Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.079
Filtrar
1.
Int J Mol Sci ; 22(18)2021 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-34576272

RESUMO

Follicle stimulating hormone (FSH) is produced by the pituitary gland in a coordinated hypothalamic-pituitary-gonadal (HPG) axis event, plays important roles in reproduction and germ cell development during different phases of reproductive development (fetal, neonatal, puberty, and adult life), and is consequently essential for fertility. FSH is a heterodimeric glycoprotein hormone of two dissociable subunits, α and ß. The FSH ß-subunit (FSHß) function starts upon coupling to its specific receptor: follicle-stimulating hormone receptor (FSHR). FSHRs are localized mainly on the surface of target cells on the testis and ovary (granulosa and Sertoli cells) and have recently been found in testicular stem cells and extra-gonadal tissue. Several reproduction disorders are associated with absent or low FSH secretion, with mutation of the FSH ß-subunit or the FSH receptor, and/or its signaling pathways. However, the influence of FSH on germ cells is still poorly understood; some studies have suggested that this hormone also plays a determinant role in the self-renewal of germinative cells and acts to increase undifferentiated spermatogonia proliferation. In addition, in vitro, together with other factors, it assists the process of differentiation of primordial germ cells (PGCLCs) into gametes (oocyte-like and SSCLCs). In this review, we describe relevant research on the influence of FSH on spermatogenesis and folliculogenesis, mainly in the germ cell of humans and other species. The possible roles of FSH in germ cell generation in vitro are also presented.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Ovário/metabolismo , Células de Sertoli/metabolismo , Animais , Dimerização , Feminino , Fertilidade , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Células Germinativas/metabolismo , Gonadotropinas/metabolismo , Humanos , Masculino , Camundongos , Ovário/embriologia , Ovário/crescimento & desenvolvimento , Hipófise/embriologia , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , Ratos , Receptores do FSH/metabolismo , Reprodução , Maturidade Sexual , Espermatogênese/genética , Espermatogônias/citologia
2.
Eur J Endocrinol ; 185(1): 121-135, 2021 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-33950863

RESUMO

Objective: The transcription factor OTX2 is implicated in ocular, craniofacial, and pituitary development. Design: We aimed to establish the contribution of OTX2 mutations in congenital hypopituitarism patients with/without eye abnormalities, study functional consequences, and establish OTX2 expression in the human brain, with a view to investigate the mechanism of action. Methods: We screened patients from the UK (n = 103), international centres (n = 24), and Brazil (n = 282); 145 were within the septo-optic dysplasia spectrum, and 264 had no eye phenotype. Transactivation ability of OTX2 variants was analysed in murine hypothalamic GT1-7 neurons. In situ hybridization was performed on human embryonic brain sections. Genetically engineered mice were generated with a series of C-terminal OTX2 variants. Results: Two chromosomal deletions and six haploinsufficient mutations were identified in individuals with eye abnormalities; an affected relative of one patient harboured the same mutation without an ocular phenotype. OTX2 truncations led to significant transactivation reduction. A missense variant was identified in another patient without eye abnormalities; however, studies revealed it was most likely not causative. In the mouse, truncations proximal to aa219 caused anophthalmia, while distal truncations and the missense variant were tolerated. During human embryogenesis, OTX2 was expressed in the posterior pituitary, retina, ear, thalamus, choroid plexus, and partially in the hypothalamus, but not in the anterior pituitary. Conclusions: OTX2 mutations are rarely associated with hypopituitarism in isolation without eye abnormalities, and may be variably penetrant, even within the same pedigree. Our data suggest that the endocrine phenotypes in patients with OTX2 mutations are of hypothalamic origin.


Assuntos
Hipopituitarismo/fisiopatologia , Microftalmia/fisiopatologia , Neurônios/fisiologia , Fatores de Transcrição Otx/genética , Hipófise/fisiopatologia , Displasia Septo-Óptica/fisiopatologia , Adolescente , Animais , Animais Geneticamente Modificados , Brasil , Linhagem Celular , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Hipopituitarismo/embriologia , Hipopituitarismo/genética , Hipotálamo/citologia , Lactente , Masculino , Camundongos , Microftalmia/embriologia , Microftalmia/genética , Mutação , Neurônios/patologia , Linhagem , Hipófise/embriologia , Hipófise/patologia , Displasia Septo-Óptica/embriologia , Displasia Septo-Óptica/genética , Reino Unido
3.
Development ; 148(4)2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33526583

RESUMO

Basement membranes (BM) are extracellular matrices assembled into complex and highly organized networks essential for organ morphogenesis and function. However, little is known about the tissue origin of BM components and their dynamics in vivo Here, we unravel the assembly and role of the BM main component, Collagen type IV (ColIV), in Drosophila ovarian stalk morphogenesis. Stalks are short strings of cells assembled through cell intercalation that link adjacent follicles and maintain ovarian integrity. We show that stalk ColIV has multiple origins and is assembled following a regulated pattern leading to a unique BM organisation. Absence of ColIV leads to follicle fusion, as observed upon ablation of stalk cells. ColIV and integrins are both required to trigger cell intercalation and maintain mechanically strong cell-cell attachment within the stalk. These results show how the dynamic assembly of a mosaic BM controls complex tissue morphogenesis and integrity.


Assuntos
Membrana Basal/metabolismo , Comunicação Celular , Drosophila/embriologia , Drosophila/metabolismo , Ovário/embriologia , Ovário/metabolismo , Animais , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Feminino , Imunofluorescência , Morfogênese , Organogênese , Hipófise/embriologia , Hipófise/metabolismo
4.
Sci Rep ; 10(1): 21920, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33318609

RESUMO

Maternal exposure to increased steroid hormones, including estrogens, androgens or glucocorticoids during pregnancy results in chronic conditions in offspring that manifest in adulthood. Little is known about effects of progesterone administration in early pregnancy on fetal development. We hypothesised that maternal early pregnancy progesterone supplementation would increase fetal progesterone, affect progesterone target tissues in the developing fetal reproductive system and be metabolised to other bioactive steroids in the fetus. We investigated the effects of progesterone treatment during early pregnancy on maternal and fetal plasma progesterone concentrations, transcript abundance in the fetal pituitary and testes and circulating steroids, at day 75 gestation, using a clinically realistic ovine model. Endogenous progesterone concentrations were lower in male than female fetuses. Maternal progesterone administration increased male, but not female, fetal progesterone concentrations, also increasing circulating 11-dehydrocorticosterone in male fetuses. Maternal progesterone administration altered fetal pituitary and testicular function in ovine male fetuses. This suggests that there may be fetal sex specific effects of the use of progesterone in early pregnancy, and highlights that progesterone supplementation should be used only when there is clear evidence of efficacy and for as limited time as necessary.


Assuntos
Desenvolvimento Fetal/efeitos dos fármacos , Feto/embriologia , Hipófise/embriologia , Progesterona/farmacologia , Ovinos/metabolismo , Testículo/embriologia , Animais , Feminino , Masculino , Gravidez , Progesterona/efeitos adversos
5.
Dev Dyn ; 249(12): 1425-1439, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32633438

RESUMO

BACKGROUND: Hedgehog signaling has various regulatory functions in tissue morphogenesis and differentiation. To investigate its involvement in anterior pituitary precursor development and the lens precursor potential for anterior pituitary precursors, we investigated Talpid mutant Japanese quail embryos, in which hedgehog signaling is defective. RESULTS: Talpid mutants develop multiple pituitary precursor-like pouches of variable sizes from the oral ectoderm (OE). The ectopic pituitary pouches initially express the pituitary-associated transcription factor (TF) LHX3 similarly to Rathke's pouch, the genuine pituitary precursor. The pouches coexpress the TFs SOX2 and PAX6, a signature of lens developmental potential. Most Talpid mutant pituitary pouches downregulate LHX3 expression and activate the lens-essential TF PROX1, leading to the development of small lens tissue expressing α-, ß-, and δ-crystallins. In contrast, mutant Rathke's pouches express a lower level of LHX3, which is primarily localized in the cytoplasm, and activate the lens developmental pathway. CONCLUSIONS: Hedgehog signaling in normal embryos regulates the development of Rathke's pouch in two steps. First, by confining Rathke's pouch development in a low hedgehog signaling region of the OE. Second, by sustaining LHX3 activity to promote anterior pituitary development, while inhibiting ectopic lens development.


Assuntos
Ectoderma/embriologia , Desenvolvimento Embrionário/fisiologia , Proteínas Hedgehog/metabolismo , Fator de Transcrição PAX6/metabolismo , Hipófise/embriologia , Fatores de Transcrição SOXB1/metabolismo , Animais , Diferenciação Celular/fisiologia , Coturnix , Ectoderma/metabolismo , Organogênese/fisiologia , Hipófise/metabolismo , Transdução de Sinais/fisiologia
6.
Dev Dyn ; 249(11): 1365-1376, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32506585

RESUMO

BACKGROUND: The nuclear progesterone receptor (Pgr) is a ligand-dependent transcription factor primarily responsible for mediating progesterone actions relevant for reproduction across vertebrates. Information on the cellular localization of Pgr expression in the reproductive system is required for developing a comprehensive approach to elucidate the role of Pgr in reproduction. RESULTS: We generated transgenic zebrafish Tg(pgr:eGFP) that express enhanced green fluorescent protein (eGFP) driven by promoter sequence of pgr gene. The tissue distribution pattern of egfp mRNA is consistent with the pgr mRNA expression in Tg(pgr:eGFP). In the pituitary, GFP signals are found in the proximal pars distalis. In order to better discern the cellular localization of GFP signals in gonads, Tg(pgr:eGFP) line was crossed with Tg(gsdf:nfsB-mCherry) line, specifically expressing nitroreductase-mCherry fusion protein in granulosa and Sertoli cells in ovary and testis, respectively. Imaging of testis tissue showed that GFP expression was confined to Leydig cells. In the ovary, GFP expression colocalized with the mCherry signal in granulosa cells. Intriguingly, we also identified some non-granulosa cells close to where blood vessels branched, expressing stronger GFP signals than granulosa cells. CONCLUSIONS: Analyzing Tg(pgr:eGFP) expression in zebrafish provided leads toward new routes to study the role of Pgr in reproduction.


Assuntos
Animais Geneticamente Modificados , Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Gônadas/embriologia , Proteínas de Fluorescência Verde , Hipófise/embriologia , Regiões Promotoras Genéticas , Receptores de Progesterona/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/genética , Feminino , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Masculino , Peixe-Zebra/embriologia , Peixe-Zebra/genética
7.
Gen Comp Endocrinol ; 295: 113524, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32526331

RESUMO

Early life stages are sensitive to environmental insults and changes during critical developmental periods; this can often result in altered adult behaviour and physiology. Examining the development of the hypothalamus-pituitary-interrenal (HPI) axis and its responsiveness, or lack thereof, during development are important for understanding the short- and long-term impacts of stressors on embryonic and larval fish. We examined the ontogeny of the HPI axis in embryonic (21, 38, 63, 83 and 103 days post-fertilisation (dpf)) and larval (1, 2, 3 and 4 weeks post-hatch (wph)) lake whitefish (Coregonus clupeaformis) by quantifying changes in mRNA levels of several genes associated with HPI axis functioning and whole animal cortisol levels throughout development and in response to a severe or mild hypoxic stress. Cortisol, and crh, crhbp1, pomc and star transcripts were detected from the earliest embryonic age studied. Cortisol levels in control embryos decreased between 21 and 63 dpf, suggesting the utilisation of maternal cortisol deposits. However, by 83 dpf (70% developed) endogenous de novo synthesis had generated a 4.5-fold increase in whole embryo cortisol. Importantly, we provide novel data showing that the HPI axis can be activated even earlier. Whole body cortisol increased in eyed lake whitefish embryos (38 dpf; ~32% developed) in response to hypoxia stress. Coincident with this hypoxia-induced increase in cortisol in 38 dpf embryos were corresponding increases in crh, crhbp1, pomc and star transcript levels. Beyond 38 dpf, the HPI axis in lake whitefish embryos was hyporesponsive to hypoxia stress at all embryonic ages examined (63, 83 and 103 dpf; 54, 72 and 85% developed, respectively). Post-hatch, larvae responded to hypoxia with an increase in cortisol levels and HPI axis genes at 1 wph, but this response was lost and larvae appeared hyporesponsive at subsequent ages (2, 3 and 4 wph). Collectively our work demonstrates that during fish embryogenesis and the larval stage there are windows where the HPI axis is responsive and windows where it is truly hyporesponsive; both could be beneficial in ensuring undisrupted development particularly in the face of increasing environmental changes.


Assuntos
Hipotálamo/embriologia , Hipóxia/embriologia , Lagos , Hipófise/embriologia , Salmonidae/embriologia , Animais , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Hidrocortisona/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Salmonidae/genética
8.
Compr Physiol ; 10(2): 389-413, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32163208

RESUMO

The development of the anterior pituitary gland occurs in distinct sequential developmental steps, leading to the formation of a complex organ containing five different cell types secreting six different hormones. During this process, the temporal and spatial expression of a cascade of signaling molecules and transcription factors plays a crucial role in organ commitment, cell proliferation, patterning, and terminal differentiation. The morphogenesis of the gland and the emergence of distinct cell types from a common primordium are governed by complex regulatory networks involving transcription factors and signaling molecules that may be either intrinsic to the developing pituitary or extrinsic, originating from the ventral diencephalon, the oral ectoderm, and the surrounding mesenchyme. Endocrine cells of the pituitary gland are organized into structural and functional networks that contribute to the coordinated response of endocrine cells to stimuli; these cellular networks are formed during embryonic development and are maintained or may be modified in adulthood, contributing to the plasticity of the gland. Abnormalities in any of the steps of pituitary development may lead to congenital hypopituitarism that includes a spectrum of disorders from isolated to combined hormone deficiencies including syndromic disorders such as septo-optic dysplasia. Over the past decade, the acceleration of next-generation sequencing has allowed for rapid analysis of the patient genome to identify novel mutations and novel candidate genes associated with hypothalmo-pituitary development. Subsequent functional analysis using patient fibroblast cells, and the generation of stem cells derived from patient cells, is fast replacing the need for animal models while providing a more physiologically relevant characterization of novel mutations. Furthermore, CRISPR-Cas9 as the method for gene editing is replacing previous laborious and time-consuming gene editing methods that were commonly used, thus yielding knockout cell lines in a fraction of the time. © 2020 American Physiological Society. Compr Physiol 10:389-413, 2020.


Assuntos
Hipófise/crescimento & desenvolvimento , Animais , Humanos , Morfogênese , Doenças da Hipófise/genética , Doenças da Hipófise/metabolismo , Doenças da Hipófise/patologia , Hipófise/citologia , Hipófise/embriologia , Hipófise/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Endocrinology ; 161(2)2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913463

RESUMO

Dietary vitamin A is metabolized into bioactive retinoic acid (RA) in vivo and regulates the development of many embryonic tissues. RA signaling is active in the oral ectoderm-derived tissues of the neuroendocrine system, but its role there has not yet been fully explored. We show here that RA signaling is active during pituitary organogenesis and dependent on the pituitary transcription factor Prop1. Prop1-mutant mice show reduced expression of the aldehyde dehydrogenase gene Aldh1a2, which metabolizes the vitamin A-intermediate retinaldehyde into RA. To elucidate the specific function of RA signaling during neuroendocrine development, we studied a conditional deletion of Aldh1a2 and a dominant-negative mouse model of inhibited RA signaling during pituitary organogenesis. These models partially phenocopy Prop1-mutant mice by exhibiting embryonic pituitary dysmorphology and reduced hormone expression, especially thyrotropin. These findings establish the role of RA in embryonic pituitary stem cell progression to differentiated hormone cells and raise the question of gene-by-environment interactions as contributors to pituitary development and disease.


Assuntos
Família Aldeído Desidrogenase 1/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipófise/embriologia , Retinal Desidrogenase/metabolismo , Tretinoína/metabolismo , Família Aldeído Desidrogenase 1/genética , Animais , Camundongos Endogâmicos C57BL , Organogênese , Hipófise/metabolismo , Retinal Desidrogenase/genética , Transdução de Sinais , Células-Tronco/metabolismo , Tireotropina/metabolismo
10.
Genomics ; 112(1): 934-942, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31200027

RESUMO

Long non-coding RNAs are transcribed into RNA molecules that are >200 nucleotides in length. However, the expression and function analysis of lncRNAs in the sheep pituitary gland are still lacking. In this study, we identified 1755 lncRNAs (545 annotated lncRNAs and 1210 novel lncRNAs) from RNA-seq data in the pituitary gland of embryonic and adult sheep. A total of 235 lncRNAs were differentially expressed between embryonic and adult group. We verified the presence of some lncRNAs using RT-PCR and DNA sequencing, and identified some differentially expressed lncRNAs using qPCR. We also investigated the role of cis-acting lncRNAs on target genes. GO and KEGG enrichment analysis revealed that the target genes of lncRNAs were involved in the regulation of hormones secretion and some signaling pathways in the sheep pituitary gland. Our study provides comprehensive expression profiles of lncRNAs and valuable resource for understanding their function in the pituitary gland.


Assuntos
Hipófise/metabolismo , RNA Longo não Codificante/metabolismo , Ovinos/genética , Animais , Feminino , Regulação da Expressão Gênica , Ontologia Genética , Hipófise/embriologia , RNA Mensageiro/metabolismo , Ovinos/embriologia , Ovinos/metabolismo
11.
Sci Rep ; 9(1): 5413, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931985

RESUMO

The hypothalamus-pituitary-thyroid (HPT) axis plays a crucial role in the metabolism, homeostasis, somatic growth and development of teleostean fishes. Thyroid hormones regulate essential biological functions such as growth and development, regulation of stress, energy expenditure, tissue compound, and psychological processes. Teleost thyroid follicles produce the same thyroid hormones as in other vertebrates: thyroxin (T4) and triiodothyronine (T3), making the zebrafish a very useful model to study hypo- and hyperthyroidism in other vertebrate taxa, including humans. Here we investigate morphological changes in T3 hyperthyroid cases in the zebrafish to better understand malformations provoked by alterations of T3 levels. In particular, we describe musculoskeletal abnormalities during the development of the zebrafish appendicular skeleton and muscles, compare our observations with those recently done by us on the normal developmental of the zebrafish, and discuss these comparisons within the context of evolutionary developmental pathology (Evo-Devo-Path), including human pathologies.


Assuntos
Hipertireoidismo/fisiopatologia , Músculos/fisiologia , Desenvolvimento Musculoesquelético/fisiologia , Glândula Tireoide/fisiologia , Animais , Evolução Biológica , Padronização Corporal/fisiologia , Embrião não Mamífero/embriologia , Embrião não Mamífero/fisiologia , Humanos , Hipertireoidismo/embriologia , Hipertireoidismo/metabolismo , Hipotálamo/embriologia , Hipotálamo/fisiologia , Larva/fisiologia , Modelos Animais , Músculos/embriologia , Sistema Musculoesquelético/embriologia , Hipófise/embriologia , Hipófise/fisiologia , Glândula Tireoide/embriologia , Glândula Tireoide/metabolismo , Hormônios Tireóideos/metabolismo , Peixe-Zebra
12.
Elife ; 82019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30912742

RESUMO

SOX2 positive pituitary stem cells (PSCs) are specified embryonically and persist throughout life, giving rise to all pituitary endocrine lineages. We have previously shown the activation of the STK/LATS/YAP/TAZ signalling cascade in the developing and postnatal mammalian pituitary. Here, we investigate the function of this pathway during pituitary development and in the regulation of the SOX2 cell compartment. Through loss- and gain-of-function genetic approaches, we reveal that restricting YAP/TAZ activation during development is essential for normal organ size and specification from SOX2+ PSCs. Postnatal deletion of LATS kinases and subsequent upregulation of YAP/TAZ leads to uncontrolled clonal expansion of the SOX2+ PSCs and disruption of their differentiation, causing the formation of non-secreting, aggressive pituitary tumours. In contrast, sustained expression of YAP alone results in expansion of SOX2+ PSCs capable of differentiation and devoid of tumourigenic potential. Our findings identify the LATS/YAP/TAZ signalling cascade as an essential component of PSC regulation in normal pituitary physiology and tumourigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Hipófise/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Células-Tronco/fisiologia , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Deleção de Genes , Redes Reguladoras de Genes , Camundongos , Hipófise/embriologia , Hipófise/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/análise , Células-Tronco/química , Proteínas de Sinalização YAP
13.
Endocrinology ; 159(12): 3910-3924, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30335147

RESUMO

Transcription factors and signaling pathways that regulate stem cells and specialized hormone-producing cells in the pituitary gland have been the subject of intense study and have yielded a mechanistic understanding of pituitary organogenesis and disease. However, the regulation of stem cell proliferation and differentiation, the heterogeneity among specialized hormone-producing cells, and the role of nonendocrine cells in the gland remain important, unanswered questions. Recent advances in single-cell RNA sequencing (scRNAseq) technologies provide new avenues to address these questions. We performed scRNAseq on ∼13,663 cells pooled from six whole pituitary glands of 7-week-old C57BL/6 male mice. We identified pituitary endocrine and stem cells in silico, as well as other support cell types such as endothelia, connective tissue, and red and white blood cells. Differential gene expression analyses identify known and novel markers of pituitary endocrine and stem cell populations. We demonstrate the value of scRNAseq by in vivo validation of a novel gonadotrope-enriched marker, Foxp2. We present novel scRNAseq data of in vivo pituitary tissue, including data from agnostic clustering algorithms that suggest the presence of a somatotrope subpopulation enriched in sterol/cholesterol synthesis genes. Additionally, we show that incomplete transcriptome annotation can cause false negatives on some scRNAseq platforms that only generate 3' transcript end sequences, and we use in vivo data to recover reads of the pituitary transcription factor Prop1. Ultimately, scRNAseq technologies represent a significant opportunity to address long-standing questions regarding the development and function of the different populations of the pituitary gland throughout life.


Assuntos
Biomarcadores/análise , Hipófise/citologia , Hipófise/metabolismo , Hormônios Hipofisários/metabolismo , Análise de Célula Única/métodos , Células-Tronco/classificação , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular/genética , Separação Celular/métodos , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Organogênese/genética , Hipófise/embriologia , Hormônios Hipofisários/genética , Análise de Sequência de RNA/métodos , Células-Tronco/citologia , Transcriptoma
14.
Endocrinology ; 159(9): 3287-3305, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30085028

RESUMO

The pituitary gland is a critical organ that is necessary for many physiological processes, including growth, reproduction, and stress response. The secretion of pituitary hormones from specific cell types regulates these essential processes. Pituitary hormone cell types arise from a common pool of pituitary progenitors, and mutations that disrupt the formation and differentiation of pituitary progenitors result in hypopituitarism. Canonical WNT signaling through CTNNB1 (ß-catenin) is known to regulate the formation of the POU1F1 lineage of pituitary cell types. When ß-catenin is deleted during the initial formation of the pituitary progenitors, Pou1f1 is not transcribed, which leads to the loss of the POU1F1 lineage. However, when ß-catenin is deleted after lineage specification, there is no observable effect. Similarly, the generation of a ß-catenin gain-of-function allele in early pituitary progenitors or stem cells results in the formation of craniopharyngiomas, whereas stimulating ß-catenin in differentiated cell types has no effect. PROP1 is a pituitary-specific transcription factor, and the peak of PROP1 expression coincides with a critical time point in pituitary organogenesis-that is, after pituitary progenitor formation but before lineage specification. We used a Prop1-cre to conduct both loss- and gain-of-function studies on ß-catenin during this critical time point. Our results demonstrate that pituitary progenitors remain sensitive to both loss and gain of ß-catenin at this time point, and that either manipulation results in hypopituitarism.


Assuntos
Craniofaringioma/genética , Hipopituitarismo/genética , Hipófise/embriologia , Células-Tronco/metabolismo , Fator de Transcrição Pit-1/genética , beta Catenina/genética , Animais , Linhagem da Célula , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Lactotrofos/citologia , Camundongos , Organogênese , Hipófise/metabolismo , Somatotrofos/citologia , Células-Tronco/citologia , Tireotrofos/citologia , Fator de Transcrição Pit-1/metabolismo , Via de Sinalização Wnt
15.
Best Pract Res Clin Obstet Gynaecol ; 52: 114-125, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29724668

RESUMO

Preterm birth (PTB) is the leading cause of neonatal morbidity and mortality worldwide. The only medicinal therapy currently recommended to prevent PTB is prophylactic progestin therapy in the form of micronized progesterone (P4) administered daily via vaginal suppository from the 24th to the 34th week of gestation or 17α-hydroxyprogesterone caproate in oil administered weekly from the 16th to the 36th week of gestation via an intramuscular injection. These therapies decrease the risk of PTB in women with an elevated risk of PTB indicated by a history of PTB or by a short cervix measured by sonography at mid-gestation. The mechanism by which progestin therapy prevents PTB in some women is not clear but may involve non-progestin mechanism and/or supplementation of localized progestin deficiency. Advances in understanding the molecular biology and physiology of P4 signaling via the P4 receptor isoforms in uterine cells reveal novel therapeutic targets; this may improve the effectiveness of progestin therapy to prevent PTB in the majority of pregnancies by targeting key steps in the pathway leading to inflammation-induced parturition.


Assuntos
Caproato de 17 alfa-Hidroxiprogesterona/administração & dosagem , Nascimento Prematuro/prevenção & controle , Progestinas/administração & dosagem , Caproato de 17 alfa-Hidroxiprogesterona/farmacologia , Administração Intravaginal , Medida do Comprimento Cervical , Ensaios Clínicos como Assunto , Feminino , Humanos , Injeções Intramusculares , Hipófise/efeitos dos fármacos , Hipófise/embriologia , Gravidez , Nascimento Prematuro/metabolismo , Progestinas/farmacologia , Útero/efeitos dos fármacos
16.
Biol Reprod ; 99(3): 504-513, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29757344

RESUMO

The coordination of pituitary development is complicated and requires input from multiple cellular processes. Recent research has provided insight into key molecular determinants that govern cell fate specification in the pituitary. Moreover, increasing research aimed to identify, characterize, and functionally describe the presumptive pituitary stem cell population has allowed for a better understanding of the processes that govern endocrine cell differentiation in the developing pituitary. The culmination of this research has led to the ability of investigators to recapitulate some of embryonic pituitary development in vitro, the first steps to developing novel regenerative therapies for pituitary diseases. In this current review, we cover the major players in pituitary stem/progenitor cell function and maintenance, and the key molecular determinants of endocrine cell specification. In addition, we discuss the contribution of peripheral hormonal regulation of pituitary gland development, an understudied area of research.


Assuntos
Hipófise/embriologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular , Feminino , Expressão Gênica , Gonadotropinas Hipofisárias/biossíntese , Hormônio do Crescimento/biossíntese , Humanos , Camundongos , Células-Tronco Multipotentes/citologia , Hipófise/citologia , Gravidez , Prolactina/biossíntese , Células-Tronco/citologia , Tireotropina/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
17.
Endocrinology ; 159(8): 2891-2904, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29796621

RESUMO

The process by which the somatotrope lineage emerges in the developing pituitary is regulated by the activity of specific signaling and transcription factors expressed during development. We set out to understand the contribution of FOXO1 to that process by using a mouse model in which FOXO1 is prematurely expressed in the pituitary primordium. Expression of FOXO1 in the oral ectoderm as early as embryonic day (e)9.5 resulted in pituitary gland hypoplasia and reduced expression of anterior lobe hormone transcripts at e18.5. Of note, the relative numbers of somatotropes and thyrotropes were also decreased at e18.5. LHX3 and PITX2, markers of pituitary identity, were present in a reduced number of cells during the formation of the Rathke pouch. Thus, premature expression of FOXO1 may affect adoption of pituitary identity during differentiation. Our results demonstrate that the timing of FOXO1 activation affects its role in pituitary gland organogenesis and somatotrope differentiation.


Assuntos
Proteína Forkhead Box O1/genética , Regulação da Expressão Gênica no Desenvolvimento , Organogênese/genética , Adeno-Hipófise/embriologia , Animais , Diferenciação Celular/genética , Linhagem da Célula , Ectoderma/embriologia , Ectoderma/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Tamanho do Órgão , Hipófise/citologia , Hipófise/embriologia , Hipófise/metabolismo , Hipófise/patologia , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Adeno-Hipófise/patologia , Somatotrofos/citologia , Somatotrofos/metabolismo , Tireotrofos/citologia , Tireotrofos/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Proteína Homeobox PITX2
18.
Acta Neuropathol ; 135(5): 757-777, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29541918

RESUMO

Adamantinomatous craniopharyngiomas (ACPs) are clinically challenging tumours, the majority of which have activating mutations in CTNNB1. They are histologically complex, showing cystic and solid components, the latter comprised of different morphological cell types (e.g. ß-catenin-accumulating cluster cells and palisading epithelium), surrounded by a florid glial reaction with immune cells. Here, we have carried out RNA sequencing on 18 ACP samples and integrated these data with an existing ACP transcriptomic dataset. No studies so far have examined the patterns of gene expression within the different cellular compartments of the tumour. To achieve this goal, we have combined laser capture microdissection with computational analyses to reveal groups of genes that are associated with either epithelial tumour cells (clusters and palisading epithelium), glial tissue or immune infiltrate. We use these human ACP molecular signatures and RNA-Seq data from two ACP mouse models to reveal that cell clusters are molecularly analogous to the enamel knot, a critical signalling centre controlling normal tooth morphogenesis. Supporting this finding, we show that human cluster cells express high levels of several members of the FGF, TGFB and BMP families of secreted factors, which signal to neighbouring cells as evidenced by immunostaining against the phosphorylated proteins pERK1/2, pSMAD3 and pSMAD1/5/9 in both human and mouse ACP. We reveal that inhibiting the MAPK/ERK pathway with trametinib, a clinically approved MEK inhibitor, results in reduced proliferation and increased apoptosis in explant cultures of human and mouse ACP. Finally, we analyse a prominent molecular signature in the glial reactive tissue to characterise the inflammatory microenvironment and uncover the activation of inflammasomes in human ACP. We validate these results by immunostaining against immune cell markers, cytokine ELISA and proteome analysis in both solid tumour and cystic fluid from ACP patients. Our data support a new molecular paradigm for understanding ACP tumorigenesis as an aberrant mimic of natural tooth development and opens new therapeutic opportunities by revealing the activation of the MAPK/ERK and inflammasome pathways in human ACP.


Assuntos
Craniofaringioma/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias Hipofisárias/metabolismo , Transcriptoma , Microambiente Tumoral/fisiologia , Animais , Biologia Computacional , Craniofaringioma/patologia , Craniofaringioma/terapia , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Inflamação/terapia , Microdissecção e Captura a Laser , Camundongos , Neuroglia/metabolismo , Odontogênese/fisiologia , Hipófise/embriologia , Hipófise/patologia , Neoplasias Hipofisárias/patologia , Neoplasias Hipofisárias/terapia , Análise de Sequência de RNA , Técnicas de Cultura de Tecidos
19.
Nat Commun ; 9(1): 345, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29367605

RESUMO

Noisy gene expression is of fundamental importance to single cells, and is therefore widely studied in single-celled organisms. Extending these studies to multicellular organisms is challenging since their cells are generally not isolated, but individuals in a tissue. Cell-cell coupling via signalling, active transport or pure diffusion, ensures that tissue-bound cells are neither fully independent of each other, nor an entirely homogeneous population. In this article, we show that increasing the strength of coupling between cells can either increase or decrease the single-cell variability (and, therefore, the heterogeneity of the tissue), depending on the statistical properties of the underlying genetic network. We confirm these predictions using spatial stochastic simulations of simple genetic networks, and experimental data from animal and plant tissues. The results suggest that cell-cell coupling may be one of several noise-control strategies employed by multicellular organisms, and highlight the need for a deeper understanding of multicellular behaviour.


Assuntos
Arabidopsis/citologia , Comunicação Celular , Redes Reguladoras de Genes , Modelos Biológicos , Hipófise/citologia , Animais , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Hipófise/embriologia , Hipófise/fisiologia , Folhas de Planta/citologia , Folhas de Planta/genética , Ratos , Processos Estocásticos
20.
J Neuroendocrinol ; 30(3): e12570, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29356182

RESUMO

Development of the anterior pituitary proceeds via spatiotemporal patterning of transcription factors and signalling molecules. Among them, retinoic acid (RA) functions as an important signalling molecule for vertebrate organogenesis in many tissues. However, little is known regarding the target genes in the developing pituitary. The present study aimed to clarify the relationship between endogenous RA signalling and mRNA expression of the pituitary-specific transcription factor Prop1 in the pituitary primordium of Rathke's pouch. Gene expression analysis and in situ hybridisation demonstrated that retinaldehyde dehydrogenases (Raldhs) and all types of RA receptors (Rars) are expressed at the level of transcription in the rat Rathke's pouch. Ex vivo organ culture using Rathke's pouch and an in vitro reporter assay demonstrated that RA signalling increases the expression level of Prop1 via RARα. Moreover, a reporter assay using serial truncated constructs of the 5'-upstream region of mouse Prop1 revealed a predicted cis-regulatory element of RARα. This is the first report of a relationship between RA signalling and Prop1-expression during early pituitary development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Hipófise/metabolismo , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Animais , Proteínas de Homeodomínio/genética , Camundongos , Hipófise/embriologia , Ratos , Ratos Wistar , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...