Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Reproduction ; 168(2)2024 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-38833564

RESUMO

In brief: Atrazine, like oestrogen, disorganises laminin formation and reduces the number of germ cells and Sertoli cells in the developing testes of the tammar wallaby. This study suggests that interfering with the balance of androgen and oestrogen affects the integrity of laminin structure and testis differentiation. Abstract: The herbicide atrazine was banned in Europe in 2003 due to its endocrine disrupting activity but remains widely used. The integrity of the laminin structure in fetal testis cords requires oestrogen signalling but overexposure to xenoestrogens in the adult can cause testicular dysgenesis. However, whether xenoestrogens affect laminin formation in developing testes has not been investigated. Here we examined the effects of atrazine in the marsupial tammar wallaby during early development and compare it with the effects of the anti-androgen flutamide, oestrogen, and the oestrogen degrader fulvestrant. The tammar, like all marsupials, gives birth to altricial young, allowing direct treatment of the developing young during the male programming window (day 20-40 post partum (pp)). Male pouch young were treated orally with atrazine (5 mg/kg), flutamide (10 mg/kg), 17ß-oestradiol (2.5 mg/kg) and fulvestrant (1 mg/kg) daily from day 20 to 40 pp. Distribution of laminin, vimentin, SOX9 and DDX4, cell proliferation and mRNA expression of SRY, SOX9, AMH, and SF1 were examined in testes at day 50 post partum after the treatment. Direct exposure to atrazine, flutamide, 17ß-oestradiol, and fulvestrant all disorganised laminin but had no effect on vimentin distribution in testes. Atrazine reduced the number of germ cells and Sertoli cells when examined at day 40-50 pp and day 20 to 40 pp, respectively. Both flutamide and fulvestrant reduced the number of germ cells and Sertoli cells. Atrazine also downregulated SRY expression and impaired SOX9 nuclear translocation. Our results demonstrate that atrazine can compromise normal testicular differentiation during the critical male programming window.


Assuntos
Atrazina , Diferenciação Celular , Herbicidas , Laminina , Testículo , Masculino , Animais , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/citologia , Atrazina/farmacologia , Laminina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Herbicidas/farmacologia , Macropodidae/metabolismo , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Células de Sertoli/citologia , Estrogênios/farmacologia , Estrogênios/metabolismo , Disruptores Endócrinos/farmacologia , Contagem de Células , Antagonistas de Androgênios/farmacologia , Flutamida/farmacologia
2.
Nat Commun ; 15(1): 3953, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729967

RESUMO

Efficient milk production in mammals confers evolutionary advantages by facilitating the transmission of energy from mother to offspring. However, the regulatory mechanism responsible for the gradual establishment of milk production efficiency in mammals, from marsupials to eutherians, remains elusive. Here, we find that mammary gland of the marsupial sugar glider contained milk components during adolescence, and that mammary gland development is less dynamically cyclic compared to that in placental mammals. Furthermore, fused in sarcoma (FUS) is found to be partially responsible for this establishment of low efficiency. In mouse model, FUS inhibit mammary epithelial cell differentiation through the cyclin-dependent kinase inhibitor p57Kip2, leading to lactation failure and pup starvation. Clinically, FUS levels are negatively correlated with milk production in lactating women. Overall, our results shed light on FUS as a negative regulator of milk production, providing a potential mechanism for the establishment of milk production from marsupial to eutherian mammals.


Assuntos
Lactação , Glândulas Mamárias Animais , Leite , Animais , Feminino , Glândulas Mamárias Animais/metabolismo , Humanos , Camundongos , Leite/metabolismo , Diferenciação Celular , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/genética , Células Epiteliais/metabolismo , Macropodidae/metabolismo , Mamíferos , Marsupiais
3.
Genes (Basel) ; 11(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31963388

RESUMO

Congenital anomalies in phalluses caused by endocrine disruptors have gained a great deal of attention due to its annual increasing rate in males. However, the endocrine-driven molecular regulatory mechanism of abnormal phallus development is complex and remains largely unknown. Here, we review the direct effect of androgen and oestrogen on molecular regulation in phalluses using the marsupial tammar wallaby, whose phallus differentiation occurs after birth. We summarize and discuss the molecular mechanisms underlying phallus differentiation mediated by sonic hedgehog (SHH) at day 50 pp and phallus elongation mediated by insulin-like growth factor 1 (IGF1) and insulin-like growth factor binding protein 3 (IGFBP3), as well as multiple phallus-regulating genes expressed after day 50 pp. We also identify hormone-responsive long non-coding RNAs (lncRNAs) that are co-expressed with their neighboring coding genes. We show that the activation of SHH and IGF1, mediated by balanced androgen receptor (AR) and estrogen receptor 1 (ESR1) signalling, initiates a complex regulatory network in males to constrain the timing of phallus differentiation and to activate the downstream genes that maintain urethral closure and phallus elongation at later stages.


Assuntos
Macropodidae/crescimento & desenvolvimento , Pênis/crescimento & desenvolvimento , Diferenciação Sexual/efeitos dos fármacos , Androgênios/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Disruptores Endócrinos , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Genitália Feminina , Genitália Masculina/crescimento & desenvolvimento , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Macropodidae/metabolismo , Masculino , Pênis/metabolismo , Receptores Androgênicos/metabolismo
4.
Biol Reprod ; 99(4): 806-816, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29767687

RESUMO

Environmental endocrine disruptors (EEDs) that affect androgen or estrogen activity may disrupt gene regulation during phallus development to cause hypospadias or a masculinized clitoris. We treated developing male tammar wallabies with estrogen and females with androgen from day 20-40 postpartum (pp) during the androgen imprinting window of sensitivity. Estrogen inhibited phallus elongation but had no effect on urethral closure and did not significantly depress testicular androgen synthesis. Androgen treatment in females did not promote phallus elongation but initiated urethral closure. Phalluses were collected for transcriptome sequencing at day 50 pp when they first become sexually dimorphic to examine changes in two signaling pathways, sonic hedgehog (SHH) and wingless-type MMTV integration site family (WNT)/ß-catenin. SHH mRNA and ß-catenin were predominantly expressed in the urethral epithelium in the tammar phallus, as in eutherian mammals. Estrogen treatment and castration of males induced an upregulation of SHH, while androgen treatment downregulated SHH. These effects appear to be direct since we detected putative estrogen receptor α (ERα) and androgen receptor (AR) binding sites near SHH. WNT5A, like SHH, was downregulated by androgen, while WNT4 was upregulated in female phalluses after androgen treatment. After estrogen treatment, WIF1 and WNT7A were both downregulated in male phalluses. After castration, WNT9A was upregulated. These results suggest that SHH and WNT pathways are regulated by both estrogen and androgen to direct the proliferation and elongation of the phallus during differentiation. Their response to exogenous hormones makes these genes potential targets of EEDs in the etiology of abnormal phallus development including hypospadias.


Assuntos
Macropodidae/crescimento & desenvolvimento , Macropodidae/genética , Pênis/crescimento & desenvolvimento , Pênis/metabolismo , Transdução de Sinais/genética , Uretra/crescimento & desenvolvimento , Uretra/metabolismo , Androgênios/metabolismo , Animais , Disruptores Endócrinos/toxicidade , Estrogênios/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/crescimento & desenvolvimento , Genitália Feminina/metabolismo , Genitália Masculina/efeitos dos fármacos , Genitália Masculina/crescimento & desenvolvimento , Genitália Masculina/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Macropodidae/metabolismo , Masculino , Pênis/efeitos dos fármacos , Diferenciação Sexual/efeitos dos fármacos , Diferenciação Sexual/genética , Diferenciação Sexual/fisiologia , Transdução de Sinais/efeitos dos fármacos , Uretra/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Sci Rep ; 7(1): 16887, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203823

RESUMO

Organotypic co-cultures bridge the gap between standard two-dimensional culture and mouse models. Such assays increase the fidelity of pre-clinical studies, to better inform lead compound development and address the increasing attrition rates of lead compounds within the pharmaceutical industry, which are often a result of screening in less faithful two-dimensional models. Using large-scale acid-extraction techniques, we demonstrate a step-by-step process to isolate collagen I from commercially available animal byproducts. Using the well-established rat tail tendon collagen as a benchmark, we apply our novel kangaroo tail tendon collagen as an alternative collagen source for our screening-ready three-dimensional organotypic co-culture platform. Both collagen sources showed equal applicability for invasive, proliferative or survival assessment of well-established cancer models and clinically relevant patient-derived cancer cell lines. Additional readouts were also demonstrated when comparing these alternative collagen sources for stromal contributions to stiffness, organization and ultrastructure via atomic force microscopy, second harmonic generation imaging and scanning electron microscopy, among other vital biological readouts, where only minor differences were found between the preparations. Organotypic co-cultures represent an easy, affordable and scalable model to investigate drug responses within a physiologically relevant 3D platform.


Assuntos
Técnicas de Cultura de Células/métodos , Colágeno/química , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Colágeno/isolamento & purificação , Matriz Extracelular/metabolismo , Gefitinibe/farmacologia , Humanos , Macropodidae/metabolismo , Camundongos , Microscopia de Força Atômica , Ratos , Tendões/metabolismo
6.
Biol Reprod ; 97(2): 217-229, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-29044428

RESUMO

The first sign of mammalian germ cell sexual differentiation is the initiation of meiosis in females and of mitotic arrest in males. In the mouse, retinoic acid induces ovarian Stra8 expression and entry of germ cells into meiosis. In developing mouse testes, cytochrome P450 family 26, subfamily b, polypeptide 1 (CYP26B1) produced by the Sertoli cells degrades retinoic acid, preventing Stimulated by Retinoic Acid Gene 8 (Stra8), expression and inhibiting meiosis. However, in developing humans, there is no evidence that CYP26B1 acts a meiosis-inhibiting factor. We therefore examined aspects of the retinoic acid/STRA8/CYP26B1 pathway during gonadal development in the tammar wallaby, a marsupial, to understand whether retinoic acid stimulation of STRA8 and CYP26B1 degradation of retinoic acid was conserved between widely divergent mammals. In tammar ovaries, as in human ovaries and unlike the pattern in mice, CYP26B1 expression was not downregulated before the onset of meiosis. Exposure of pre-meiotic tammar ovaries to exogenous retinoic acid in vitro upregulated STRA8 expression compared to controls. We conclude that retinoic acid and STRA8 are conserved factors that control the initiation of meiosis amongst mammals but the role of CYP26B1 as a meiosis-inhibiting factor may be specific to rodents. The identity of the marsupial meiosis-inhibiting factor remains unknown.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Macropodidae/metabolismo , Ácido Retinoico 4 Hidroxilase/metabolismo , Processos de Determinação Sexual/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Meiose , Camundongos , Oogênese/fisiologia , Ácido Retinoico 4 Hidroxilase/genética , Especificidade da Espécie , Espermatogênese/fisiologia , Tretinoína/metabolismo
7.
Mol Reprod Dev ; 84(10): 1076-1085, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28688214

RESUMO

Pregnancy in mammals requires remodeling of the uterus to become receptive to the implanting embryo. Remarkably similar morphological changes to the uterine epithelium occur in both eutherian and marsupial mammals, irrespective of placental type. Nevertheless, molecular differences in uterine remodeling indicate that the marsupial uterus employs maternal defences, including molecular reinforcement of the uterine epithelium, to regulate embryonic invasion. Non-invasive (epitheliochorial) embryonic attachment in marsupials likely evolved secondarily from invasive attachment, so uterine defences in these species may prevent embryonic invasion. We tested this hypothesis by identifying localization patterns of Talin, a key basal anchoring molecule, in the uterine epithelium during pregnancy in the tammar wallaby (Macropus eugenii; Macropodidae) and the brush tail possum (Trichosurus vulpecula; Phalangeridae). Embryonic attachment is non-invasive in both species, yet Talin undergoes a clear distributional change during pregnancy in M. eugenii, including recruitment to the base of the uterine epithelium just before attachment, that closely resembles that of invasive implantation in the marsupial species Sminthopsis crassicaudata. Basal localization occurs throughout pregnancy in T. vulpecula, although, as for M. eugenii, this pattern is most specific prior to attachment. Such molecular reinforcement of the uterine epithelium for non-invasive embryonic attachment in marsupials supports the hypothesis that less-invasive and non-invasive embryonic attachment in marsupials may have evolved via accrual of maternal defences. Recruitment of basal molecules, including Talin, to the uterine epithelium may have played a key role in this transition.


Assuntos
Implantação do Embrião/fisiologia , Macropodidae/fisiologia , Prenhez , Trichosurus/fisiologia , Útero/metabolismo , Animais , Células Epiteliais/metabolismo , Feminino , Macropodidae/metabolismo , Phalangeridae/metabolismo , Phalangeridae/fisiologia , Gravidez , Ratos , Talina/metabolismo , Trichosurus/metabolismo , Trofoblastos/metabolismo , Útero/citologia , Útero/fisiologia
8.
J Vet Pharmacol Ther ; 40(6): 682-686, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28568310

RESUMO

This study was designed to investigate the pharmacokinetics of clindamycin, a lincosamide antibiotic, in Bennett's wallabies. The pharmacokinetic properties of a single intravenous (IV) dose of clindamycin were determined in six wallabies. A single 20-min IV infusion of 20 mg/kg of clindamycin was administered, followed by blood collection prior to, and up to 12 hr after clindamycin administration. Plasma clindamycin concentrations were determined by high-pressure liquid chromatography (HPLC) with ultraviolet (UV) detection. Pharmacokinetic variables were calculated using a two-compartment model with first order elimination which best fit the data. The mean volume of distribution at steady-state, distribution half-life, and elimination half-life were 898.25 ml/kg, 0.16 hr, 1.79 hr, respectively. No adverse effects were noted after IV administration.


Assuntos
Antibacterianos/farmacocinética , Clindamicina/análogos & derivados , Macropodidae/metabolismo , Animais , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Cromatografia Líquida de Alta Pressão/veterinária , Clindamicina/administração & dosagem , Clindamicina/sangue , Clindamicina/farmacocinética , Feminino , Meia-Vida , Infusões Intravenosas/veterinária , Masculino
9.
Reproduction ; 152(5): 491-505, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27486272

RESUMO

The marsupial tammar wallaby has the longest period of embryonic diapause of any mammal, up to 11 months, during which there is no cell division or blastocyst growth. Since the blastocyst in diapause is surrounded by acellular coats, the signals that maintain or terminate diapause involve factors that reside in uterine secretions. The nature of such factors remains to be resolved. In this study, uterine flushings (UFs) were used to assess changes in uterine secretions of tammars using liquid chromatography-mass spectrometry (LC-MS/MS) during diapause (day 0 and 3) and reactivation days (d) 4, 5, 6, 8, 9, 11 and 24 after removal of pouch young (RPY), which initiates embryonic development. This study supports earlier suggestions that the presence of specific factors stimulate reactivation, early embryonic growth and cell proliferation. A mitogen, hepatoma-derived growth factor and soluble epidermal growth factor receptors were observed from d3 until at least d11 RPY when these secreted proteins constituted 21% of the UF proteome. Binding of these factors to specific cellular receptors or growth factors may directly stimulate DNA synthesis and division in endometrial gland cells. Proteins involved in the p53/CDKN1A (p21) cell cycle inhibition pathway were also observed in the diapause samples. Progesterone and most of the oestrogen-regulated proteins were present in the UF after d3, which is concomitant with the start of blastocyst mitoses at d4. We propose that once the p21 inhibition of the cell cycle is lost, growth factors including HDGF and EGFR are responsible for reactivation of the diapausing blastocyst via the uterine secretions.


Assuntos
Blastocisto/metabolismo , Implantação Tardia do Embrião/fisiologia , Desenvolvimento Embrionário , Macropodidae/metabolismo , Metamorfose Biológica/fisiologia , Proteoma/metabolismo , Útero/metabolismo , Animais , Blastocisto/citologia , Endométrio/crescimento & desenvolvimento , Endométrio/metabolismo , Feminino , Macropodidae/crescimento & desenvolvimento , Gravidez , Espectrometria de Massas em Tandem , Útero/crescimento & desenvolvimento
10.
Int J Dev Biol ; 58(2-4): 175-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25023683

RESUMO

The marsupial tammar wallaby has the longest period of embryonic diapause of any mammal. Reproduction in the tammar is seasonal, regulated by photoperiod and also lactation. Reactivation is triggered by falling daylength after the austral summer solstice in December. Young are born late January and commence a 9-10-month lactation. Females mate immediately after birth. The resulting conceptus develops over 6- 7 days to form a unilaminar blastocyst of 80-100 cells and enters lactationally, and later seasonally, controlled diapause. The proximate endocrine signal for reactivation is an increase in progesterone which alters uterine secretions. Since the diapausing blastocyst is surrounded by the zona and 2 other acellular coats, the mucoid layer and shell coat, the uterine signals that maintain or terminate diapause must involve soluble factors in the secretions rather than any direct cellular interaction between uterus and embryo. Our studies suggest involvement of a number of cytokines in the regulation of diapause in tammars. The endometrium secretes platelet activating factor (PAF) and leukaemia inhibitory factor, which increase after reactivation. Receptors for PAF are low on the blastocyst during diapause but are upregulated at reactivation. Conversely, there is endometrial expression of the muscle segment homeobox gene MSX2 throughout diapause, but it is rapidly downregulated at reactivation. These patterns are consistent with those observed in diapausing mice and mink after reactivation, despite the very different patterns of endocrine control of diapause in these 3 divergent species. These common patterns suggest a similar underlying mechanism for diapause, perhaps common to all mammals, but which is activated in only a few.


Assuntos
Implantação Tardia do Embrião/fisiologia , Embrião de Mamíferos/metabolismo , Endométrio/metabolismo , Macropodidae/embriologia , Animais , Feminino , Humanos , Macropodidae/metabolismo , Camundongos
11.
Reproduction ; 147(1): 21-31, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24123130

RESUMO

The control of reactivation from embryonic diapause in the tammar wallaby (Macropus eugenii) involves sequential activation of the corpus luteum, secretion of progesterone that stimulates endometrial secretion and subsequent changes in the uterine environment that activate the embryo. However, the precise signals between the endometrium and the blastocyst are currently unknown. In eutherians, both the phospholipid Paf and its receptor, platelet-activating factor receptor (PTAFR), are present in the embryo and the endometrium. In the tammar, endometrial Paf release in vitro increases around the time of the early progesterone pulse that occurs around the time of reactivation, but whether Paf can reactivate the blastocyst is unknown. We cloned and characterised the expression of PTAFR in the tammar embryo and endometrium at entry into embryonic diapause, during its maintenance and after reactivation. Tammar PTAFR sequence and protein were highly conserved with mammalian orthologues. In the endometrium, PTAFR was expressed at a constant level in the glandular epithelium across all stages and in the luminal epithelium during both diapause and reactivation. Thus, the presence of the receptor appears not to be a limiting factor for Paf actions in the endometrium. However, the low levels of PTAFR in the embryo during diapause, together with its up-regulation and subsequent internalisation at reactivation, supports earlier results suggesting that endometrial Paf could be involved in reactivation of the tammar blastocyst from embryonic diapause.


Assuntos
Endométrio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Macropodidae/embriologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Corpo Lúteo/metabolismo , Feminino , Macropodidae/metabolismo , Gravidez , Progesterona/metabolismo , Útero/metabolismo
12.
Matrix Biol ; 32(6): 342-51, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23665481

RESUMO

Asynchronous concurrent lactation (ACL) is an extreme lactation strategy in macropod marsupials including the tammar wallaby, that may hold the key to understanding local control of mammary epithelial cell function. Marsupials have a short gestation and a long lactation consisting of three phases; P2A, P2B and P3, representing early, mid and late lactation respectively and characterised by profound changes in milk composition. A lactating tammar is able to concurrently produce phase 2A and 3 milk from adjacent glands in order to feed a young newborn and an older sibling at heel. Physiological effectors of ACL remain unknown and in this study the extracellular matrix (ECM) is investigated for its role in switching mammary phenotypes between phases of tammar wallaby lactation. Using the level of expression of the genes for the phase specific markers tELP, tWAP, and tLLP-B representing phases 2A, 2B and 3 respectively we show for the first time that tammar wallaby mammary epithelial cells (WallMECs) extracted from P2B acquire P3 phenotype when cultured on P3 ECM. Similarly P2A cells acquire P2B phenotype when cultured on P2B ECM. We further demonstrate that changes in phase phenotype correlate with phase-specific changes in ECM composition. This study shows that progressive changes in ECM composition in individual mammary glands provide a local regulatory mechanism for milk protein gene expression thereby enabling the mammary glands to lactate independently.


Assuntos
Células Epiteliais/metabolismo , Matriz Extracelular/genética , Lactação/genética , Macropodidae/genética , Glândulas Mamárias Animais/metabolismo , Proteínas do Leite/genética , Fatores Etários , Animais , Biomarcadores/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Epiteliais/ultraestrutura , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Feminino , Regulação da Expressão Gênica , Genótipo , Lactação/metabolismo , Macropodidae/metabolismo , Glândulas Mamárias Animais/ultraestrutura , Proteínas do Leite/metabolismo , Fenótipo , Inibidores de Proteases/metabolismo , Transdução de Sinais , Proteínas do Soro do Leite
13.
Reprod Fertil Dev ; 25(2): 456-61, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22951148

RESUMO

The corpus luteum (CL) of the tammar wallaby is inhibited by prolactin during lactation and seasonal quiescence. In seasonal quiescence a daily transient pulse of prolactin (PRL) of less than 2h duration is sufficient to maintain inhibition. We investigated whether the same inhibition applies in lactation and, if so, how. Our results show that inhibition of the CL during lactation is maintained by a transient pulse of prolactin once a day. They also show that the minimum time without a PRL pulse for the CL to escape inhibition is more than 48 h and less than 72 h. Nevertheless, some animals had a longer refractory period than 72 h, which was reflected in a longer interval to the progesterone peak and birth. These results support the previous conclusion that PRL exercises its effect on a rate-limiting step in progesterone synthesis and secretion rate from the CL, which precedes any increase in its mass. Therefore, we conclude that the role of PRL is to act as a luteostatic agent, an effect that is in marked contrast to its luteotrophic effect in many eutherian species, including rodents.


Assuntos
Corpo Lúteo/metabolismo , Lactação/fisiologia , Macropodidae/fisiologia , Prolactina/metabolismo , Animais , Território da Capital Australiana , Feminino , Modelos Lineares , Macropodidae/metabolismo , Progesterona/biossíntese , Prolactina/sangue , Radioimunoensaio , Estações do Ano , Especificidade da Espécie , Fatores de Tempo
14.
Biol Reprod ; 84(3): 595-603, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21123819

RESUMO

Kallmann syndrome is characterized by hypogonadotrophic hypogonadism and anosmia. The syndrome can be caused by mutations in several genes, but the X-linked form is caused by mutation in the Kallmann syndrome 1 (KAL1). KAL1 plays a critical role in gonadotropin-releasing hormone (GnRH) neuronal migration that is essential for the normal development of the hypothalamic-pituitary-gonadal axis. Interestingly, KAL1 appears to be missing from the rodent X, and no orthologue has been detected as yet. We investigated KAL1 during development and in adults of an Australian marsupial, the tammar wallaby, Macropus eugenii. Marsupial KAL1 maps to an autosome within a group of genes that was added as a block to the X chromosome in eutherian evolution. KAL1 expression was widespread in embryonic and adult tissues. In the adult testis, tammar KAL1 mRNA and protein were detected in the germ cells at specific stages of differentiation. In the adult testis, the protein encoded by KAL1, anosmin-1, was restricted to the round spermatids and elongated spermatids. In the adult ovary, anosmin-1 was not only detected in the oocytes but was also localized in the granulosa cells throughout folliculogenesis. This is the first examination of KAL1 mRNA and protein localization in adult mammalian gonads. The protein localization suggests that KAL1 participates in gametogenesis not only through the development of the hypothalamic-pituitary-gonadal axis by activation of GnRH neuronal migration, but also directly within the gonads themselves. Because KAL1 is autosomal in marsupials but is X-linked in eutherians, its conserved involvement in gametogenesis supports the hypothesis that reproduction-related genes were actively recruited to the eutherian X chromosome.


Assuntos
Gônadas/metabolismo , Síndrome de Kallmann/genética , Marsupiais/genética , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Clonagem Molecular , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Expressão Gênica , Gônadas/embriologia , Síndrome de Kallmann/metabolismo , Macropodidae/embriologia , Macropodidae/genética , Macropodidae/metabolismo , Masculino , Marsupiais/embriologia , Marsupiais/metabolismo , Camundongos , Dados de Sequência Molecular , Organogênese/genética , Filogenia , Homologia de Sequência
15.
BMC Biol ; 8: 113, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20807406

RESUMO

BACKGROUND: Hormones are critical for early gonadal development in nonmammalian vertebrates, and oestrogen is required for normal ovarian development. In contrast, mammals determine sex by the presence or absence of the SRY gene, and hormones are not thought to play a role in early gonadal development. Despite an XY sex-determining system in marsupial mammals, exposure to oestrogen can override SRY and induce ovarian development of XY gonads if administered early enough. Here we assess the effect of exogenous oestrogen on the molecular pathways of mammalian gonadal development. RESULTS: We examined the expression of key testicular (SRY, SOX9, AMH and FGF9) and ovarian (WNT4, RSPO1, FOXL2 and FST) markers during gonadal development in the marsupial tammar wallaby (Macropus eugenii) and used these data to determine the effect of oestrogen exposure on gonadal fate. During normal development, we observed male specific upregulation of AMH and SOX9 as in the mouse and human testis, but this upregulation was initiated before the peak in SRY expression and 4 days before testicular cord formation. Similarly, key genes for ovarian development in mouse and human were also upregulated during ovarian differentiation in the tammar. In particular, there was early sexually dimorphic expression of FOXL2 and WNT4, suggesting that these genes are key regulators of ovarian development in all therian mammals. We next examined the effect of exogenous oestrogen on the development of the mammalian XY gonad. Despite the presence of SRY, exogenous oestrogen blocked the key male transcription factor SOX9 from entering the nuclei of male somatic cells, preventing activation of the testicular pathway and permitting upregulation of key female genes, resulting in ovarian development of the XY gonad. CONCLUSIONS: We have uncovered a mechanism by which oestrogen can regulate gonadal development through the nucleocytoplasmic shuttling of SOX9. This may represent an underlying ancestral mechanism by which oestrogen promotes ovarian development in the gonads of nonmammalian vertebrates. Furthermore, oestrogen may retain this function in adult female mammals to maintain granulosa cell fate in the differentiated ovary by suppressing nuclear translocation of the SOX9 protein. See commentary: http://www.biomedcentral.com/1741-7007/8/110.


Assuntos
Estrogênios/metabolismo , Gônadas/crescimento & desenvolvimento , Gônadas/metabolismo , Macropodidae/crescimento & desenvolvimento , Fatores de Transcrição SOX9/metabolismo , Animais , Feminino , Macropodidae/metabolismo , Masculino , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
16.
BMC Biol ; 8: 110, 2010 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-20828373

RESUMO

Oestrogen exerts a robust yet imperfectly understood effect on sexual development in vertebrate embryos. New work by Pask and colleagues in BMC Biology indicates that it may interfere with male development by preventing nuclear localization of SOX9, a master regulator of the testis differentiation pathway. See research article http://www.biomedcentral.com/1741-7007/8/113.


Assuntos
Estrogênios/metabolismo , Gônadas/crescimento & desenvolvimento , Macropodidae/crescimento & desenvolvimento , Fatores de Transcrição SOX9/metabolismo , Animais , Gônadas/metabolismo , Macropodidae/metabolismo , Masculino
17.
Reproduction ; 139(3): 599-611, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028801

RESUMO

Oestrogen has wide ranging effects in development mediated mainly via the two oestrogen receptors, alpha (ESR1, also known as ERalpha) and beta (ESR2, also known as ERbeta). Oestrogen is the key factor that directs the indifferent gonad to become an ovary in many non-mammalian vertebrates. Oestrogen is not required for early ovarian differentiation in mammals but can disrupt normal testicular development in eutherians. Surprisingly, exogenous oestrogen can cause sex reversal of an XY gonad in two marsupials, the North American opossum and the tammar wallaby. To understand the mechanism by which oestrogen induces sex reversal, we characterised the genes for ESR1 and ESR2 and examined their expression during gonadal differentiation in the tammar wallaby, Macropus eugenii. Both receptors were expressed in the somatic cells and germ cells of the indifferent gonad in both XX and XY foetuses throughout all stages of development, and persisted in these cells into adulthood. ERs were also present in many other tissues including kidney, pituitary and mammary gland. ER mRNA was not significantly altered by exogenous oestrogen in cultured XY gonads but the receptors translocated to the nucleus in its presence. These findings confirm that there is conserved expression of the ERs in the indifferent gonad despite the lack of available ligand during early gonadal development. The receptors can respond to exogenous estrogen at this early stage and are capable of transducing signals in the early mammalian gonad. However, the selective forces that maintained conserved ER expression in this tissue remain unknown.


Assuntos
Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Gônadas/embriologia , Gônadas/metabolismo , Macropodidae , Sequência de Aminoácidos , Animais , Células Cultivadas , Clonagem Molecular , Embrião de Mamíferos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Macropodidae/embriologia , Macropodidae/genética , Macropodidae/metabolismo , Masculino , Dados de Sequência Molecular , Gravidez , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
18.
Evol Dev ; 11(4): 363-75, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19601970

RESUMO

Whey acidic protein (WAP) belongs to a family of four disulfide core (4-DSC) proteins rich in cysteine residues and is the principal whey protein found in milk of a number of mammalian species. Eutherian WAPs have two 4-DSC domains, whereas marsupial WAPs are characterized by the presence of an additional domain at the amino terminus. Structural and expression differences between marsupial and eutherian WAPs have presented challenges to identifying physiological functions of the WAP protein. We have characterized the genomic structure of tammar WAP (tWAP) gene, identified its chromosomal localization and investigated the potential function of tWAP. We have demonstrated that tWAP and domain III (DIII) of the protein alone stimulate proliferation of a mouse mammary epithelial cell line (HC11) and primary cultures of tammar mammary epithelial cells (Wall-MEC), whereas deletion of DIII from tWAP abolishes this proliferative effect. However, tWAP does not induce proliferation of human embryonic kidney (HEK293) cells. DNA synthesis and expression of cyclin D1 and cyclin-dependent kinase-4 genes were significantly up-regulated when Wall-MEC and HC11 cells were grown in the presence of either tWAP or DIII. These data suggest that DIII is the functional domain of the tWAP protein and that evolutionary pressure has led to the loss of this domain in eutherians, most likely as a consequence of adopting a reproductive strategy that relies on greater investment in development of the newborn during pregnancy.


Assuntos
Macropodidae/genética , Proteínas do Leite/química , Proteínas do Leite/metabolismo , Sequência de Aminoácidos , Animais , Ciclo Celular , Linhagem Celular , Células Cultivadas , Humanos , Macropodidae/metabolismo , Camundongos , Proteínas do Leite/genética , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência
19.
Vet Immunol Immunopathol ; 127(3-4): 269-76, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19046773

RESUMO

Cathelicidins are important components of the innate immune system and have been identified in skin and epithelia of a range of mammals. In this study molecular techniques, including RACE-PCR, were used to identify the full cDNA sequence of a cathelicidin gene, MaeuCath8, from the Australian marsupial, the tammar wallaby, Macropus eugenii. This cathelicidin was not homologous to other such genes previously isolated from a tammar wallaby mammary gland EST library, however, it did contain 4 conserved cysteine residues which characterise the pre-propeptide and had 80% identity with a previously isolated bandicoot cathelicidin. Reverse transcriptase-PCR established the expression profile of MaeuCath8 in a range of tissues, including spleen, thymus, gastrointestinal tract, skin and liver, of the tammar wallaby from birth to adulthood. Expression of MaeuCath8 was observed in spleen and gastrointestinal tract of newborn animals and was observed in most tissues by 7 days post-partum. The results indicate that pouch young could synthesize their own antimicrobial peptides from an early age suggesting that this ability most likely plays a role in protecting the pouch young from infection prior to the development of immunocompetence.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Regulação da Expressão Gênica/fisiologia , Macropodidae/metabolismo , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/genética , Sequência de Bases , Dados de Sequência Molecular , Filogenia , Catelicidinas
20.
J Exp Biol ; 211(Pt 8): 1344-51, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18375859

RESUMO

Marsupials are born in a relatively underdeveloped state and develop during a period of intensive maturation in the postnatal period. During this period, the young marsupial lacks a competent immune system, but manages to survive despite the potential of exposure to environmental pathogens. Passive immune transfer via the milk is one well-recognised strategy to compensate the neonate, but there also may be innate immune mechanisms in place. In this study, CD14 and Toll-like receptor 4 (TLR4), integral molecular components of pathogen recognition, were identified and characterised for the first time in a marsupial, the tammar wallaby (Macropus eugenii). Functional motifs of tammar CD14 and the toll/interleukin receptor (TIR) domain of TLR4 were highly conserved. The lipopolysaccharide (LPS) binding residues and the TLR4 interaction site of CD14 were conserved in all marsupials. The TIR signalling domain had 84% identity within marsupials and 77% with eutherians. Stimulation of adult tammar leukocytes resulted in the induction of a biphasic pattern of CD14 and TLR4 expression, and coincided with increased production of the pro-inflammatory cytokine TNF-alpha. Differential patterns of expression of CD14 and TLR4 were observed in tammar pouch young early in development, suggesting that early maturation of the innate immune system in these animals may have developed as an immune survival strategy to protect the marsupial neonate from exposure to microbial pathogens.


Assuntos
Animais Recém-Nascidos/metabolismo , Leucotrieno A4/farmacologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macropodidae/crescimento & desenvolvimento , Macropodidae/metabolismo , Receptor 4 Toll-Like/metabolismo , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos/genética , Animais Recém-Nascidos/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Interações Hidrofóbicas e Hidrofílicas , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Receptores de Lipopolissacarídeos/química , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/isolamento & purificação , Macropodidae/genética , Dados de Sequência Molecular , Especificidade de Órgãos/efeitos dos fármacos , Filogenia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Tempo , Receptor 4 Toll-Like/química , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA