Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 5(1): 492, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606540

RESUMEN

Hibernation involves prolonged intervals of profound metabolic suppression periodically interrupted by brief arousals to euthermy, the function of which is unknown. Annual cycles in mammals are timed by a photoperiodically-regulated thyroid-hormone-dependent mechanism in hypothalamic tanycytes, driven by thyrotropin (TSH) in the pars tuberalis (PT), which regulates local TH-converting deiodinases and triggers remodeling of neuroendocrine pathways. We demonstrate that over the course of hibernation in continuous darkness, arctic ground squirrels (Urocitellus parryii) up-regulate the retrograde TSH/Deiodinase/TH pathway, remodel hypothalamic tanycytes, and activate the reproductive axis. Forcing the premature termination of hibernation by warming animals induced hypothalamic deiodinase expression and the accumulation of secretory granules in PT thyrotrophs and pituitary gonadotrophs, but did not further activate the reproductive axis. We suggest that periodic arousals may allow for the transient activation of hypothalamic thyroid hormone signaling, cellular remodeling, and re-programming of brain circuits in preparation for the short Arctic summer.


Asunto(s)
Hibernación , Animales , Hibernación/fisiología , Yoduro Peroxidasa , Sciuridae/fisiología , Hormonas Tiroideas , Tirotropina
2.
Andrology ; 9(5): 1652-1661, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33998165

RESUMEN

BACKGROUND: Circulating prolactin concentration in rodents and humans is sexually dimorphic. Oestrogens are a well-characterised stimulator of prolactin release. Circulating prolactin fluctuates throughout the menstrual/oestrous cycle of females in response to oestrogen levels, but remains continually low in males. We have previously identified androgens as an inhibitor of prolactin release through characterisation of males of a mouse line with a conditional pituitary androgen receptor knockout (PARKO) which have an increase in circulating prolactin, but unchanged lactotroph number. OBJECTIVES: In the present study, we aimed to specify the cell type that androgens act on to repress prolactin release. MATERIALS AND METHODS: PARKO, lactotroph-specific, Pit1 lineage-specific and neural-specific conditional androgen receptor knockout male mice were investigated using prolactin ELISA, pituitary electron microscopy, immunohistochemistry and qRT-PCR. RESULTS: Lactotroph-specific, Pit1 lineage-specific and neural-specific conditional AR knockouts did not duplicate the high circulating prolactin seen in the PARKO line. Using electron microscopy to examine ultrastructure, we showed that pituitary androgen receptor knockout male mice develop lactotrophs that resemble those seen in female mice. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males. When expression of selected oestrogen-regulated anterior pituitary genes was examined, there were no differences in expression level between controls and knockouts. DISCUSSION: The cell type that androgens act on to repress prolactin release is not the lactotroph, cells in the Pit1-lineage, or the dopaminergic neurons in the hypothalamus. PARKO males develop a female-specific lactotroph ultrastructure that this is likely to contribute to the increase in circulating prolactin. Castrated PARKO males have significantly reduced circulating prolactin compared to intact males, which suggests that removal of both circulating oestrogens and androgens reduces the stimulation of pituitary prolactin release. CONCLUSION: Further investigation is needed into prolactin regulation by changes in androgen-oestrogen balance, which is involved sexual dimorphism of development and diseases including hyperprolactinemia.


Asunto(s)
Hiperprolactinemia/genética , Lactotrofos , Receptores Androgénicos/deficiencia , Animales , Estrógenos/metabolismo , Masculino , Ratones , Ratones Noqueados , Hipófisis/metabolismo , Prolactina/metabolismo
3.
J Neuroendocrinol ; 32(10): e12903, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32959418

RESUMEN

The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRß) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRß+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRß+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.


Asunto(s)
Comunicación Celular/fisiología , Hipófisis/citología , Animales , Biomarcadores/metabolismo , Células Endocrinas/citología , Células Endocrinas/fisiología , Células Endoteliales/citología , Células Endoteliales/fisiología , Ratones , Ratones Endogámicos C57BL , Pericitos/citología , Pericitos/fisiología , Hipófisis/metabolismo , Adenohipófisis/citología , Adenohipófisis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción SOXB1/metabolismo
4.
Nat Commun ; 10(1): 3960, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481663

RESUMEN

Translation is a basic cellular process and its capacity is adapted to cell function. In particular, secretory cells achieve high protein synthesis levels without triggering the protein stress response. It is unknown how and when translation capacity is increased during differentiation. Here, we show that the transcription factor Creb3l2 is a scaling factor for translation capacity in pituitary secretory cells and that it directly binds ~75% of regulatory and effector genes for translation. In parallel with this cell-autonomous mechanism, implementation of the physiological UPR pathway prevents triggering the protein stress response. Knockout mice for Tpit, a pituitary differentiation factor, show that Creb3l2 expression and its downstream regulatory network are dependent on Tpit. Further, Creb3l2 acts by direct targeting of translation effector genes in parallel with signaling pathways that otherwise regulate protein synthesis. Expression of Creb3l2 may be a useful means to enhance production of therapeutic proteins.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Hipófisis/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Diferenciación Celular/fisiología , Línea Celular , Retículo Endoplásmico/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Hipófisis/citología , Proopiomelanocortina/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteína 1 de Unión a la X-Box/metabolismo , Xenopus laevis
5.
JCI Insight ; 52019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31265437

RESUMEN

Hormones produced by the anterior pituitary gland regulate an array of important physiological functions, but pituitary hormone disorders are not fully understood. Herein we report that genetically-engineered mice with deletion of the hedgehog signaling receptor Patched1 by S100a4 promoter-driven Cre recombinase (S100a4-Cre;Ptch1fl/fl mutants) exhibit adult-onset hypogonadotropic hypogonadism and multiple pituitary hormone disorders. During the transition from puberty to adult, S100a4-Cre;Ptch1fl/fl mice of both sexes develop hypogonadism coupled with reduced gonadotropin levels. Their pituitary glands also display severe structural and functional abnormalities, as revealed by transmission electron microscopy and expression of key genes regulating pituitary endocrine functions. S100a4-Cre activity in the anterior pituitary gland is restricted to CD45+ cells of hematopoietic origin, including folliculo-stellate cells and other immune cell types, causing sex-specific changes in the expression of genes regulating the local microenvironment of the anterior pituitary. These findings provide in vivo evidence for the importance of pituitary hematopoietic cells in regulating fertility and endocrine function, in particular during sexual maturation and likely through sexually dimorphic mechanisms. These findings support a previously unrecognized role of hematopoietic cells in causing hypogonadotropic hypogonadism and provide inroads into the molecular and cellular basis for pituitary hormone disorders in humans.


Asunto(s)
Hipogonadismo/metabolismo , Integrasas/metabolismo , Receptor Patched-1/metabolismo , Hipófisis/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Animales , Epidídimo/patología , Femenino , Humanos , Hipogonadismo/genética , Hipogonadismo/patología , Masculino , Ratones , Ratones Noqueados , Ovario/patología , Receptor Patched-1/genética , Adenohipófisis/metabolismo , Reproducción/fisiología , Vesículas Seminales/patología , Maduración Sexual , Transducción de Señal , Testículo , Testosterona/sangre , Útero/patología
6.
PLoS Comput Biol ; 15(6): e1007030, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31194728

RESUMEN

Prolactin is a major hormone product of the pituitary gland, the central endocrine regulator. Despite its physiological importance, the cell-level mechanisms of prolactin production are not well understood. Having significantly improved the resolution of real-time-single-cell-GFP-imaging, the authors recently revealed that prolactin gene transcription is highly dynamic and stochastic yet shows space-time coordination in an intact tissue slice. However, it still remains an open question as to what kind of cellular communication mediates the observed space-time organization. To determine the type of interaction between cells we developed a statistical model. The degree of similarity between two expression time series was studied in terms of two distance measures, Euclidean and geodesic, the latter being a network-theoretic distance defined to be the minimal number of edges between nodes, and this was used to discriminate between juxtacrine from paracrine signalling. The analysis presented here suggests that juxtacrine signalling dominates. To further determine whether the coupling is coordinating transcription or post-transcriptional activities we used stochastic switch modelling to infer the transcriptional profiles of cells and estimated their similarity measures to deduce that their spatial cellular coordination involves coupling of transcription via juxtacrine signalling. We developed a computational model that involves an inter-cell juxtacrine coupling, yielding simulation results that show space-time coordination in the transcription level that is in agreement with the above analysis. The developed model is expected to serve as the prototype for the further study of tissue-level organised gene expression for epigenetically regulated genes, such as prolactin.


Asunto(s)
Comunicación Celular/genética , Modelos Biológicos , Comunicación Paracrina/genética , Animales , Comunicación Celular/fisiología , Biología Computacional , Regulación de la Expresión Génica/genética , Humanos , Masculino , Comunicación Paracrina/fisiología , Hipófisis/metabolismo , Prolactina/genética , Prolactina/metabolismo , Ratas , Ratas Transgénicas , Procesos Estocásticos
7.
Cereb Cortex ; 29(5): 2148-2159, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29850799

RESUMEN

Synaptosomal associated protein 25 kDa (SNAP25) is an essential component of the SNARE complex regulating synaptic vesicle fusion. SNAP25 deficiency has been implicated in a variety of cognitive disorders. We ablated SNAP25 from selected neuronal populations by generating a transgenic mouse (B6-Snap25tm3mcw (Snap25-flox)) with LoxP sites flanking exon5a/5b. In the presence of Cre-recombinase, Snap25-flox is recombined to a truncated transcript. Evoked synaptic vesicle release is severely reduced in Snap25 conditional knockout (cKO) neurons as shown by live cell imaging of synaptic vesicle fusion and whole cell patch clamp recordings in cultured hippocampal neurons. We studied Snap25 cKO in subsets of cortical projection neurons in vivo (L5-Rbp4-Cre; L6-Ntsr1-Cre; L6b-Drd1a-Cre). cKO neurons develop normal axonal projections, but axons are not maintained appropriately, showing signs of swelling, fragmentation and eventually complete absence. Onset and progression of degeneration are dependent on the neuron type, with L5 cells showing the earliest and most severe axonal loss. Ultrastructural examination revealed that cKO neurites contain autophagosome/lysosome-like structures. Markers of inflammation such as Iba1 and lipofuscin are increased only in adult cKO cortex. Snap25 cKO can provide a model to study genetic interactions with environmental influences in several disorders.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Neuronas/patología , Neuronas/fisiología , Proteína 25 Asociada a Sinaptosomas/fisiología , Animales , Axones/patología , Axones/fisiología , Axones/ultraestructura , Encéfalo/ultraestructura , Femenino , Masculino , Ratones Noqueados , Neuronas/ultraestructura , Transmisión Sináptica , Vesículas Sinápticas
8.
J Biol Chem ; 293(4): 1151-1162, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29123029

RESUMEN

Normal renin synthesis and secretion is important for the maintenance of juxtaglomerular apparatus architecture. Mice lacking a functional Ren1d gene are devoid of renal juxtaglomerular cell granules and exhibit an altered macula densa morphology. Due to the species-specificity of renin activity, transgenic mice are ideal models for experimentally investigating and manipulating expression patterns of the human renin gene in a native cellular environment without confounding renin-angiotensin system interactions. A 55-kb transgene encompassing the human renin locus was crossed onto the mouse Ren1d-null background, restoring granulation in juxtaglomerular cells. Correct processing of human renin in dense core granules was confirmed by immunogold labeling. After stimulation of the renin-angiotensin system, juxtaglomerular cells contained rhomboid protogranules with paracrystalline contents, dilated rough endoplasmic reticulum, and electron-lucent granular structures. However, complementation of Ren1d-/- mice with human renin was unable to rescue the abnormality seen in macula densa structure. The juxtaglomerular apparatus was still able to respond to tubuloglomerular feedback in isolated perfused juxtaglomerular apparatus preparations, although minor differences in glomerular tuft contractility and macula densa cell calcium handling were observed. This study reveals that the human renin protein is able to complement the mouse Ren1d-/- non-granulated defect and suggests that granulopoiesis requires a structural motif that is conserved between the mouse Ren1d and human renin proteins. It also suggests that the altered macula densa phenotype is related to the activity of the renin-1d enzyme in a local juxtaglomerular renin-angiotensin system.


Asunto(s)
Prueba de Complementación Genética , Aparato Yuxtaglomerular/enzimología , Renina/biosíntesis , Transgenes , Animales , Humanos , Aparato Yuxtaglomerular/patología , Ratones , Ratones Noqueados , Renina/genética
9.
Sci Rep ; 7: 45541, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28361877

RESUMEN

Despite being unable to activate the cognate ghrelin receptor (GHS-R), unacylated ghrelin (UAG) possesses a unique activity spectrum that includes promoting bone marrow adipogenesis. Since a receptor mediating this action has not been identified, we re-appraised the potential interaction of UAG with GHS-R in the regulation of bone marrow adiposity. Surprisingly, the adipogenic effects of intra-bone marrow (ibm)-infused acylated ghrelin (AG) and UAG were abolished in male GHS-R-null mice. Gas chromatography showed that isolated tibial marrow adipocytes contain the medium-chain fatty acids utilised in the acylation of UAG, including octanoic acid. Additionally, immunohistochemistry and immunogold electron microscopy revealed that tibial marrow adipocytes show prominent expression of the UAG-activating enzyme ghrelin O-acyl transferase (GOAT), which is located in the membranes of lipid trafficking vesicles and in the plasma membrane. Finally, the adipogenic effect of ibm-infused UAG was completely abolished in GOAT-KO mice. Thus, the adipogenic action of exogenous UAG in tibial marrow is dependent upon acylation by GOAT and activation of GHS-R. This suggests that UAG is subject to target cell-mediated activation - a novel mechanism for manipulating hormone activity.


Asunto(s)
Aciltransferasas/metabolismo , Adipogénesis , Médula Ósea/metabolismo , Ghrelina/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Ghrelina/metabolismo , Acilación , Animales , Cromatografía de Gases , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Inmunoelectrónica , Receptores de Ghrelina/deficiencia
10.
Am J Physiol Renal Physiol ; 312(4): F778-F790, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28179256

RESUMEN

Zebrafish provide an excellent model in which to assess the role of the renin-angiotensin system in renal development, injury, and repair. In contrast to mammals, zebrafish kidney organogenesis terminates with the mesonephros. Despite this, the basic functional structure of the nephron is conserved across vertebrates. The relevance of teleosts for studies relating to the regulation of the renin-angiotensin system was established by assessing the phenotype and functional regulation of renin-expressing cells in zebrafish. Transgenic fluorescent reporters for renin (ren), smooth muscle actin (acta2), and platelet-derived growth factor receptor-beta (pdgfrb) were studied to determine the phenotype and secretory ultrastructure of perivascular renin-expressing cells. Whole kidney ren transcription responded to altered salinity, pharmacological renin-angiotensin system inhibition, and renal injury. Mesonephric ren-expressing cells occupied niches at the preglomerular arteries and afferent arterioles, forming intermittent epithelioid-like multicellular clusters exhibiting a granular secretory ultrastructure. In contrast, renin cells of the efferent arterioles were thin bodied and lacked secretory granules. Renin cells expressed the perivascular cell markers acta2 and pdgfrb Transcriptional responses of ren to physiological challenge support the presence of a functional renin-angiotensin system and are consistent with the production of active renin. The reparative capability of the zebrafish kidney was harnessed to demonstrate that ren transcription is a marker for renal injury and repair. Our studies demonstrate substantive conservation of renin regulation across vertebrates, and ultrastructural studies of renin cells reveal at least two distinct morphologies of mesonephric perivascular ren-expressing cells.


Asunto(s)
Forma de la Célula , Sistema Renina-Angiotensina , Renina/metabolismo , Conductos Mesonéfricos/enzimología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Animales Modificados Genéticamente , Regulación del Desarrollo de la Expresión Génica , Genotipo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Microscopía Fluorescente , Miocitos del Músculo Liso/metabolismo , Pericitos/metabolismo , Fenotipo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Renina/genética , Transcripción Genética , Conductos Mesonéfricos/ultraestructura , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
11.
Sci Rep ; 7: 40620, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28094777

RESUMEN

Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) stimulates calcium release from acidic stores such as lysosomes and is a highly potent calcium-mobilising second messenger. NAADP plays an important role in calcium signalling in the heart under basal conditions and following ß-adrenergic stress. Nevertheless, the spatial interaction of acidic stores with other parts of the calcium signalling apparatus in cardiac myocytes is unknown. We present evidence that lysosomes are intimately associated with the sarcoplasmic reticulum (SR) in ventricular myocytes; a median separation of 20 nm in 2D electron microscopy and 3.3 nm in 3D electron tomography indicates a genuine signalling microdomain between these organelles. Fourier analysis of immunolabelled lysosomes suggests a sarcomeric pattern (dominant wavelength 1.80 µm). Furthermore, we show that lysosomes form close associations with mitochondria (median separation 6.2 nm in 3D studies) which may provide a basis for the recently-discovered role of NAADP in reperfusion-induced cell death. The trigger hypothesis for NAADP action proposes that calcium release from acidic stores subsequently acts to enhance calcium release from the SR. This work provides structural evidence in cardiac myocytes to indicate the formation of microdomains between acidic and SR calcium stores, supporting emerging interpretations of NAADP physiology and pharmacology in heart.


Asunto(s)
Lisosomas/metabolismo , Lisosomas/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/ultraestructura , Animales , Biomarcadores , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , NADP/análogos & derivados , NADP/metabolismo , Orgánulos/metabolismo , Conejos
12.
Elife ; 52016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27897970

RESUMEN

Hepcidin is the master regulator of systemic iron homeostasis. Derived primarily from the liver, it inhibits the iron exporter ferroportin in the gut and spleen, the sites of iron absorption and recycling respectively. Recently, we demonstrated that ferroportin is also found in cardiomyocytes, and that its cardiac-specific deletion leads to fatal cardiac iron overload. Hepcidin is also expressed in cardiomyocytes, where its function remains unknown. To define the function of cardiomyocyte hepcidin, we generated mice with cardiomyocyte-specific deletion of hepcidin, or knock-in of hepcidin-resistant ferroportin. We find that while both models maintain normal systemic iron homeostasis, they nonetheless develop fatal contractile and metabolic dysfunction as a consequence of cardiomyocyte iron deficiency. These findings are the first demonstration of a cell-autonomous role for hepcidin in iron homeostasis. They raise the possibility that such function may also be important in other tissues that express both hepcidin and ferroportin, such as the kidney and the brain.


Asunto(s)
Hepcidinas/metabolismo , Homeostasis , Hierro/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Hepcidinas/genética , Ratones
13.
Mol Cell ; 64(4): 746-759, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863227

RESUMEN

Excitation-transcription coupling, linking stimulation at the cell surface to changes in nuclear gene expression, is conserved throughout eukaryotes. How closely related coexpressed transcription factors are differentially activated remains unclear. Here, we show that two Ca2+-dependent transcription factor isoforms, NFAT1 and NFAT4, require distinct sub-cellular InsP3 and Ca2+ signals for physiologically sustained activation. NFAT1 is stimulated by sub-plasmalemmal Ca2+ microdomains, whereas NFAT4 additionally requires Ca2+ mobilization from the inner nuclear envelope by nuclear InsP3 receptors. NFAT1 is rephosphorylated (deactivated) more slowly than NFAT4 in both cytoplasm and nucleus, enabling a more prolonged activation phase. Oscillations in cytoplasmic Ca2+, long considered the physiological form of Ca2+ signaling, play no role in activating either NFAT protein. Instead, effective sustained physiological activation of NFAT4 is tightly linked to oscillations in nuclear Ca2+. Our results show how gene expression can be controlled by coincident yet geographically distinct Ca2+ signals, generated by a freely diffusible InsP3 message.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Fosfatos de Inositol/metabolismo , Factores de Transcripción NFATC/genética , Proteínas Recombinantes de Fusión/genética , Animales , Basófilos/citología , Basófilos/efectos de los fármacos , Basófilos/metabolismo , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Línea Celular , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Leucotrieno C4/farmacología , Factores de Transcripción NFATC/metabolismo , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Tapsigargina/farmacología , Transcripción Genética
14.
Kidney Int ; 90(6): 1251-1261, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27678158

RESUMEN

Pericytes, perivascular cells embedded in the microvascular wall, are crucial for vascular homeostasis. These cells also play diverse roles in tissue development and regeneration as multi-lineage progenitors, immunomodulatory cells and as sources of trophic factors. Here, we establish that pericytes are renin producing cells in the human kidney. Renin was localized by immunohistochemistry in CD146 and NG2 expressing pericytes, surrounding juxtaglomerular and afferent arterioles. Similar to pericytes from other organs, CD146+CD34-CD45-CD56- renal fetal pericytes, sorted by flow cytometry, exhibited tri-lineage mesodermal differentiation potential in vitro. Additionally, renin expression was triggered in cultured kidney pericytes by cyclic AMP as confirmed by immuno-electron microscopy, and secretion of enzymatically functional renin, capable of generating angiotensin I. Pericytes derived from second trimester human placenta also expressed renin in an inducible fashion although the renin activity was much lower than in renal pericytes. Thus, our results confirm and extend the recently discovered developmental plasticity of microvascular pericytes, and may open new perspectives to the therapeutic regulation of the renin-angiotensin system.


Asunto(s)
Riñón/ultraestructura , Pericitos/metabolismo , Renina/metabolismo , Humanos , Riñón/embriología , Células Madre Mesenquimatosas , Cultivo Primario de Células
15.
Endocrinology ; 157(9): 3631-46, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27580811

RESUMEN

Thyrotrope hyperplasia and hypertrophy are common responses to primary hypothyroidism. To understand the genetic regulation of these processes, we studied gene expression changes in the pituitaries of Cga(-/-) mice, which are deficient in the common α-subunit of TSH, LH, and FSH. These mice have thyrotrope hypertrophy and hyperplasia and develop thyrotrope adenoma. We report that cell proliferation is increased, but the expression of most stem cell markers is unchanged. The α-subunit is required for secretion of the glycoprotein hormone ß-subunits, and mutants exhibit elevated expression of many genes involved in the unfolded protein response, consistent with dilation and stress of the endoplasmic reticulum. Mutants have elevated expression of transcription factors that are important in thyrotrope function, such as Gata2 and Islet 1, and those that stimulate proliferation, including Nupr1, E2f1, and Etv5. We characterized the expression and function of a novel, overexpressed gene, transcription elongation factor A (SII)-like 5 (Tceal5). Stable expression of Tceal5 in a pituitary progenitor cell line is sufficient to increase cell proliferation. Thus, Tceal5 may act as a proto-oncogene. This study provides a rich resource for comparing pituitary transcriptomes and an analysis of gene expression networks.


Asunto(s)
Adenoma/metabolismo , Hipotiroidismo/metabolismo , Neoplasias Hipofisarias/metabolismo , Tirotrofos/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Retículo Endoplásmico/ultraestructura , Estrés del Retículo Endoplásmico , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Masculino , Ratones , Datos de Secuencia Molecular , Proto-Oncogenes Mas , Tirotropina de Subunidad beta/metabolismo , Factores de Elongación Transcripcional/metabolismo , Respuesta de Proteína Desplegada
16.
Stem Cells ; 34(8): 2063-78, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27097283

RESUMEN

An expanded hexanucleotide repeat in a noncoding region of the C9orf72 gene is a major cause of amyotrophic lateral sclerosis (ALS), accounting for up to 40% of familial cases and 7% of sporadic ALS in European populations. We have generated induced pluripotent stem cells (iPSCs) from fibroblasts of patients carrying C9orf72 hexanucleotide expansions, differentiated these to functional motor and cortical neurons, and performed an extensive phenotypic characterization. In C9orf72 iPSC-derived motor neurons, decreased cell survival is correlated with dysfunction in Ca(2+) homeostasis, reduced levels of the antiapoptotic protein Bcl-2, increased endoplasmic reticulum (ER) stress, and reduced mitochondrial membrane potential. Furthermore, C9orf72 motor neurons, and also cortical neurons, show evidence of abnormal protein aggregation and stress granule formation. This study is an extensive characterization of iPSC-derived motor neurons as cellular models of ALS carrying C9orf72 hexanucleotide repeats, which describes a novel pathogenic link between C9orf72 mutations, dysregulation of calcium signaling, and altered proteostasis and provides a potential pharmacological target for the treatment of ALS and the related neurodegenerative disease frontotemporal dementia. Stem Cells 2016;34:2063-2078.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/genética , Calcio/metabolismo , Expansión de las Repeticiones de ADN/genética , Retículo Endoplásmico/metabolismo , Demencia Frontotemporal/patología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/metabolismo , Esclerosis Amiotrófica Lateral/genética , Apoptosis , Caspasa 3/metabolismo , Diferenciación Celular , Reprogramación Celular , Corteza Cerebral/patología , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Retículo Endoplásmico/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/patología , Demencia Frontotemporal/genética , Homeostasis/genética , Humanos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Péptidos/metabolismo , Agregado de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN/genética
17.
Stem Cell Reports ; 6(3): 342-56, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26905200

RESUMEN

Heterozygous mutations in the glucocerebrosidase gene (GBA) represent the strongest common genetic risk factor for Parkinson's disease (PD), the second most common neurodegenerative disorder. However, the molecular mechanisms underlying this association are still poorly understood. Here, we have analyzed ten independent induced pluripotent stem cell (iPSC) lines from three controls and three unrelated PD patients heterozygous for the GBA-N370S mutation, and identified relevant disease mechanisms. After differentiation into dopaminergic neurons, we observed misprocessing of mutant glucocerebrosidase protein in the ER, associated with activation of ER stress and abnormal cellular lipid profiles. Furthermore, we observed autophagic perturbations and an enlargement of the lysosomal compartment specifically in dopamine neurons. Finally, we found increased extracellular α-synuclein in patient-derived neuronal culture medium, which was not associated with exosomes. Overall, ER stress, autophagic/lysosomal perturbations, and elevated extracellular α-synuclein likely represent critical early cellular phenotypes of PD, which might offer multiple therapeutic targets.


Asunto(s)
Autofagia , Neuronas Dopaminérgicas/metabolismo , Estrés del Retículo Endoplásmico , Glucosilceramidasa/genética , Células Madre Pluripotentes Inducidas/citología , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Animales , Línea Celular , Células Cultivadas , Neuronas Dopaminérgicas/citología , Exosomas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lisosomas/metabolismo , Ratones , Mutación Missense , Neurogénesis , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
18.
Elife ; 5: e08494, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26828110

RESUMEN

Transcription at individual genes in single cells is often pulsatile and stochastic. A key question emerges regarding how this behaviour contributes to tissue phenotype, but it has been a challenge to quantitatively analyse this in living cells over time, as opposed to studying snap-shots of gene expression state. We have used imaging of reporter gene expression to track transcription in living pituitary tissue. We integrated live-cell imaging data with statistical modelling for quantitative real-time estimation of the timing of switching between transcriptional states across a whole tissue. Multiple levels of transcription rate were identified, indicating that gene expression is not a simple binary 'on-off' process. Immature tissue displayed shorter durations of high-expressing states than the adult. In adult pituitary tissue, direct cell contacts involving gap junctions allowed local spatial coordination of prolactin gene expression. Our findings identify how heterogeneous transcriptional dynamics of single cells may contribute to overall tissue behaviour.


Asunto(s)
Regulación de la Expresión Génica , Hipófisis/fisiología , Transcripción Genética , Animales , Perfilación de la Expresión Génica , Genes Reporteros , Imagen Óptica , Ratas Endogámicas F344 , Análisis Espacio-Temporal
19.
Curr Biol ; 25(20): 2651-62, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26412130

RESUMEN

Persistent free-running circannual (approximately year-long) rhythms have evolved in animals to regulate hormone cycles, drive metabolic rhythms (including hibernation), and time annual reproduction. Recent studies have defined the photoperiodic input to this rhythm, wherein melatonin acts on thyrotroph cells of the pituitary pars tuberalis (PT), leading to seasonal changes in the control of thyroid hormone metabolism in the hypothalamus. However, seasonal rhythms persist in constant conditions in many species in the absence of a changing photoperiod signal, leading to the generation of circannual cycles. It is not known which cells, tissues, and pathways generate these remarkable long-term rhythmic processes. We show that individual PT thyrotrophs can be in one of two binary states reflecting either a long (EYA3(+)) or short (CHGA(+)) photoperiod, with the relative proportion in each state defining the phase of the circannual cycle. We also show that a morphogenic cycle driven by the PT leads to extensive re-modeling of the PT and hypothalamus over the circannual cycle. We propose that the PT may employ a recapitulated developmental pathway to drive changes in morphology of tissues and cells. Our data are consistent with the hypothesis that the circannual timer may reside within the PT thyrotroph and is encoded by a binary switch timing mechanism, which may regulate the generation of circannual neuroendocrine rhythms, leading to dynamic re-modeling of the hypothalamic interface. In summary, the PT-ventral hypothalamus now appears to be a prime structure involved in long-term rhythm generation.


Asunto(s)
Relojes Circadianos , Fotoperiodo , Ovinos/fisiología , Tirotrofos/fisiología , Animales , Masculino
20.
Proc Natl Acad Sci U S A ; 112(10): 3164-9, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713362

RESUMEN

Iron is essential to the cell. Both iron deficiency and overload impinge negatively on cardiac health. Thus, effective iron homeostasis is important for cardiac function. Ferroportin (FPN), the only known mammalian iron-exporting protein, plays an essential role in iron homeostasis at the systemic level. It increases systemic iron availability by releasing iron from the cells of the duodenum, spleen, and liver, the sites of iron absorption, recycling, and storage respectively. However, FPN is also found in tissues with no known role in systemic iron handling, such as the heart, where its function remains unknown. To explore this function, we generated mice with a cardiomyocyte-specific deletion of Fpn. We show that these animals have severely impaired cardiac function, with a median survival of 22 wk, despite otherwise unaltered systemic iron status. We then compared their phenotype with that of ubiquitous hepcidin knockouts, a recognized model of the iron-loading disease hemochromatosis. The phenotype of the hepcidin knockouts was far milder, with normal survival up to 12 mo, despite far greater iron loading in the hearts. Histological examination demonstrated that, although cardiac iron accumulates within the cardiomyocytes of Fpn knockouts, it accumulates predominantly in other cell types in the hepcidin knockouts. We conclude, first, that cardiomyocyte FPN is essential for intracellular iron homeostasis and, second, that the site of deposition of iron within the heart determines the severity with which it affects cardiac function. Both findings have significant implications for the assessment and treatment of cardiac complications of iron dysregulation.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Corazón/fisiología , Homeostasis , Hierro/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...