Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 435
Filtrar
1.
Endocr J ; 71(6): 547-559, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38346768

RESUMO

The pituitary gland is endocrine tissue composed of two distinct parts with different origins: the adenohypophysis (adenohypophyseal placode origin) and the neurohypophysis (neuroectoderm origin). Differentiation of endocrine cells in the pituitary gland leads to hormone synthesis, secretion into the capillary network, and transportation to target organs. In 1988, the discovery of the pituitary transcription factor PIT1 sparked research on endocrine cell differentiation. In the twenty-first century, the discovery that SOX2-positive stem/progenitor cells give rise to all types of pituitary endocrine cells advanced research on differentiation processes using diverse marker molecules. Lineage tracing using specific marker genes from early embryos revealed that during construction of the anterior pituitary from the adenohypophyseal placodal cells the developing anterior pituitary incorporates diverse cell types originating from the neural crest-derived and ectodermal-derived cells. Consequently, the postnatal anterior pituitary becomes a mosaic of terminally differentiated cells of different origin and with different life histories. It has also been revealed that most of the postnatal stem/progenitor cells form at least solid clusters in the parenchyma. Moreover, the classification and role of S100ß-positive cells had been ambiguous, but now they are identified as a major component of postnatal stem/progenitor cells. This paper provides an updated overview of pituitary development.


Assuntos
Diferenciação Celular , Linhagem da Célula , Adeno-Hipófise , Células-Tronco , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Humanos , Animais , Células-Tronco/fisiologia , Células-Tronco/citologia
2.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34958103

RESUMO

The specific role of gonadotropin-releasing hormone (GnRH) on brain sexual differentiation remains unclear. To investigate whether gonadotropin and, in turn, testosterone (T) secretion is regulated by GnRH during the critical period for brain differentiation in sheep fetuses, we attempted to selectively suppress pituitary-testicular activation during midgestation with the long-acting GnRH antagonist degarelix. Fetuses received subcutaneous injections of the antagonist or vehicle on day 62 of gestation. After 2 to 3 weeks we examined consequences of the intervention on baseline and GnRH-stimulated plasma luteinizing hormone (LH) and T levels. In addition, we measured the effect of degarelix-treatment on messenger RNA (mRNA) expression for the pituitary gonadotropins and key gonadal steroidogenic enzymes. Baseline and GnRH-stimulated plasma LH levels were significantly suppressed in degarelix-treated male and female fetuses compared to control values. Similarly, T concentrations were suppressed in degarelix-treated males. The percentage of LHß-immunoreactive cells colocalizing c-fos was significantly reduced by degarelix treatment indicating that pituitary sensitivity was inhibited. Degarelix treatment also led to the significant suppression of mRNA expression coding for the pituitary gonadotropin subunits and for the gonadal enzymes involved in androgen synthesis. These findings demonstrate that pharmacologic inhibition of GnRH early in gestation results in suppression of LH secretion and deficits in the plasma T levels of male lamb fetuses. We conclude that GnRH signaling plays a pivotal role for regulating T exposure during the critical period of sheep gestation when the brain is masculinized. Thus, disturbance to gonadotropin secretion during this phase of gestation could have long-term consequence on adult sexual behaviors and fertility.


Assuntos
Idade Gestacional , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Gonadotropinas Hipofisárias/metabolismo , Oligopeptídeos/administração & dosagem , Adeno-Hipófise/embriologia , Ovinos/embriologia , Animais , Encéfalo/embriologia , Feminino , Sangue Fetal/química , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/fisiologia , Gonadotropinas Hipofisárias/genética , Injeções Subcutâneas/veterinária , Hormônio Luteinizante/sangue , Masculino , Ovário/química , Ovário/embriologia , Adeno-Hipófise/química , Adeno-Hipófise/efeitos dos fármacos , Gravidez , RNA Mensageiro/análise , Diferenciação Sexual/fisiologia , Testículo/química , Testículo/embriologia , Testosterona/sangue
3.
Curr Top Dev Biol ; 141: 207-239, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33602489

RESUMO

Modern vertebrates consist of two sister groups: cyclostomes and gnathostomes. Cyclostomes are a monophyletic jawless group that can be further divided into hagfishes and lampreys, which show conspicuously different developmental and morphological patterns. However, during early pharyngula development, there appears to be a stage when the embryos of hagfishes and lampreys resemble each other by showing an "ancestral" craniofacial pattern; this pattern enables morphological comparison of hagfish and lamprey craniofacial development at late stages. This cyclostome developmental pattern, or more accurately, this developmental pattern of the jawless grade of vertebrates in early pharyngula was very likely shared by the gnathostome stem before the division of the nasohypophyseal placode led to the jaw and paired nostrils. The craniofacial pattern of the modern jawed vertebrates seems to have begun in fossil ostracoderms (including galeaspids), and was completed by the early placoderm lineages. The transition from jawless to jawed vertebrates was thus driven by heterotopy of development, mainly caused by separation and shift of ectodermal placodes and resultant ectomesenchymal distribution, and shifts of the epithelial-mesenchymal interactions that underlie craniofacial differentiation. Thus, the evolution of the jaw was not a simple modification of the mandibular arch, but a heterotopic shift of the developmental interactions involving not only the mandibular arch, but also the premandibular region rostral to the mandibular arch.


Assuntos
Evolução Biológica , Feiticeiras (Peixe)/anatomia & histologia , Lampreias/anatomia & histologia , Vertebrados , Animais , Embrião não Mamífero , Fósseis , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Feiticeiras (Peixe)/embriologia , Lampreias/embriologia , Adeno-Hipófise/embriologia , Crânio/anatomia & histologia , Vertebrados/anatomia & histologia , Vertebrados/embriologia
4.
Nat Commun ; 11(1): 5275, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-33077725

RESUMO

The anterior pituitary gland plays a central role in regulating various physiological processes, including body growth, reproduction, metabolism and stress response. Here, we perform single-cell RNA-sequencing (scRNA-seq) of 4113 individual cells from human fetal pituitaries. We characterize divergent developmental trajectories with distinct transitional intermediate states in five hormone-producing cell lineages. Corticotropes exhibit an early intermediate state prior to full differentiation. Three cell types of the PIT-1 lineage (somatotropes, lactotropes and thyrotropes) segregate from a common progenitor coexpressing lineage-specific transcription factors of different sublineages. Gonadotropes experience two multistep developmental trajectories. Furthermore, we identify a fetal gonadotrope cell subtype expressing the primate-specific hormone chorionic gonadotropin. We also characterize the cellular heterogeneity of pituitary stem cells and identify a hybrid epithelial/mesenchymal state and an early-to-late state transition. Here, our results provide insights into the transcriptional landscape of human pituitary development, defining distinct cell substates and subtypes and illustrating transcription factor dynamics during cell fate commitment.


Assuntos
Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , Transcriptoma , Diferenciação Celular , Células Cultivadas , Feminino , Feto/embriologia , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gonadotrofos/citologia , Gonadotrofos/metabolismo , Gonadotropinas/metabolismo , Humanos , Masculino , Adeno-Hipófise/citologia , Proteínas/genética , Proteínas/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Science ; 370(6515): 463-467, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33093109

RESUMO

Vertebrate sensory organs arise from epithelial thickenings called placodes. Along with neural crest cells, cranial placodes are considered ectodermal novelties that drove evolution of the vertebrate head. The anterior-most placode generates the endocrine lobe [adenohypophysis (ADH)] of the pituitary, a master gland controlling growth, metabolism, and reproduction. In addition to known ectodermal contributions, we use lineage tracing and time-lapse imaging in zebrafish to identify an endodermal contribution to the ADH. Single-cell RNA sequencing of the adult pituitary reveals similar competency of endodermal and ectodermal epithelia to generate all endocrine cell types. Further, endoderm can generate a rudimentary ADH-like structure in the near absence of ectodermal contributions. The fish condition supports the vertebrate pituitary arising through interactions of an ancestral endoderm-derived proto-pituitary with newly evolved placodal ectoderm.


Assuntos
Endoderma/embriologia , Adeno-Hipófise/embriologia , Animais , Linhagem da Célula , Endoderma/citologia , Adeno-Hipófise/citologia , RNA-Seq , Análise de Célula Única , Peixe-Zebra
6.
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
7.
Development ; 144(19): 3521-3532, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28974641

RESUMO

Placodes are discrete thickenings of the vertebrate cranial ectoderm that generate morpho-functionally distinct structures, such as the adenohypophysis, olfactory epithelium and lens. All placodes arise from a horseshoe-shaped preplacodal ectoderm in which the precursors of individual placodes are intermingled. However, fate-map studies indicated that cells positioned at the preplacodal midline give rise to only the adenohypophyseal placode, suggesting a unique organization of these precursors within the preplacode. To test this possibility, we combined embryological and molecular approaches in chick embryos to show that, at gastrula stage, adenohypophyseal precursors are clustered in the median preplacodal ectoderm, largely segregated from those of the adjacent olfactory placode. Median precursors are elongated, densely packed and, at neurula stage, express a molecular signature that distinguishes them from the remaining preplacodal cells. Olfactory placode precursors and midline neural cells can replace ablated adenohypophyseal precursors up to head-fold stage, although with a more plastic organization. We thus propose that adenohypophyseal placode precursors are unique within the preplacodal ectoderm possibly because they originate the only single placode and the only one with an endocrine character.


Assuntos
Ectoderma/embriologia , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Células-Tronco/citologia , Animais , Padronização Corporal , Forma Celular , Embrião de Galinha , Ectoderma/citologia , Cristalino/citologia , Cristalino/embriologia , Mucosa Olfatória/citologia , Mucosa Olfatória/embriologia
8.
Oncotarget ; 8(22): 36553-36565, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28402262

RESUMO

Follicle-stimulating hormone (FSH) secreted by adenohypophyseal cells plays an important role in the regulation of reproduction, but whether microRNAs (miRNAs) regulate the secretion of FSH remains unclear. In the present study, we predicted and screened miRNAs that might act on the follicle-stimulating hormone beta-subunit (FSHb) gene of rats using the TargetScan program and luciferase reporter assays, and the results identified two miRNAs, miR-21-3p and miR-433. We then transfected these miRNAs into rat anterior adenohypophyseal cells and assessed the FSHb expression levels in and FSH secretion by the transfected cells through quantitative PCR and ELISA. The results showed that both miR-21-3p and miR-433 down-regulated the expression levels of FSHb and resulted in the decrease of the secretion of FSH compared with the control group, and treatment with miR-21-3p and miR-433 inhibitors up-regulated the expression levels of FSHb and resulted in the increase of the secretion of FSH. Taken together, our results indicate that miR-21-3p and miR-433 can down-regulate the expression of FSHb by directly targeting the FSHb 3'UTR in rat primary pituitary cells. Our findings provide evidence that miRNAs can regulate FSHb expression and further affect the secretion of FSH and might contribute to the use of miRNAs for the regulation of animal reproduction.


Assuntos
Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica/efeitos dos fármacos , MicroRNAs/genética , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Regiões 3' não Traduzidas , Animais , Células Cultivadas , Hormônio Foliculoestimulante/metabolismo , Perfilação da Expressão Gênica , Masculino , Adeno-Hipófise/embriologia , Interferência de RNA , Ratos , Reprodutibilidade dos Testes , Transfecção
9.
Nat Commun ; 7: 11121, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27079169

RESUMO

The anterior pituitary harbours five distinct hormone-producing cell types, and their cellular differentiation is a highly regulated and coordinated process. Here we show that ZBTB20 is essential for anterior pituitary development and lactotrope specification in mice. In anterior pituitary, ZBTB20 is highly expressed by all the mature endocrine cell types, and to some less extent by somatolactotropes, the precursors of prolactin (PRL)-producing lactotropes. Disruption of Zbtb20 leads to anterior pituitary hypoplasia, hypopituitary dwarfism and a complete loss of mature lactotropes. In ZBTB20-null mice, although lactotrope lineage commitment is normally initiated, somatolactotropes exhibit profound defects in lineage specification and expansion. Furthermore, endogenous ZBTB20 protein binds to Prl promoter, and its knockdown decreases PRL expression and secretion in a lactotrope cell line MMQ. In addition, ZBTB20 overexpression enhances the transcriptional activity of Prl promoter in vitro. In conclusion, our findings point to ZBTB20 as a critical regulator of anterior pituitary development and lactotrope specification.


Assuntos
Linhagem da Célula/genética , Lactotrofos/metabolismo , Adeno-Hipófise/metabolismo , Fatores de Transcrição/genética , Animais , Western Blotting , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Hipotálamo/embriologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Lactotrofos/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Adeno-Hipófise/embriologia , Adeno-Hipófise/crescimento & desenvolvimento , Prolactina/genética , Prolactina/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
10.
Endocrinology ; 157(4): 1385-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26812162

RESUMO

Mutations in PROP1, the most common known cause of combined pituitary hormone deficiency in humans, can result in the progressive loss of all hormones of the pituitary anterior lobe. In mice, Prop1 mutations result in the failure to initiate transcription of Pou1f1 (also known as Pit1) and lack somatotropins, lactotropins, and thyrotropins. The basis for this species difference is unknown. We hypothesized that Prop1 is expressed in a progenitor cell that can develop into all anterior lobe cell types, and not just the somatotropes, thyrotropes, and lactotropes, which are collectively known as the PIT1 lineage. To test this idea, we produced a transgenic Prop1-cre mouse line and conducted lineage-tracing experiments of Prop1-expressing cells. The results reveal that all hormone-secreting cell types of both the anterior and intermediate lobes are descended from Prop1-expressing progenitors. The Prop1-cre mice also provide a valuable genetic reagent with a unique spatial and temporal expression for generating tissue-specific gene rearrangements early in pituitary gland development. We also determined that the minimal essential sequences for reliable Prop1 expression lie within 10 kilobases of the mouse gene and demonstrated that human PROP1 can substitute functionally for mouse Prop1. These studies enhance our understanding of the pathophysiology of disease in patients with PROP1 mutations.


Assuntos
Proteínas de Homeodomínio/metabolismo , Adeno-Hipófise/metabolismo , Hipófise/metabolismo , Células-Tronco/metabolismo , Animais , Linhagem da Célula/genética , Teste de Complementação Genética , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Mutação , Hipófise/citologia , Hipófise/embriologia , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia
11.
Nat Commun ; 7: 10351, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26762480

RESUMO

Anterior pituitary is critical for endocrine systems. Its hormonal responses to positive and negative regulators are indispensable for homeostasis. For this reason, generating human anterior pituitary tissue that retains regulatory hormonal control in vitro is an important step for the development of cell transplantation therapy for pituitary diseases. Here we achieve this by recapitulating mouse pituitary development using human embryonic stem cells. We find that anterior pituitary self-forms in vitro following the co-induction of hypothalamic and oral ectoderm. The juxtaposition of these tissues facilitated the formation of pituitary placode, which subsequently differentiated into pituitary hormone-producing cells. They responded normally to both releasing and feedback signals. In addition, after transplantation into hypopituitary mice, the in vitro-generated corticotrophs rescued physical activity levels and survival of the hosts. Thus, we report a useful methodology for the production of regulator-responsive human pituitary tissue that may benefit future studies in regenerative medicine.


Assuntos
Corticotrofos/transplante , Células-Tronco Embrionárias/fisiologia , Hipopituitarismo/terapia , Adeno-Hipófise/embriologia , Engenharia Tecidual/métodos , Hormônio Adrenocorticotrópico/metabolismo , Animais , Técnicas de Cultura de Células , Desenvolvimento Embrionário , Hormônio do Crescimento/metabolismo , Humanos , Camundongos , Adeno-Hipófise/metabolismo
12.
Endocrinology ; 156(8): 2934-48, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25965960

RESUMO

Dopaminergic (DA) neurons located in the preoptico-hypothalamic region of the brain exert a major neuroendocrine control on reproduction, growth, and homeostasis by regulating the secretion of anterior pituitary (or adenohypophysis) hormones. Here, using a retrograde tract tracing experiment, we identified the neurons playing this role in the zebrafish. The DA cells projecting directly to the anterior pituitary are localized in the most anteroventral part of the preoptic area, and we named them preoptico-hypophyseal DA (POHDA) neurons. During development, these neurons do not appear before 72 hours postfertilization (hpf) and are the last dopaminergic cell group to differentiate. We found that the number of neurons in this cell population continues to increase throughout life proportionally to the growth of the fish. 5-Bromo-2'-deoxyuridine incorporation analysis suggested that this increase is due to continuous neurogenesis and not due to a phenotypic change in already-existing neurons. Finally, expression profiles of several genes (foxg1a, dlx2a, and nr4a2a/b) were different in the POHDA compared with the adjacent suprachiasmatic DA neurons, suggesting that POHDA neurons develop as a distinct DA cell population in the preoptic area. This study offers some insights into the regional identity of the preoptic area and provides the first bases for future functional genetic studies on the development of DA neurons controlling anterior pituitary functions.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Neurogênese/fisiologia , Adeno-Hipófise/fisiologia , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Neurônios Dopaminérgicos/citologia , Embrião não Mamífero , Feminino , Sistemas Neurossecretores/citologia , Sistemas Neurossecretores/crescimento & desenvolvimento , Adeno-Hipófise/embriologia , Adeno-Hipófise/crescimento & desenvolvimento , Hormônios Adeno-Hipofisários/metabolismo , Área Pré-Óptica/embriologia , Área Pré-Óptica/crescimento & desenvolvimento , Peixe-Zebra/embriologia , Peixe-Zebra/genética
13.
Neuroendocrinology ; 101(1): 18-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25428763

RESUMO

The hypothalamic-pituitary system is essential to maintain life and control systemic homeostasis, but it is negatively affected by various diseases, leading to serious symptoms. Embryonic stem (ES) cells differentiate into neuroectodermal progenitors when cultured as floating aggregates under serum-free conditions. Recently, our colleagues have shown that strict removal of exogenous patterning factors during early differentiation steps induced efficient generation of rostral hypothalamic-like progenitors from mouse ES cell-derived neuroectodermal cells. The use of growth factor-free chemically defined medium was critical for this induction. The ES cell-derived hypothalamic-like progenitors generated rostral-dorsal hypothalamic neurons, especially magnocellular vasopressinergic neurons that release the hormone upon stimulation. Subsequently, we reported efficient self-formation of 3-dimensional adenohypophysis tissues in aggregate cultures of mouse ES cells. The ES cells were stimulated to differentiate into nonneural head ectoderm and hypothalamic neuroectoderm in adjacent layers within the aggregate and then treated with hedgehog. Self-organization of Rathke's pouch-like structures occurred at the interface of the two epithelia, as observed in vivo, and various endocrine cells including corticotrophs and somatotrophs were subsequently produced. The corticotrophs efficiently secreted adrenocorticotropic hormone in response to corticotropin-releasing hormone. Furthermore, when engrafted in vivo, these cells rescued the systemic glucocorticoid level in hypopituitary mice. Our present research aims are to prepare hypothalamic and pituitary tissues from human induced pluripotent stem cells and establish effective transplantation techniques with clinical applications. To replicate the complex and precise control of the hypothalamic-pituitary system, regenerative medicine using pluripotent cells may be a hopeful option.


Assuntos
Diferenciação Celular , Sistema Hipotálamo-Hipofisário/embriologia , Sistema Hipotálamo-Hipofisário/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Técnicas de Cultura de Células , Humanos , Sistema Hipotálamo-Hipofisário/citologia , Camundongos , Neurônios/citologia , Neurônios/fisiologia , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Adeno-Hipófise/fisiologia
14.
Hum Mol Genet ; 24(4): 939-53, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25315894

RESUMO

OTX2 is a homeodomain transcription factor that is necessary for normal head development in mouse and man. Heterozygosity for loss-of-function alleles causes an incompletely penetrant, haploinsufficiency disorder. Affected individuals exhibit a spectrum of features that range from developmental defects in eye and/or pituitary development to acephaly. To investigate the mechanism underlying the pituitary defects, we used different cre lines to inactivate Otx2 in early head development and in the prospective anterior and posterior lobes. Mice homozygous for Otx2 deficiency in early head development and pituitary oral ectoderm exhibit craniofacial defects and pituitary gland dysmorphology, but normal pituitary cell specification. The morphological defects mimic those observed in humans and mice with OTX2 heterozygous mutations. Mice homozygous for Otx2 deficiency in the pituitary neural ectoderm exhibited altered patterning of gene expression and ablation of FGF signaling. The posterior pituitary lobe and stalk, which normally arise from neural ectoderm, were extremely hypoplastic. Otx2 expression was intact in Rathke's pouch, the precursor to the anterior lobe, but the anterior lobe was hypoplastic. The lack of FGF signaling from the neural ectoderm was sufficient to impair anterior lobe growth, but not the differentiation of hormone-producing cells. This study demonstrates that Otx2 expression in the neural ectoderm is important intrinsically for the development of the posterior lobe and pituitary stalk, and it has significant extrinsic effects on anterior pituitary growth. Otx2 expression early in head development is important for establishing normal craniofacial features including development of the brain, eyes and pituitary gland.


Assuntos
Ectoderma/embriologia , Ectoderma/metabolismo , Deleção de Genes , Organogênese/genética , Fatores de Transcrição Otx/genética , Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , Animais , Proliferação de Células , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Mutação , Fatores de Transcrição Otx/metabolismo , Fenótipo , Adeno-Hipófise/patologia , Transdução de Sinais
15.
Endocrinology ; 155(1): 33-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24189142

RESUMO

Somatostatin (SRIF), by acting mainly through sst2 and sst5 receptors, is a potent inhibitor of hormonal secretion by the human anterior pituitary gland. However, the pattern of protein expression of these SRIF receptors remains unknown during pituitary development. To get further insights into the physiological role of SRIF receptors in human development and pituitary function, the present study examined the developmental expression of the sst2 and sst5 receptors in the individual cell types of the anterior human pituitary. Thirteen fetal human pituitaries were investigated between 13 to 38 weeks of gestation (WG) by double-labeling immunofluorescence with antibodies raised against sst2 or sst5 receptors and GH, LH, FSH, TSH, or pro-opiomelanocortin proteins. SRIF immunoreactivity in the hypothalamus and median eminence was investigated at the same developmental ages. Immunoreactivity for the sst2 receptor was evident as early as 13 to 15 WG and onward mainly in TSH-, LH-, and FSH-expressing cells, whereas sst5 immunoreactivity was apparent at the late development stages (35-38 WG). GH-expressing cells mainly expressed sst5 immunoreactivity. SRIF-positive fibers and cells were detected as soon as 13 to 16 WG in the hypothalamus and median eminence and their densities increased with gestational age. The early appearance of hypothalamic SRIF cells and fibers suggests a physiological link between SRIF and its receptors during pituitary development. Whereas sst2 receptors might play a primary role in the differentiation and regulation of TSH, LH, and FSH cells, sst5 receptors appear to be mainly involved in GH regulation from birth onward.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Adeno-Hipófise/embriologia , Receptores de Somatostatina/metabolismo , Diferenciação Celular , Retroalimentação Fisiológica , Feminino , Perfilação da Expressão Gênica , Idade Gestacional , Humanos , Masculino , Microscopia de Fluorescência , Fatores de Tempo
16.
Fish Physiol Biochem ; 40(3): 897-909, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24310491

RESUMO

In this study, we describe for the first time the details of the pituitary gland morphogenesis and the ontogeny of adenohypophyseal cells of a South American Characiform species with great importance for Brazilian Aquaculture, Salminus brasiliensis (Characiformes, Characidae), from hatching to 25 days after hatching (dah), by histochemical and immunocytochemical methods. The pituitary placode was first detected at hatching (0 dah), and the pituitary anlage became more defined at 0.5 dah. The neurohypophysis (NH) development started at 3 dah, and the early formation of its stalk at 12.5 dah. An increase in adenohypophyseal and NH tissues was also observed, and in juveniles at 25 dah, the pituitary displayed similar morphology to that found in adults of this species, displaying the main features of the teleost pituitary. PRL cells were detected at 0.5 dah, together with ACTH and α-MSH cells, followed by GH and SL cells at 1.5 dah. ß-FSH cells were detected at 25 dah, while ß-LH cells at 5 dah. The pituitary development in this species comprises a dynamic process similar to other teleosts. Our findings in S. brasiliensis corroborate the heterogeneity in the ontogeny of adenohypophyseal cells in teleosts and suggest a role for adenohypophyseal hormones in the early development of this species.


Assuntos
Characidae/embriologia , Adeno-Hipófise/embriologia , Animais , Characidae/crescimento & desenvolvimento , Organogênese , Adeno-Hipófise/citologia
17.
Int J Dev Biol ; 57(9-10): 753-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24307294

RESUMO

Cranial ectodermal placodes, a vertebrate innovation, contribute to the adenohypophysis and peripheral nervous system of the head, including the paired sense organs (eyes, nose, ears) and sensory ganglia of the Vth, VIIth, IXth and Xth cranial nerves. Fate-maps of groups of cells in amphibians, teleosts and amniotes have demonstrated that all placodes have a common origin in a horseshoe shaped territory, known as the preplacodal region (PPR), which surrounds the presumptive neural plate of the late gastrula/early neurula stage embryo. Given the extensive regional overlap of progenitors for different placodes in the chick embryo, it has been a matter of debate as to whether individual cells in the PPR are truly multipotent progenitors, with regard to placodal identity, or rather are lineage-biased or restricted to a specific placodal type prior to overt differentiation. Utilizing clonal analyses in vivo, we demonstrate here that the anterior PPR comprises some precursors that contribute either to the olfactory or lens placode well before they are spatially segregated or committed to either of these placodal fates. This suggests that lineage bias towards a specific placodal fate may coincide with induction of the PPR.


Assuntos
Evolução Clonal/fisiologia , Cristalino/embriologia , Mucosa Olfatória/embriologia , Sistema Nervoso Periférico/embriologia , Animais , Embrião de Galinha , Ectoderma/citologia , Ectoderma/embriologia , Gástrula/embriologia , Gástrula/inervação , Cabeça/embriologia , Crista Neural/citologia , Crista Neural/embriologia , Placa Neural/embriologia , Neurulação/fisiologia , Adeno-Hipófise/embriologia , Coloração e Rotulagem
18.
Development ; 140(24): 4947-58, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24227653

RESUMO

The Insm1 gene encodes a zinc finger factor expressed in many endocrine organs. We show here that Insm1 is required for differentiation of all endocrine cells in the pituitary. Thus, in Insm1 mutant mice, hormones characteristic of the different pituitary cell types (thyroid-stimulating hormone, follicle-stimulating hormone, melanocyte-stimulating hormone, adrenocorticotrope hormone, growth hormone and prolactin) are absent or produced at markedly reduced levels. This differentiation deficit is accompanied by upregulated expression of components of the Notch signaling pathway, and by prolonged expression of progenitor markers, such as Sox2. Furthermore, skeletal muscle-specific genes are ectopically expressed in endocrine cells, indicating that Insm1 participates in the repression of an inappropriate gene expression program. Because Insm1 is also essential for differentiation of endocrine cells in the pancreas, intestine and adrenal gland, it is emerging as a transcription factor that acts in a pan-endocrine manner. The Insm1 factor contains a SNAG domain at its N-terminus, and we show here that the SNAG domain recruits histone-modifying factors (Kdm1a, Hdac1/2 and Rcor1-3) and other proteins implicated in transcriptional regulation (Hmg20a/b and Gse1). Deletion of sequences encoding the SNAG domain in mice disrupted differentiation of pituitary endocrine cells, and resulted in an upregulated expression of components of the Notch signaling pathway and ectopic expression of skeletal muscle-specific genes. Our work demonstrates that Insm1 acts in the epigenetic and transcriptional network that controls differentiation of endocrine cells in the anterior pituitary gland, and that it requires the SNAG domain to exert this function in vivo.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Endócrinas/metabolismo , Adeno-Hipófise/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proteínas Correpressoras , Proteínas de Ligação a DNA/genética , Células Endócrinas/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Histona Desmetilases , Histonas/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Estrutura Terciária de Proteína , Ratos , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/metabolismo , Deleção de Sequência , Fatores de Transcrição/genética , Transcrição Gênica
19.
Cell Tissue Res ; 353(3): 473-81, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23673414

RESUMO

In the anterior pituitary gland, folliculo-stellate cells and five types of hormone-producing cells are surrounded by an extracellular matrix (ECM) essential for these cells to perform their respective roles. Syndecans-type I transmembrane cell-surface heparan sulfate proteoglycans act as major ECM coreceptors via their respective heparan sulfate chains and efficiently transduce intracellular signals through the convergent action of their transmembrane and cytoplasmic domains. The syndecans comprise four family members in vertebrates: syndecan-1, -2, -3 and -4. However, whether syndecans are produced in the pituitary gland or whether they have a role as a coreceptor is not known. We therefore used (1) reverse transcription plus the polymerase chain reaction to analyze the expression of syndecan genes and (2) immunohistochemical techniques to identify the cells that produce the syndecans in the anterior pituitary gland of adult rat. Syndecan-2 mRNA expression was clearly detected in the corticotropes of the anterior pituitary gland. Moreover, the expression of syndecan-2 in the developing pituitary gland had a distinct temporospatial pattern. To identify the cells expressing syndecan-2 in the developing pituitary gland, we used double-immunohistochemistry for syndecan-2 and the cell markers E-cadherin (immature cells) and Ki-67 (proliferating cells). Some E-cadherin- and Ki-67-immunopositive cells expressed syndecan-2. Therefore, syndecan-2 expression occurs in developmentally regulated patterns and syndecan-2 probably has different roles in adult and developing anterior pituitary glands.


Assuntos
Regulação da Expressão Gênica/fisiologia , Adeno-Hipófise/embriologia , Sindecana-2/biossíntese , Animais , Caderinas/biossíntese , Caderinas/genética , Imuno-Histoquímica , Antígeno Ki-67/biossíntese , Antígeno Ki-67/genética , Adeno-Hipófise/citologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sindecana-2/genética
20.
Mol Endocrinol ; 27(3): 422-36, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23371388

RESUMO

Multiple homeodomain transcription factors are crucial for pituitary organogenesis and cellular differentiation. A homeodomain repressor, Msx1, is expressed from the ventral aspect of the developing anterior pituitary and implicated in gonadotrope differentiation. Here, we find that Msx1 represses transcription of lineage-specific pituitary genes such as the common α-glycoprotein subunit (αGSU) and GnRH receptor (GnRHR) promoters in the mouse gonadotrope-derived cell lines, αT3-1 and LßT2. Repression of the mouse GnRHR promoter by Msx1 is mediated through a consensus-binding motif in the downstream activin regulatory element (DARE). Truncation and mutation analyses of the human αGSU promoter map Msx1 repression to a site at -114, located at the junctional regulatory element (JRE). Dlx activators are closely related to the Msx repressors, acting through the same elements, and Dlx3 and Dlx2 act as transcriptional activators for GnRHR and αGSU, respectively. Small interfering RNA knockdown of Msx1 in αT3-1 cells increases endogenous αGSU and GnRHR mRNA expression. Msx1 gene expression reaches its maximal expression at the rostral edge at e13.5. The subsequent decline in Msx1 expression specifically coincides with the onset of expression of both αGSU and GnRHR. The expression levels of both αGSU and GnRHR in Msx1-null mice at e18.5 are higher compared with wild type, further confirming a role for Msx1 in the repression of αGSU and GnRHR. In summary, Msx1 functions as a negative regulator early in pituitary development by repressing the gonadotrope-specific αGSU and GnRHR genes, but a temporal decline in Msx1 expression alleviates this repression allowing induction of GnRHR and αGSU, thus serving to time the onset of gonadotrope-specific gene program.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa de Hormônios Glicoproteicos/genética , Gonadotrofos/metabolismo , Fator de Transcrição MSX1/metabolismo , Receptores LHRH/genética , Proteínas Repressoras/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Diferenciação Celular/genética , Linhagem Celular , Sequência Consenso/genética , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Gonadotrofos/citologia , Proteínas de Homeodomínio/metabolismo , Humanos , Fator de Transcrição MSX1/genética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Receptores LHRH/metabolismo , Proteínas Repressoras/genética , Elementos de Resposta/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA