Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 179
Filtrar
1.
Reproduction ; 168(2)2024 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-38833564

RESUMEN

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.


Asunto(s)
Atrazina , Diferenciación Celular , Herbicidas , Laminina , Testículo , Masculino , Animales , Testículo/efectos de los fármacos , Testículo/metabolismo , Testículo/citología , Atrazina/farmacología , Laminina/metabolismo , Diferenciación Celular/efectos de los fármacos , Herbicidas/farmacología , Macropodidae/metabolismo , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Células de Sertoli/citología , Estrógenos/farmacología , Estrógenos/metabolismo , Disruptores Endocrinos/farmacología , Recuento de Células , Antagonistas de Andrógenos/farmacología , Flutamida/farmacología
2.
Heredity (Edinb) ; 132(1): 5-17, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37952041

RESUMEN

The imprinted isoform of the Mest gene in mice is involved in key mammalian traits such as placental and fetal growth, maternal care and mammary gland maturation. The imprinted isoform has a distinct differentially methylated region (DMR) at its promoter in eutherian mammals but in marsupials, there are no differentially methylated CpG islands between the parental alleles. Here, we examined similarities and differences in the MEST gene locus across mammals using a marsupial, the tammar wallaby, a monotreme, the platypus, and a eutherian, the mouse, to investigate how imprinting of this gene evolved in mammals. By confirming the presence of the short isoform in all mammalian groups (which is imprinted in eutherians), this study suggests that an alternative promoter for the short isoform evolved at the MEST gene locus in the common ancestor of mammals. In the tammar, the short isoform of MEST shared the putative promoter CpG island with an antisense lncRNA previously identified in humans and an isoform of a neighbouring gene CEP41. The antisense lncRNA was expressed in tammar sperm, as seen in humans. This suggested that the conserved lncRNA might be important in the establishment of MEST imprinting in therian mammals, but it was not imprinted in the tammar. In contrast to previous studies, this study shows that MEST is not imprinted in marsupials. MEST imprinting in eutherians, therefore must have occurred after the marsupial-eutherian split with the acquisition of a key epigenetic imprinting control region, the differentially methylated CpG islands between the parental alleles.


Asunto(s)
Impresión Genómica , Macropodidae , Proteínas , ARN Largo no Codificante , Animales , Femenino , Humanos , Masculino , Ratones , Embarazo , Metilación de ADN , Euterios/genética , Euterios/metabolismo , Macropodidae/genética , Macropodidae/metabolismo , Placenta/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas/genética , Proteínas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Semen/metabolismo
3.
Cereb Cortex ; 28(1): 145-157, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29253253

RESUMEN

A hallmark of mammalian brain evolution is the emergence of the neocortex, which has expanded in all mammalian infraclasses (Eutheria, Marsupialia, Monotremata). In eutherians, neocortical neurons derive from distinct neural stem and progenitor cells (NPCs). However, precise data on the presence and abundance of the NPCs, especially of basal radial glia (bRG), in the neocortex of marsupials are lacking. This study characterized and quantified the NPCs in the developing neocortex of a marsupial, the tammar wallaby (Macropus eugenii). Our data demonstrate that its neocortex is characterized by high NPC diversity. Importantly, we show that bRG exist at high relative abundance in the tammar indicating that this cell type is not specific to the eutherian neocortex and that similar mechanisms may underlie the formation of an expanded neocortex in eutherian and marsupial mammals. We also show that bRG are likely to have been present in the therian ancestor, so did not emerge independently in the eutherian and marsupial lineages. Moreover, our data support the concept that changes in multiple parameters contribute to neocortex expansion and demonstrate the importance of bRG and other NPCs for the development and expansion of the mammalian neocortex.


Asunto(s)
Evolución Biológica , Células Ependimogliales/citología , Macropodidae/anatomía & histología , Neocórtex/citología , Animales , División Celular/fisiología , Ventrículos Cerebrales , Células Ependimogliales/metabolismo , Inmunohistoquímica , Macropodidae/crecimiento & desarrollo , Macropodidae/metabolismo , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Factor de Transcripción PAX6/metabolismo , Filogenia , Oveja Doméstica/anatomía & histología , Oveja Doméstica/crecimiento & desarrollo , Oveja Doméstica/metabolismo , Especificidad de la Especie , Proteínas de Dominio T Box/metabolismo
4.
Biol Reprod ; 99(4): 806-816, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29767687

RESUMEN

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.


Asunto(s)
Macropodidae/crecimiento & desarrollo , Macropodidae/genética , Pene/crecimiento & desarrollo , Pene/metabolismo , Transducción de Señal/genética , Uretra/crecimiento & desarrollo , Uretra/metabolismo , Andrógenos/metabolismo , Animales , Disruptores Endocrinos/toxicidad , Estrógenos/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genitales Femeninos/efectos de los fármacos , Genitales Femeninos/crecimiento & desarrollo , Genitales Femeninos/metabolismo , Genitales Masculinos/efectos de los fármacos , Genitales Masculinos/crecimiento & desarrollo , Genitales Masculinos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Macropodidae/metabolismo , Masculino , Pene/efectos de los fármacos , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Diferenciación Sexual/fisiología , Transducción de Señal/efectos de los fármacos , Uretra/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Biol Reprod ; 97(2): 217-229, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29044428

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Macropodidae/metabolismo , Ácido Retinoico 4-Hidroxilasa/metabolismo , Procesos de Determinación del Sexo/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Meiosis , Ratones , Oogénesis/fisiología , Ácido Retinoico 4-Hidroxilasa/genética , Especificidad de la Especie , Espermatogénesis/fisiología , Tretinoina/metabolismo
6.
Mol Reprod Dev ; 84(10): 1076-1085, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28688214

RESUMEN

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.


Asunto(s)
Implantación del Embrión/fisiología , Macropodidae/fisiología , Preñez , Trichosurus/fisiología , Útero/metabolismo , Animales , Células Epiteliales/metabolismo , Femenino , Macropodidae/metabolismo , Phalangeridae/metabolismo , Phalangeridae/fisiología , Embarazo , Ratas , Talina/metabolismo , Trichosurus/metabolismo , Trofoblastos/metabolismo , Útero/citología , Útero/fisiología
7.
J Vet Pharmacol Ther ; 40(6): 682-686, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28568310

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacocinética , Clindamicina/análogos & derivados , Macropodidae/metabolismo , Animales , Antibacterianos/administración & dosificación , Antibacterianos/sangre , Cromatografía Líquida de Alta Presión/veterinaria , Clindamicina/administración & dosificación , Clindamicina/sangre , Clindamicina/farmacocinética , Femenino , Semivida , Infusiones Intravenosas/veterinaria , Masculino
8.
Development ; 140(5): 965-75, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23344710

RESUMEN

Early cell lineage specification in eutherian mammals results in the formation of a pluripotent inner cell mass (ICM) and trophoblast. By contrast, marsupials have no ICM. Here, we present the first molecular analysis of mechanisms of early cell lineage specification in a marsupial, the tammar wallaby. There was no overt differential localisation of key lineage-specific transcription factors in cleavage and early unilaminar blastocyst stages. Pluriblast cells (equivalent to the ICM) became distinguishable from trophoblast cells by differential expression of POU5F1 and, to a greater extent, POU2, a paralogue of POU5F1. Unlike in the mouse, pluriblast-trophoblast differentiation coincided with a global nuclear-to-cytoplasmic transition of CDX2 localisation. Also unlike in the mouse, Hippo pathway factors YAP and WWTR1 showed mutually distinct localisation patterns that suggest non-redundant roles. NANOG and GATA6 were conserved as markers of epiblast and hypoblast, respectively, but some differences to the mouse were found in their mode of differentiation. Our results suggest that there is considerable evolutionary plasticity in the mechanisms regulating early lineage specification in mammals.


Asunto(s)
Tipificación del Cuerpo , Linaje de la Célula , Mamíferos , Marsupiales/embriología , Animales , Tipificación del Cuerpo/genética , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Variación Genética/fisiología , Macropodidae/embriología , Macropodidae/genética , Macropodidae/metabolismo , Macropodidae/fisiología , Mamíferos/embriología , Mamíferos/genética , Mamíferos/metabolismo , Mamíferos/fisiología , Marsupiales/genética , Marsupiales/metabolismo , Ratones , Especificidad de Órganos/genética , Embarazo , Transducción de Señal/genética , Factores de Tiempo , Distribución Tisular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Reproduction ; 152(5): 491-505, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27486272

RESUMEN

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.


Asunto(s)
Blastocisto/metabolismo , Implantación Tardía del Embrión/fisiología , Desarrollo Embrionario , Macropodidae/metabolismo , Metamorfosis Biológica/fisiología , Proteoma/metabolismo , Útero/metabolismo , Animales , Blastocisto/citología , Endometrio/crecimiento & desarrollo , Endometrio/metabolismo , Femenino , Macropodidae/crecimiento & desarrollo , Embarazo , Espectrometría de Masas en Tándem , Útero/crecimiento & desarrollo
10.
J Exp Biol ; 218(Pt 21): 3425-34, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26538176

RESUMEN

Fundamental differences in methane (CH4) production between macropods (kangaroos) and ruminants have been suggested and linked to differences in the composition of the forestomach microbiome. Using six western grey kangaroos (Macropus fuliginosus) and four red kangaroos (Macropus rufus), we measured daily absolute CH4 production in vivo as well as CH4 yield (CH4 per unit of intake of dry matter, gross energy or digestible fibre) by open-circuit respirometry. Two food intake levels were tested using a chopped lucerne hay (alfalfa) diet. Body mass-specific absolute CH4 production resembled values previously reported in wallabies and non-ruminant herbivores such as horses, and did not differ with food intake level, although there was no concomitant proportionate decrease in fibre digestibility with higher food intake. In contrast, CH4 yield decreased with increasing intake, and was intermediate between values reported for ruminants and non-ruminant herbivores. These results correspond to those in ruminants and other non-ruminant species where increased intake (and hence a shorter digesta retention in the gut) leads to a lower CH4 yield. We hypothesize that rather than harbouring a fundamentally different microbiome in their foregut, the microbiome of macropods is in a particular metabolic state more tuned towards growth (i.e. biomass production) rather than CH4 production. This is due to the short digesta retention time in macropods and the known distinct 'digesta washing' in the gut of macropods, where fluids move faster than particles and hence most likely wash out microbes from the forestomach. Although our data suggest that kangaroos only produce about 27% of the body mass-specific volume of CH4 of ruminants, it remains to be modelled with species-specific growth rates and production conditions whether or not significantly lower CH4 amounts are emitted per kg of meat in kangaroo than in beef or mutton production.


Asunto(s)
Digestión , Sistema Digestivo/metabolismo , Ingestión de Alimentos , Macropodidae/metabolismo , Metano/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Fibras de la Dieta , Medicago sativa , Especificidad de la Especie
11.
Dev Dyn ; 243(2): 324-38, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24115631

RESUMEN

BACKGROUND: At birth, marsupial neonates have precociously developed forelimbs. The development of the tammar wallaby (Macropus eugenii) hindlimbs lags significantly behind that of the forelimbs. This differs from the grey short-tailed opossum, Monodelphis domestica, which has relatively similar fore- and hindlimbs at birth. This study examines the expression of the key patterning genes TBX4, TBX5, PITX1, FGF8, and SHH in developing limb buds in the tammar wallaby. RESULTS: All genes examined were highly conserved with orthologues from opossum and mouse. TBX4 expression appeared earlier in development than in the mouse, but later than in the opossum. SHH expression is restricted to the zone of polarising activity, while TBX5 (forelimb) and PITX1 (hindlimb) showed diffuse mRNA expression. FGF8 is specifically localised to the apical ectodermal ridge, which is more prominent than in the opossum. CONCLUSIONS: The most marked divergence in limb size in marsupials occurs in the kangaroos and wallabies. The faster development of the fore limb compared to that of the hind limb correlates with the early timing of the expression of the key patterning genes in these limbs.


Asunto(s)
Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Esbozos de los Miembros/crecimiento & desarrollo , Esbozos de los Miembros/metabolismo , Macropodidae/crecimiento & desarrollo , Factores de Edad , Animales , Cartilla de ADN/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Técnicas Histológicas , Hibridación in Situ , Esbozos de los Miembros/ultraestructura , Macropodidae/metabolismo , Microscopía Electrónica de Rastreo , Especificidad de la Especie , Proteínas de Dominio T Box/metabolismo
12.
Semin Cell Dev Biol ; 23(5): 547-56, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22498725

RESUMEN

The role of milk extends beyond simply providing nutrition to the suckled young. Milk has a comprehensive role in programming and regulating growth and development of the suckled young, and provides a number of potential autocrine factors so that the mammary gland functions appropriately during the lactation cycle. This central role of milk is best studied in animal models such as marsupials that have evolved a different lactation strategy to eutherians and allow researchers to more easily identify regulatory mechanisms that are not as readily apparent in eutherian species. For example, the tammar wallaby (Macropus eugenii) has evolved with a unique reproductive strategy of a short gestation, birth of an altricial young and a relatively long lactation during which the mother progressively changes the composition of the major, and many of the minor components of milk. Consequently, in contrast to eutherians, there is a far greater investment in development of the young during lactation and it is likely that many of the signals that regulate development of eutherian embryos in utero are delivered by the milk. This requires the co-ordinated development and function of the mammary gland since inappropriate timing of these signalling events may result in either limited or abnormal development of the young, and potentially a higher incidence of mature onset disease. Milk proteins play a significant role in these processes by providing timely presentation of signalling molecules and antibacterial protection for the young and the mammary gland at times when there is increased susceptibility to infection. This review describes studies exploiting the unique reproductive strategy of the tammar wallaby to investigate the role of several proteins secreted at specific times during the lactation cycle and that are correlated with potential roles in the young and mammary gland. Interestingly, alternative splicing of some milk protein genes has been utilised by the mammary gland to deliver domain-specific functions at specific times during lactation.


Asunto(s)
Macropodidae/metabolismo , Proteínas de la Leche/metabolismo , Animales , Femenino , Humanos , Lactancia , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Modelos Biológicos
13.
Reproduction ; 147(1): 21-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24123130

RESUMEN

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.


Asunto(s)
Endometrio/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Macropodidae/embriología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Cuerpo Lúteo/metabolismo , Femenino , Macropodidae/metabolismo , Embarazo , Progesterona/metabolismo , Útero/metabolismo
14.
Reproduction ; 147(3): 279-89, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24307230

RESUMEN

The X-linked aristaless gene, ARX, is essential for the development of the gonads, forebrain, olfactory bulb, pancreas, and skeletal muscle in mice and humans. Mutations cause neurological diseases, often accompanied by ambiguous genitalia. There are a disproportionately high number of testis and brain genes on the human and mouse X chromosomes. It is still unknown whether the X chromosome accrued these genes during its evolution or whether genes that find themselves on the X chromosome evolve such roles. ARX was originally autosomal in mammals and remains so in marsupials, whereas in eutherian mammals it translocated to the X chromosome. In this study, we examined autosomal ARX in tammars and compared it with the X-linked Arx in mice. We detected ARX mRNA in the neural cells of the forebrain, midbrain and hindbrain, and olfactory bulbs in developing tammars, consistent with the expression in mice. ARX was detected by RT-PCR and mRNA in situ hybridization in the developing tammar wallaby gonads of both sexes, suggestive of a role in sexual development as in mice. We also detected ARX/Arx mRNA in the adult testis in both tammars and mice, suggesting a potential novel role for ARX/Arx in spermiogenesis. ARX transcripts were predominantly observed in round spermatids. Arx mRNA localization distributions in the mouse adult testis suggest that it escaped meiotic sex chromosome inactivation during spermatogenesis. Our findings suggest that ARX in the therian mammal ancestor already played a role in male reproduction before it was recruited to the X chromosome in eutherians.


Asunto(s)
Células Germinativas/metabolismo , Proteínas de Homeodominio/genética , Marsupiales/genética , Ratones/genética , Espermatogénesis/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Macropodidae/genética , Macropodidae/metabolismo , Masculino , Marsupiales/crecimiento & desarrollo , Marsupiales/metabolismo , Ratones/crecimiento & desarrollo , Ratones/metabolismo , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Maduración Sexual/genética , Espermatogonias/metabolismo , Factores de Transcripción/metabolismo , Cromosoma X
15.
Nat Commun ; 15(1): 3953, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729967

RESUMEN

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.


Asunto(s)
Lactancia , Macropodidae , Glándulas Mamarias Animales , Animales , Femenino , Humanos , Ratones , Diferenciación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Células Epiteliales/metabolismo , Macropodidae/metabolismo , Mamíferos , Glándulas Mamarias Animales/metabolismo , Marsupiales , Leche/metabolismo
16.
Mol Phylogenet Evol ; 69(1): 4-16, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23707702

RESUMEN

S100 proteins are calcium-binding proteins involved in controlling diverse intracellular and extracellular processes such as cell growth, differentiation, and antimicrobial function. We recently identified a S100-like cDNA from the tammar wallaby (Macropus eugenii) stomach. Phylogentic analysis shows wallaby S100A19 forms a new clade with other marsupial and monotreme S100A19, while this group shows similarity to eutherian S100A7 and S100A15 genes. This is also supported by amino acid and domain comparisons. We show S100A19 is developmentally-regulated in the tammar wallaby gut by demonstrating the gene is expressed in the forestomach of young animals at a time when the diet consists of only milk, but is absent in older animals when the diet is supplemented with herbage. During this transition the forestomach phenotype changes from a gastric stomach into a fermentation sac and intestinal flora changes with diet. We also show that S100A19 is expressed in the mammary gland of the tammar wallaby only during specific stages of lactation; the gene is up-regulated during pregnancy and involution and not expressed during the milk production phase of lactation. Comparison of the tammar wallaby S100A19 protein sequence with S100 protein sequences from eutherian, monotreme and other marsupial species suggest the marsupial S100A19 has two functional EF hand domains, and an extended His tail. An evolutionary analysis of S100 family proteins was carried out to gain a better understanding of the relationship between the S100 family member functions. We propose that S100A19 gene/protein is the ancestor of the eutherian S100A7 gene/protein, which has subsequently modified its original function in eutherians. This modified function may have arisen due to differentiation of evolutionary pressures placed on gut and mammary gland developmental during mammal evolution. The highly regulated differential expression patterns of S100A19 in the tammar wallaby suggests that S100A19 may play a role in gut development, which differs between metatherians and eutherians, and/or include a potential antibacterial role in order to establish the correct flora and protect against spiral bacteria in the immature forestomach. In the mammary gland it may protect the tissue from infection at times of vulnerability during the lactation cycle.


Asunto(s)
Evolución Molecular , Marsupiales/genética , Filogenia , Isoformas de Proteínas/genética , Proteínas S100/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario/genética , ADN Complementario/metabolismo , Femenino , Mucosa Gástrica/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Lactancia/fisiología , Macropodidae/clasificación , Macropodidae/genética , Macropodidae/metabolismo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/metabolismo , Marsupiales/clasificación , Marsupiales/metabolismo , Datos de Secuencia Molecular , Embarazo , Isoformas de Proteínas/clasificación , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas S100/clasificación , Proteínas S100/metabolismo , Análisis de Secuencia de ADN , Estómago/crecimiento & desarrollo
17.
Reprod Fertil Dev ; 25(2): 456-61, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22951148

RESUMEN

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.


Asunto(s)
Cuerpo Lúteo/metabolismo , Lactancia/fisiología , Macropodidae/fisiología , Prolactina/metabolismo , Animales , Territorio de la Capital Australiana , Femenino , Modelos Lineales , Macropodidae/metabolismo , Progesterona/biosíntesis , Prolactina/sangre , Radioinmunoensayo , Estaciones del Año , Especificidad de la Especie , Factores de Tiempo
18.
J Endocrinol ; 258(3)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37343228

RESUMEN

Since the discovery in 1968 that dihydrotestosterone (DHT) is a major mediator of androgen action, a convincing body of evidence has accumulated to indicate that the major pathway of DHT formation is the 5α-reduction of circulating testosterone in androgen target tissues. However, we now know that DHT can also be formed in peripheral tissues by the oxidation of 5α-androstane-3α,17ß-diol (adiol). This pathway is responsible for the formation of the male phenotype. We discuss the serendipitous discovery in the tammar wallaby of an alternate pathway by which adiol is formed in the testes, secreted into plasma and converted in peripheral tissues to DHT. This alternate pathway is responsible for virilisation of the urogenital system in this species and is present in the testes at the onset of male puberty of all mammals studied so far. This is the first clear-cut function for steroid 5α-reductase 1 in males. Unexpectedly, the discovery of this pathway in this Australian marsupial has had a major impact in understanding the pathophysiology of aberrant virilisation in female newborns. Overactivity of the alternate pathway appears to explain virilisation in congenital adrenal hyperplasia CAH, in X-linked 46,XY disorders of sex development. It also appears to be important in polycystic ovarian syndrome (PCOS) since PCOS ovaries have enhanced the expression of genes and proteins of the alternate pathway. It is now clear that normal male development in marsupials, rodents and humans requires the action of both the classic and the alternate (backdoor) pathways.


Asunto(s)
Andrógenos , Testosterona , Recién Nacido , Humanos , Animales , Masculino , Femenino , Andrógenos/metabolismo , Australia , Testosterona/metabolismo , Dihidrotestosterona , Macropodidae/metabolismo , Virilismo
19.
Am Nat ; 179(2): 169-77, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22218307

RESUMEN

Rensch's rule, which states that the magnitude of sexual size dimorphism tends to increase with increasing body size, has evolved independently in three lineages of large herbivorous mammals: bovids (antelopes), cervids (deer), and macropodids (kangaroos). This pattern can be explained by a model that combines allometry, life-history theory, and energetics. The key features are that female group size increases with increasing body size and that males have evolved under sexual selection to grow large enough to control these groups of females. The model predicts relationships among body size and female group size, male and female age at first breeding, death and growth rates, and energy allocation of males to produce body mass and weapons. Model predictions are well supported by data for these megaherbivores. The model suggests hypotheses for why some other sexually dimorphic taxa, such as primates and pinnipeds (seals and sea lions), do or do not conform to Rensh's rule.


Asunto(s)
Tamaño Corporal , Herbivoria , Macropodidae/anatomía & histología , Modelos Biológicos , Rumiantes/anatomía & histología , Animales , Caniformia/anatomía & histología , Caniformia/metabolismo , Femenino , Macropodidae/metabolismo , Masculino , Primates/anatomía & histología , Primates/metabolismo , Reproducción , Rumiantes/metabolismo , Caracteres Sexuales , Conducta Social , Especificidad de la Especie
20.
Chem Senses ; 37(6): 567-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22383629

RESUMEN

The vomeronasal organ (VNO) detects pheromones via 2 large families of receptors: vomeronasal receptor 1, associated with the protein Giα2, and vomeronasal receptor 2, associated with Goα. We investigated the distribution of Goα in the developing and adult VNO and adult olfactory bulb of a marsupial, the tammar wallaby. Some cells expressed Goα as early as day 5 postpartum, but by day 30, Goα expressing cells were distributed throughout the receptor epithelium of the VNO. In the adult tammar, Goα appeared to be expressed in sensory neurons whose nuclei were mostly basally located in the vomeronasal receptor epithelium. Goα expressing vomeronasal receptor cells led to all areas of the accessory olfactory bulb (AOB). The lack of regionally restricted projection of the vomeronasal receptor cell type 2 in the tammar was similar to the uniform type, with the crucial difference that the uniform type only shows expression of Giα2 and no expression of Goα. The observed Goα staining pattern suggests that the tammar may have a third accessory olfactory type that could be intermediate to the segregated and uniform types already described.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/biosíntesis , Macropodidae/metabolismo , Bulbo Olfatorio/metabolismo , Órgano Vomeronasal/metabolismo , Animales , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/análisis , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda