Shh expression, Gli3 and HoxD13, also showed altered expression levels in the emu forelimb bud. CONCLUSIONS: These data reveal heterochronic but otherwise normal expression of most patterning genes in the emu vestigial forelimb. Delayed Shh expression may be related to the small and vestigial structure of the emu forelimb bud. However, the genetic mechanism driving retarded emu wing development is likely to rest within the forelimb field of the lateral plate mesoderm, predating the expression of patterning genes.

14.
Genome Biol ; 14(3): R26, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23531366

RESUMEN

BACKGROUND: Birds have a ZZ male: ZW female sex chromosome system and while the Z-linked DMRT1 gene is necessary for testis development, the exact mechanism of sex determination in birds remains unsolved. This is partly due to the poor annotation of the W chromosome, which is speculated to carry a female determinant. Few genes have been mapped to the W and little is known of their expression. RESULTS: We used RNA-seq to produce a comprehensive profile of gene expression in chicken blastoderms and embryonic gonads prior to sexual differentiation. We found robust sexually dimorphic gene expression in both tissues pre-dating gonadogenesis, including sex-linked and autosomal genes. This supports the hypothesis that sexual differentiation at the molecular level is at least partly cell autonomous in birds. Different sets of genes were sexually dimorphic in the two tissues, indicating that molecular sexual differentiation is tissue specific. Further analyses allowed the assembly of full-length transcripts for 26 W chromosome genes, providing a view of the W transcriptome in embryonic tissues. This is the first extensive analysis of W-linked genes and their expression profiles in early avian embryos. CONCLUSION: Sexual differentiation at the molecular level is established in chicken early in embryogenesis, before gonadal sex differentiation. We find that the W chromosome is more transcriptionally active than previously thought, expand the number of known genes to 26 and present complete coding sequences for these W genes. This includes two novel W-linked sequences and three small RNAs reassigned to the W from the Un_Random chromosome.


Asunto(s)
Regulación de la Expresión Génica , Gónadas/embriología , Anotación de Secuencia Molecular , Análisis de Secuencia de ARN/métodos , Caracteres Sexuales , Cromosomas Sexuales/genética , Diferenciación Sexual/genética , Animales , Blastodermo/metabolismo , Pollos/genética , Secuencia Conservada , Femenino , Perfilación de la Expresión Génica , Ligamiento Genético , Gónadas/metabolismo , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Int J Dev Biol ; 53(1): 59-67, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19123127

RESUMEN

Birds have a ZZ/ZW sex chromosome system, but the mechanism of sex determination remains unknown. The heterogametic sex is female (ZW) and one hypothesis holds that the W chromosome carries a dominant-acting ovary-determining gene. The strongest candidate ovary-determinant on the W chromosome is HINTW, which encodes an aberrant nucleotide hydrolase enzyme. HINTW is conserved amongst all carinate (flying) birds and it is strongly expressed in the gonads and other tissues of female chicken embryos. This and other lines of circumstantial evidence support the proposal that HINTW is the female-determining gene in birds. However, in vivo gain-of-function or loss-of-function studies have not hitherto been reported to test this hypothesis. We tested the potential role of HINTW by mis-expressing it in genetically male (ZZ) embryos, using the RCASBP avian retroviral vector. Strong, widespread expression was delivered throughout the embryo, including the urogenital system, as assessed by whole mount in situ hybridisation. This expression pattern mimicked that seen in normal ZW females, in which HINTW is widely expressed. Strong mis-expression was observed throughout the gonads of genetic male (ZZ) embryos. However, despite strong HINTW expression, ZZ gonads developed normally as bilateral testes. In tissue sections of ZZ urogenital systems transgenic for HINTW, normal testicular histology was observed. Female (ZW) gonads over-expressing HINTW also developed normally, with normal ovarian structure and left/right asymmetry. These results provide genetic evidence against a dominant role for HINTW in avian sex determination.


Asunto(s)
Hidrolasas/metabolismo , Cromosomas Sexuales/genética , Procesos de Determinación del Sexo , Animales , Biomarcadores/metabolismo , Línea Celular , Embrión de Pollo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros/genética , Gónadas/embriología , Gónadas/metabolismo , Hidrolasas/genética , Masculino
16.
Dev Dyn ; 238(8): 2073-80, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19618468

RESUMEN

Balanced production and degradation of retinoids is important in regulating development of several organ systems in the vertebrate embryo. Among these, it is known that retinoic acid (RA), and the retinoid-catabolyzing enzyme CYP26B1 together regulate the sex-specific behavior of germ cells in developing mouse gonads. We report here that the gene encoding a cytosolic class-1 aldehyde dehydrogenase, ALDH1A1, a weak catalyst of RA production, is strongly expressed in a male-specific manner in somatic cells of the developing mouse testis, beginning shortly after Sry expression is first detectable. This expression pattern is conserved in the developing male gonad of the chicken and is dependent on the testis-specific transcription factor SOX9. Our data suggest that low levels of RA may be required for early developmental events in the testis, or that Aldh1a1 expression in the fetus may prefigure a later requirement for ALDH1A1 in regulating spermatogenesis postnatally.


Asunto(s)
Aldehído Deshidrogenasa/genética , Isoenzimas/genética , Retinoides/metabolismo , Testículo/embriología , Testículo/enzimología , Aldehído Deshidrogenasa/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Secuencia de Bases , Embrión de Pollo , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes sry , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Embarazo , Retinal-Deshidrogenasa , Factor de Transcripción SOX9/metabolismo , Diferenciación Sexual , Testículo/metabolismo , Tretinoina/metabolismo
17.
Mol Reprod Dev ; 74(5): 531-8, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17290426

RESUMEN

Cell cycle progression is prevented by signal transduction pathways known as checkpoints which are activated in response to replication interference and DNA damage. We cloned a G2/M cell cycle phase-related checkpoint gene from a neonatal mouse testis cDNA library which was identified as mouse claspin, a proposed adaptor protein for Chk1. As part of a study on germ cell differentiation we examined the expression of the checkpoint gene, Chk1, and claspin at 12.5 and 14.5 days post coitum (dpc) and in the post-natal phase. Chk1 mRNA expression increased from 12.5 to 14.5 dpc in female gonads and was strong in males at both time points. Claspin however, was not detected until 14.5 dpc. This suggests there may be some dissociation of claspin expression from Chk1 in fetal germ cell development. Chk1 and claspin expression was also studied in testis over the first 3 days following birth, when apoptosis regulates germ stem cell number. We modulated checkpoint-related gene expression in testis using the anti-metabolite, 5-fluorouracil, resulting in increased apoptosis and upregulation of Chk1 (P<0.0001) and Cdc2 (P<0.02) mRNA. Although we do not fully understand the role checkpoint gene expression has during mammalian germ cell development this report is the first to show the expression of checkpoint-related genes in early mammalian germ cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fase G2 , Células Germinativas/citología , Mitosis , Proteínas Quinasas/metabolismo , Animales , Animales Recién Nacidos , Antimetabolitos/farmacología , Apoptosis , Proteína Quinasa CDC2/metabolismo , Diferenciación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Femenino , Fluorouracilo/farmacología , Biblioteca de Genes , Células Germinativas/metabolismo , Masculino , Ratones , Embarazo , Factores Sexuales , Testículo/citología , Testículo/embriología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Regulación hacia Arriba
18.
Stem Cells ; 24(11): 2382-90, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16857898

RESUMEN

Recently, we described a rare cell type within the adult murine pituitary gland with progenitor cell hallmarks (PCFCs). PCFCs are contained exclusively within a subpopulation of cells that import fluorescent beta-Ala-Lys-Nepsilon-AMCA (7-amino-4-methylcoumarin-3-acetic acid). Herein, we investigate the utility of cell surface molecules angiotensin-converting enzyme (ACE) and stem cell antigen-1 (Sca-1) to further enrich for PCFCs. ACE and Sca-1 were expressed on 61% and 55% of AMCA(+)CD45(-)CD31(-) cells, respectively, and coexpressed on 38%. ACE(+)Sca-1(+)AMCA(+) cells enriched for PCFCs by 195-fold over unselected cells. ACE(+)AMCA(+) cells enriched for PCFCs by 170-fold, and colonies were twofold larger than for AMCA(+) selection alone. Conversely, ACE(-)-selected cells reduced both colony-forming activity and size. Notably, colonies generated from AMCA(+) cells obtained from ACE(null) mice were 2.7-fold smaller than for wild-type mice. These data identify ACE as a previously unrecognized marker of PCFCs and suggest that ACE is functionally important for PCFC proliferation. Anatomically, the cells that imported AMCA and expressed ACE were situated in the marginal epithelial cell layer of the pituitary cleft and in the adjacent subluminal zone, thus supporting previous proposals that the luminal zone is a source of precursor cells in the adult pituitary.


Asunto(s)
Células Madre Adultas/metabolismo , Proliferación Celular , Peptidil-Dipeptidasa A/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , Células Madre Adultas/enzimología , Animales , Antígenos Ly/análisis , Separación Celular/métodos , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Cumarinas/metabolismo , Dipéptidos/metabolismo , Femenino , Citometría de Flujo , Colorantes Fluorescentes/metabolismo , Proteínas de la Membrana/análisis , Ratones , Ratones Noqueados , Microscopía Fluorescente , Peptidil-Dipeptidasa A/genética , Hipófisis/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA