Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Rev Mol Cell Biol ; 14(6): 393-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23636498

RESUMEN

The past few years have marked significant anniversaries in signal transduction, including the identification of classic growth factors and morphogens, the notion of protein modification through phosphorylation and the characterization of protein interaction domains. Here, six researchers reflect on the context in which these discoveries were made, and how our concept of cell signalling has evolved during the past three decades.


Asunto(s)
Investigación Biomédica/historia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Animales , Investigación Biomédica/métodos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Fosforilación
2.
Horm Behav ; 162: 105508, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38513527

RESUMEN

Social environments modulate endocrine function, yet it is unclear whether individuals can become like their social partners in how they physiologically respond to stressors. This social transmission of hypothalamic-pituitary-adrenal (HPA) axis reactivity could have long-term consequences for health and lifespan of individuals if their social partners react to stressors with an exaggerated HPA axis response. We tested whether glucocorticoid levels in response to stress of breeding partners changes after breeding depending on whether partners had similar or dissimilar postnatal conditions. We manipulated postnatal conditions by mimicking early life stress in zebra finch chicks (Taeniopygia guttata) via postnatal corticosterone exposure. When they reached adulthood, we created breeding pairs where the female and male had experienced either the same or different early life hormonal treatment (corticosterone or control). Before and after breeding, we obtained blood samples within 3 min and after 10 min or 30 min of restraint stress (baseline, cort10, cort30). We found that corticosterone levels of individuals in response to restraint were affected by their own and their partner's early life conditions, but did not change after breeding. However, across all pairs, partners became more similar in cort30 levels after breeding, although differences between partners in cort10 remained greater in pairs with a corticosterone-treated female. Thus, we show that HPA axis response to stressors in adulthood can be modulated by reproductive partners and that similarity between partners is reduced when females are postnatally exposed to elevated glucocorticoids.


Asunto(s)
Corticosterona , Pinzones , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Estrés Psicológico , Animales , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Femenino , Sistema Hipófiso-Suprarrenal/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Masculino , Corticosterona/sangre , Estrés Psicológico/metabolismo , Estrés Psicológico/sangre , Pinzones/fisiología , Reproducción/fisiología , Restricción Física/fisiología
3.
Cell ; 135(3): 510-23, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18984162

RESUMEN

Tumor cells exhibit two different modes of individual cell movement. Mesenchymal-type movement is characterized by an elongated cellular morphology and requires extracellular proteolysis. In amoeboid movement, cells have a rounded morphology, are less dependent on proteases, and require high Rho-kinase signaling to drive elevated levels of actomyosin contractility. These two modes of cell movement are interconvertible. We show that mesenchymal-type movement in melanoma cells is driven by activation of the GTPase Rac through a complex containing NEDD9, a recently identified melanoma metastasis gene, and DOCK3, a Rac guanine nucleotide exchange factor. Rac signals through WAVE2 to direct mesenchymal movement and suppress amoeboid movement through decreasing actomyosin contractility. Conversely, in amoeboid movement, Rho-kinase signaling activates a Rac GAP, ARHGAP22, that suppresses mesenchymal movement by inactivating Rac. We demonstrate tight interplay between Rho and Rac in determining different modes of tumor cell movement, revealing how tumor cells switch between different modes of movement.


Asunto(s)
Movimiento Celular , Melanoma/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Actomiosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Quimerina 1/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
4.
Horm Behav ; 142: 105153, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35325691

RESUMEN

Most animals in the temperate zone exhibit robust seasonal rhythms in neuroendocrine, physiological and behavioral processes. The integration of predictive and supplementary environmental cues (e.g., nutrients) involves a series of discrete, and interconnected brain regions that span hypothalamic, thalamic, mesencephalic, and limbic regions. Species-specific adaptive changes in these neuroendocrine structures and cellular plasticity have likely evolved to support seasonal life-history transitions. Despite significant advances in our understanding of ecological responses to predictive and supplementary environmental cues, there remains a paucity of literature on how these diverse cues impact the underlying neural and cellular substrates. To date, most scientific approach has focused on neuroendocrine responses to annual changes in daylength, referred to as photoperiod, due to the robust physiological changes to light manipulations in laboratory settings. In this review, we highlight the relatively few animal models that have been effectively used to investigate how predictive day lengths, and supplementary cues are integrated across hypothalamic nuclei, and discuss key findings of how seasonal rhythms in physiology are governed by adaptive neuroendocrine changes. We discuss how specific brain regions integrate environmental cues to form a complex multiunit or 'modular' system that has evolved to optimize the timing of seasonal physiology. Overall, the review aims to highlight the existence of a modular network of neural regions that independently contribute to timing seasonal physiology. This paper proposes that a multi-modular neuroendocrine system has evolved in which independent neural 'units' operate to support species-specific seasonal rhythms.


Asunto(s)
Aves , Mamíferos , Animales , Aves/fisiología , Hipotálamo , Mamíferos/fisiología , Fotoperiodo , Reproducción/fisiología , Estaciones del Año
5.
Neuroendocrinology ; 110(7-8): 671-687, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31630145

RESUMEN

INTRODUCTION: The central regulation of fertility is carefully coordinated with energy homeostasis, and infertility is frequently the outcome of energy imbalance. Neurons in the hypothalamus expressing neuropeptide Y and agouti-related peptide (NPY/AgRP neurons) are strongly implicated in linking metabolic cues with fertility regulation. OBJECTIVE: We aimed here to determine the impact of selectively activating NPY/AgRP neurons, critical regulators of metabolism, on the activity of luteinizing hormone (LH) pulse generation. METHODS: We employed a suite of in vivo optogenetic and chemogenetic approaches with serial measurements of LH to determine the impact of selectively activating NPY/AgRP neurons on dynamic LH secretion. In addition, electrophysiological studies in ex vivo brain slices were employed to ascertain the functional impact of activating NPY/AgRP neurons on gonadotropin-releasing hormone (GnRH) neurons. RESULTS: Selective activation of NPY/AgRP neurons significantly decreased post-castration LH secretion. This was observed in males and females, as well as in prenatally androgenized females that recapitulate the persistently elevated LH pulse frequency characteristic of polycystic ovary syndrome (PCOS). Reduced LH pulse frequency was also observed when optogenetic stimulation was restricted to NPY/AgRP fiber projections surrounding GnRH neuron cell bodies in the rostral preoptic area. However, electrophysiological studies in ex vivo brain slices indicated these effects were likely to be indirect. CONCLUSIONS: These data demonstrate the ability of NPY/AgRP neuronal signaling to modulate and, specifically, reduce GnRH/LH pulse generation. The findings suggest a mechanism by which increased activity of this hunger circuit, in response to negative energy balance, mediates impaired fertility in otherwise reproductively fit states, and highlight a potential mechanism to slow LH pulsatility in female infertility disorders, such as PCOS, that are associated with hyperactive LH secretion.


Asunto(s)
Hambre/fisiología , Hormona Luteinizante/metabolismo , Red Nerviosa/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Red Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Neuropéptido Y/metabolismo , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Síndrome del Ovario Poliquístico/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/psicología , Vías Secretoras/fisiología
6.
J Cell Sci ; 129(24): 4466-4479, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807006

RESUMEN

Directional cell migration involves reorientation of the secretory machinery. However, the molecular mechanisms that control this reorientation are not well characterised. Here, we identify a new Rho effector protein, named FAM65A, which binds to active RHOA, RHOB and RHOC. FAM65A links RHO proteins to Golgi-localising cerebral cavernous malformation-3 protein (CCM3; also known as PDCD10) and its interacting proteins mammalian STE20-like protein kinases 3 and 4 (MST3 and MST4; also known as STK24 and STK26, respectively). Binding of active RHO proteins to FAM65A does not affect the kinase activity of MSTs but results in their relocation from the Golgi in a CCM3-dependent manner. This relocation is crucial for reorientation of the Golgi towards the leading edge and subsequent directional cell migration. Our results reveal a previously unidentified pathway downstream of RHO that regulates the polarity of migrating cells through Golgi reorientation in a FAM65A-, CCM3- and MST3- and MST4-dependent manner.


Asunto(s)
Movimiento Celular , Aparato de Golgi/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Activación Enzimática , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas de la Membrana/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(2): 596-601, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25550522

RESUMEN

Polycystic ovarian syndrome (PCOS), the leading cause of female infertility, is associated with an increase in luteinizing hormone (LH) pulse frequency, implicating abnormal steroid hormone feedback to gonadotropin-releasing hormone (GnRH) neurons. This study investigated whether modifications in the synaptically connected neuronal network of GnRH neurons could account for this pathology. The PCOS phenotype was induced in mice following prenatal androgen (PNA) exposure. Serial blood sampling confirmed that PNA elicits increased LH pulse frequency and impaired progesterone negative feedback in adult females, mimicking the neuroendocrine abnormalities of the clinical syndrome. Imaging of GnRH neurons revealed greater dendritic spine density that correlated with increased putative GABAergic but not glutamatergic inputs in PNA mice. Mapping of steroid hormone receptor expression revealed that PNA mice had 59% fewer progesterone receptor-expressing cells in the arcuate nucleus of the hypothalamus (ARN). To address whether increased GABA innervation to GnRH neurons originates in the ARN, a viral-mediated Cre-lox approach was taken to trace the projections of ARN GABA neurons in vivo. Remarkably, projections from ARN GABAergic neurons heavily contacted and even bundled with GnRH neuron dendrites, and the density of fibers apposing GnRH neurons was even greater in PNA mice (56%). Additionally, this ARN GABA population showed significantly less colocalization with progesterone receptor in PNA animals compared with controls. Together, these data describe a robust GABAergic circuit originating in the ARN that is enhanced in a model of PCOS and may underpin the neuroendocrine pathophysiology of the syndrome.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiopatología , Neuronas GABAérgicas/fisiología , Hormona Liberadora de Gonadotropina/fisiología , Síndrome del Ovario Poliquístico/fisiopatología , Andrógenos/administración & dosificación , Animales , Núcleo Arqueado del Hipotálamo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Hormona Luteinizante/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Sistemas Neurosecretores/fisiopatología , Síndrome del Ovario Poliquístico/etiología , Síndrome del Ovario Poliquístico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Receptores de Progesterona/fisiología
8.
Neuroendocrinology ; 105(2): 157-169, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27710963

RESUMEN

BACKGROUND/AIMS: Arcuate nucleus (ARN) γ-aminobutyric acid (GABA) neurons are implicated in many critical homeostatic mechanisms, from food intake to fertility. To determine the functional relevance of ARN GABA neurons, it is essential to define the neurotransmitters co-expressed with and potentially co-released from ARN GABA neurons. METHODS: The present study investigated the expression of markers of specific signaling molecules by ARN GABA neurons in brain sections from male, female, and, in some cases, prenatally androgen-treated (PNA) female, vesicular GABA transporter (VGaT)-ires-Cre/tdTomato reporter mice. Immunofluorescence for kisspeptin, ß-endorphin, neuropeptide Y (NPY), tyrosine hydroxylase (TH) and neuronal nitric oxide synthase (nNOS) was detected by confocal microscopy, and co-localization with tdTomato VGaT reporter expression throughout the ARN was quantified. RESULTS: GABA neurons rarely co-localized with kisspeptin (<2%) or ß-endorphin (<1%), and only a small proportion of kisspeptin (∼10%) or ß-endorphin (∼3%) neurons co-localized with VGaT in male and female mice. In contrast, one-third of ARN GABA neurons co-localized with NPY, and nearly all NPY neurons (>95%) co-localized with VGaT across groups. Both TH and nNOS labeling was co-localized with ∼10% of ARN GABA neurons. The proportion of TH neurons co-localized with VGaT was significantly greater in males than either control or PNA females, and the proportion of nNOS neurons co-localizing VGaT was higher in control and PNA females compared with males. CONCLUSION: These data highlight NPY as a significant subpopulation of ARN GABA neurons, demonstrate no significant impact of PNA on signal co-expression, and, for the first time, show sexually dimorphic co-expression patterns of TH and nNOS with ARN GABA neurons.


Asunto(s)
Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/fisiología , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/fisiología , Caracteres Sexuales , Andrógenos/administración & dosificación , Andrógenos/metabolismo , Animales , Recuento de Células , Femenino , Técnica del Anticuerpo Fluorescente , Kisspeptinas/metabolismo , Masculino , Ratones Transgénicos , Neuropéptido Y/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , betaendorfina/metabolismo
9.
J Cell Sci ; 127(Pt 3): 686-99, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24284074

RESUMEN

Cell polarization is essential for neuronal development in both the embryonic and postnatal brain. Here, using primary cultures, in vivo postnatal electroporation and conditional genetic ablation, we show that the Ras-like small GTPase RalA and its effector, the exocyst, regulate the morphology and polarized migration of neural progenitors derived from the subventricular zone, a major neurogenic niche in the postnatal brain. Active RalA promotes the direct binding between the exocyst subunit Exo84 and the PDZ domain of Par6 through a non-canonical PDZ-binding motif. Blocking the Exo84-Par6 interaction impairs polarization in postnatal neural progenitors and cultured embryonic neurons. Our results provide the first in vivo characterization of RalA function in the mammalian brain and highlight a novel molecular mechanism for cell polarization. Given that the exocyst and the Par complex are conserved in many tissues, the functional significance of their interaction and its regulation by RalA are likely to be important in a wide range of polarization events.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neurogénesis , Neuronas/metabolismo , Proteínas de Unión al GTP ral/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Polaridad Celular/genética , Dominios PDZ/genética , Cultivo Primario de Células , Unión Proteica , Transducción de Señal , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP ral/metabolismo
10.
J Cell Sci ; 125(Pt 7): 1814-26, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22328516

RESUMEN

Neural precursor expressed, developmentally down-regulated 9 (NEDD9), a member of the Cas family of signal transduction molecules, is amplified at the genetic level in melanoma, and elevated expression levels have been shown to correlate with melanoma progression and metastasis. NEDD9 interacts with the guanine nucleotide exchange factor DOCK3 to promote Rac activation and the elongated, mesenchymal-type of tumour cell invasion, but the molecular mechanisms through which NEDD9 promotes melanoma metastasis are not fully understood. We show that signalling through increased NEDD9 levels requires integrin ß3 signalling, which leads to elevated phosphorylation of integrin ß3. This results in increased Src and FAK but decreased ROCK signalling to drive elongated, mesenchymal-type invasion in environments that contain vitronectin. NEDD9 overexpression does not affect ROCK signalling through activation of RhoA but decreases ROCKII signalling through Src-dependent phosphorylation of a negative regulatory site Tyr722. In NEDD9-overexpressing melanoma cells, inhibition of Src with dasatinib results in a switch from Rac-driven elongated, mesenchymal-type invasion to ROCK-dependent rounded, amoeboid invasion. These findings brings into question whether dasatinib would work as a therapeutic agent to block melanoma invasion and metastasis. On the basis of the in vitro data presented here, a combination treatment of dasatinib and a ROCK inhibitor might be a better alternative in order to inhibit both elongated, mesenchymal-type and rounded, amoeboid motility.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/genética , Integrina beta3/metabolismo , Melanoma/genética , Melanoma/patología , Mesodermo/metabolismo , Fosfoproteínas/metabolismo , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Humanos , Metástasis de la Neoplasia/genética , Fosfoproteínas/genética , Fosforilación , Transducción de Señal/genética , Células Tumorales Cultivadas
11.
J Cell Sci ; 125(Pt 23): 5758-69, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22992460

RESUMEN

Cell chemotaxis, such as migration of fibroblasts towards growth factors during development and wound healing, requires precise spatial coordination of signalling events. Phosphoinositides and signalling enzymes involved in their generation and hydrolysis have been implicated in regulation of chemotaxis; however, the role and importance of specific components remain poorly understood. Here, we demonstrate that phospholipase C epsilon (PLCε) contributes to fibroblast chemotaxis towards platelet-derived growth factor (PDGF-BB). Using PLCe1 null fibroblasts we show that cells deficient in PLCε have greatly reduced directionality towards PDGF-BB without detrimental effect on their basal ability to migrate. Furthermore, we show that in intact fibroblasts, signalling events, such as activation of Rac, are spatially compromised by the absence of PLCε that affects the ability of cells to enlarge their protrusions in the direction of the chemoattractant. By further application of live cell imaging and the use of FRET-based biosensors, we show that generation of Ins(1,4,5)P(3) and recruitment of PLCε are most pronounced in protrusions responding to the PDGF-BB gradient. Furthermore, the phospholipase C activity of PLCε is critical for its role in chemotaxis, consistent with the importance of Ins(1,4,5)P(3) generation and sustained calcium responses in this process. As PLCε has extensive signalling connectivity, using transgenic fibroblasts we ruled out its activation by direct binding to Ras or Rap GTPases, and suggest instead new unexpected links for PLCε in the context of chemotaxis.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fosfoinositido Fosfolipasa C/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Células Cultivadas , Quimiotaxis/genética , Fibroblastos/citología , Ratones , Ratones Transgénicos , Fosfoinositido Fosfolipasa C/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética
12.
Cancer Cell ; 9(1): 33-44, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413470

RESUMEN

Inhibition of ERK-MAPK signaling by expression of dominant-negative MEK1 in the tumor vasculature suppresses angiogenesis and tumor growth. In an organotypic tissue culture angiogenesis assay, ERK-MAPK inhibition during the migratory phase results in loss of bipolarity, detachment, and cell death of isolated endothelial cells and retraction of sprouting tubules. These effects are the consequence of upregulated Rho-kinase signaling. Transient inhibition of Rho-kinase rescues the effects of ERK-MAPK inhibition in vitro and in vivo, promotes sprouting, and increases vessel length in tumors. We propose a regulatory role of Rho-kinase by ERK-MAPK during angiogenesis that acts through the control of actomyosin contractility. Our data delineate a mechanism by which ERK-MAPK promotes endothelial cell survival and sprouting by downregulating Rho-kinase signaling.


Asunto(s)
Células Endoteliales/fisiología , Endotelio Vascular/patología , MAP Quinasa Quinasa 1/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Neovascularización Patológica/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Actomiosina/metabolismo , Animales , Movimiento Celular , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/citología , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , MAP Quinasa Quinasa 1/genética , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neovascularización Patológica/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Transducción de Señal , Venas Umbilicales/citología , Quinasas Asociadas a rho
13.
Endocrinology ; 165(4)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38417844

RESUMEN

A series of well-described anabolic and catabolic neuropeptides are known to provide short-term, homeostatic control of energy balance. The mechanisms that govern long-term, rheostatic control of regulated changes in energy balance are less well characterized. Using the robust and repeatable seasonal changes in body mass observed in Siberian hamsters, this report examined the role of prolactin in providing long-term rheostatic control of body mass and photoinduced changes in organ mass (ie, kidney, brown adipose tissue, uterine, and spleen). Endogenous circannual interval timing was observed after 4 months in a short photoperiod, indicated by a significant increase in body mass and prolactin mRNA expression in the pituitary gland. There was an inverse relationship between body mass and the expression of somatostatin (Sst) and cocaine- and amphetamine-regulated transcript (Cart). Pharmacological inhibition of prolactin release (via bromocriptine injection), reduced body mass of animals maintained in long photoperiods to winter-short photoperiod levels and was associated with a significant increase in hypothalamic Cart expression. Administration of ovine prolactin significantly increased body mass 24 hours after a single injection and the effect persisted after 3 consecutive daily injections. The data indicate that prolactin has pleiotropic effects on homeostatic sensors of energy balance (ie, Cart) and physiological effectors (ie, kidney, BAT). We propose that prolactin release from the pituitary gland acts as an output signal of the hypothalamic rheostat controller to regulate adaptive changes in body mass.


Asunto(s)
Neuropéptidos , Prolactina , Cricetinae , Animales , Ovinos , Femenino , Prolactina/metabolismo , Estaciones del Año , Hipotálamo/metabolismo , Phodopus/metabolismo , Neuropéptidos/metabolismo , Fotoperiodo
14.
J Cell Sci ; 124(Pt 8): 1256-67, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21444753

RESUMEN

Melanoma cells can switch between an elongated mesenchymal-type and a rounded amoeboid-type migration mode. The rounded 'amoeboid' form of cell movement is driven by actomyosin contractility resulting in membrane blebbing. Unlike elongated A375 melanoma cells, rounded A375 cells do not display any obvious morphological front-back polarisation, although polarisation is thought to be a prerequisite for cell movement. We show that blebbing A375 cells are polarised, with ezrin (a linker between the plasma membrane and actin cytoskeleton), F-actin, myosin light chain, plasma membrane, phosphatidylinositol (4,5)-bisphosphate and ß1-integrin accumulating at the cell rear in a uropod-like structure. This structure does not have the typical protruding shape of classical leukocyte uropods, but, as for those structures, it is regulated by protein kinase C. We show that the ezrin-rich uropod-like structure (ERULS) is an inherent feature of polarised A375 cells and not a consequence of cell migration, and is necessary for cell invasion. Furthermore, we demonstrate that membrane blebbing is reduced at this site, leading to a model in which the rigid ezrin-containing structure determines the direction of a moving cell through localised inhibition of membrane blebbing.


Asunto(s)
Membrana Celular/metabolismo , Movimiento Celular , Polaridad Celular , Células/citología , Proteínas del Citoesqueleto/metabolismo , Melanoma/fisiopatología , Línea Celular Tumoral , Membrana Celular/genética , Células/metabolismo , Proteínas del Citoesqueleto/genética , Humanos , Melanoma/genética , Melanoma/metabolismo
15.
Cancer Cell ; 7(3): 219-26, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15766660

RESUMEN

To investigate the role of signaling by the small GTPase Ral, we have generated mice deficient for RalGDS, a guanine nucleotide exchange factor that activates Ral. We show that RalGDS is dispensable for mouse development but plays a substantial role in Ras-induced oncogenesis. Lack of RalGDS results in reduced tumor incidence, size, and progression to malignancy in multistage skin carcinogenesis, and reduced transformation by Ras in tissue culture. RalGDS does not appear to participate in the regulation of cell proliferation, but instead controls survival of transformed cells. Experiments performed in cells isolated from skin tumors suggest that RalGDS mediates cell survival through the activation of the JNK/SAPK pathway. These studies identify RalGDS as a key component in Ras-dependent carcinogenesis in vivo.


Asunto(s)
Transformación Celular Neoplásica , Transducción de Señal/fisiología , Neoplasias Cutáneas , Proteínas de Unión al GTP ral/metabolismo , Factor de Intercambio de Guanina Nucleótido ral/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteínas de Unión al GTP ral/genética , Factor de Intercambio de Guanina Nucleótido ral/genética , Proteínas ras/metabolismo
16.
Nat Rev Cancer ; 2(2): 133-42, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12635176

RESUMEN

The RAS oncogenes were identified almost 20 years ago. Since then, we have learnt that they are members of a large family of small GTPases that bind GTP and hydrolyse it to GDP. This is then exchanged for GTP and the cycle is repeated. The switching between these two states regulates a wide range of cellular processes. A branch of the RAS family--the RHO proteins--is also involved in cancer, but what is the role of these proteins and would they make good therapeutic targets?


Asunto(s)
Neoplasias/enzimología , Neoplasias/patología , Proteínas de Unión al GTP rho/metabolismo , Animales , Apoptosis , Ciclo Celular , Diseño de Fármacos , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Neoplasias/genética , Transducción de Señal , Proteínas de Unión al GTP rho/genética
17.
Elife ; 122023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38150309

RESUMEN

Annual cycles in daylength provide an initial predictive environmental cue that plants and animals use to time seasonal biology. Seasonal changes in photoperiodic information acts to entrain endogenous programs in physiology to optimize an animal's fitness. Attempts to identify the neural and molecular substrates of photoperiodic time measurement in birds have, to date, focused on blunt changes in light exposure during a restricted period of photoinducibility. The objectives of these studies were first to characterize a molecular seasonal clock in Japanese quail and second, to identify the key transcripts involved in endogenously generated interval timing that underlies photosensitivity in birds. We hypothesized that the mediobasal hypothalamus (MBH) provides the neuroendocrine control of photoperiod-induced changes in reproductive physiology, and that the pars distalis of the pituitary gland contains an endogenous internal timer for the short photoperiod-dependent development of reproductive photosensitivity. Here, we report distinct seasonal waveforms of transcript expression in the MBH, and pituitary gland and discovered the patterns were not synchronized across tissues. Follicle-stimulating hormone-ß (FSHß) expression increased during the simulated spring equinox, prior to photoinduced increases in prolactin, thyrotropin-stimulating hormone-ß, and testicular growth. Diurnal analyses of transcript expression showed sustained elevated levels of FSHß under conditions of the spring equinox, compared to autumnal equinox, short (<12L) and long (>12L) photoperiods. FSHß expression increased in quail held in non-stimulatory short photoperiod, indicative of the initiation of an endogenously programmed interval timer. These data identify that FSHß establishes a state of photosensitivity for the external coincidence timing of seasonal physiology. The independent regulation of FSHß expression provides an alternative pathway through which other supplementary environmental cues, such as temperature, can fine tune seasonal reproductive maturation and involution.


Asunto(s)
Coturnix , Hormona Folículo Estimulante de Subunidad beta , Fotoperiodo , Reproducción , Coturnix/fisiología , Hormona Folículo Estimulante de Subunidad beta/fisiología , Estaciones del Año , Masculino , Animales
18.
Nat Cell Biol ; 7(3): 255-61, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15723050

RESUMEN

Actomyosin contractility is a mechanism by which cells exert locomotory force against their environment. Signalling downstream of the small GTPase Rho increases contractility through Rho-kinase (ROCK)-mediated regulation of myosin-II light chain (MLC2) phosphorylation. Cdc42 signalling has been shown to control cell polarity. Tumour cells can move through a three-dimensional matrix with either a rounded morphology characterized by Rho-ROCK dependence or with an elongated morphology characterized by Rho-ROCK independence. Here we show that contractility necessary for elongated morphology and invasion can be generated by Cdc42-MRCK signalling. MRCK (myotonic dystrophy kinase-related Cdc42-binding kinase) cooperates with ROCK in the maintenance of elongated morphology and invasion and either MRCK or ROCK is sufficient for MLC2 phosphorylation, through the inhibitory phosphorylation of myosin phosphatase. By contrast, in rounded ROCK-dependent movement, where MLC2 phosphorylation is higher, MRCK has a smaller role. Our data show that a Cdc42-MRCK signal mediates myosin-dependent cell motility and highlight convergence between Rho and Cdc42 signalling.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Actomiosina/química , Animales , Bovinos , Movimiento Celular , Colágeno/química , Vectores Genéticos , Proteínas Fluorescentes Verdes , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Immunoblotting , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Microscopía de Contraste de Fase , Miosinas/química , Proteína Quinasa de Distrofia Miotónica , Invasividad Neoplásica , Fosforilación , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Quinasas Asociadas a rho
19.
Arterioscler Thromb Vasc Biol ; 31(3): 657-64, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148427

RESUMEN

OBJECTIVE: RhoJ/TCL was identified by our group as an endothelial-expressed Rho GTPase. The aim of this study was to determine its tissue distribution, subcellular localization, and function in endothelial migration and tube formation. METHODS AND RESULTS: Using in situ hybridization, RhoJ was localized to endothelial cells in a set of normal and cancerous tissues and in the vasculature of mouse embryos; endogenous RhoJ was localized to focal adhesions by immunofluorescence. The proangiogenic factor vascular endothelial growth factor activated RhoJ in endothelial cells. Using either small interfering (si)RNA-mediated knockdown of RhoJ expression or overexpression of constitutively active RhoJ (daRhoJ), RhoJ was found to positively regulate endothelial motility and tubule formation. Downregulating RhoJ expression increased focal adhesions and stress fibers in migrating cells, whereas daRhoJ overexpression resulted in the converse. RhoJ downregulation resulted in increased contraction of a collagen gel and increased phospho-myosin light chain, indicative of increased actomyosin contractility. Pharmacological inhibition of Rho-kinase (which phosphorylates myosin light chain) or nonmuscle myosin II reversed the defective tube formation and migration of RhoJ knockdown cells. CONCLUSIONS: RhoJ is endothelial-expressed in vivo, activated by vascular endothelial growth factor, localizes to focal adhesions, regulates endothelial cell migration and tube formation, and modulates actomyosin contractility and focal adhesion numbers.


Asunto(s)
Actomiosina/metabolismo , Movimiento Celular , Células Endoteliales/enzimología , Adhesiones Focales/metabolismo , GTP Fosfohidrolasas/metabolismo , Neovascularización Fisiológica , Proteínas de Unión al GTP rho/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Forma de la Célula , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , GTP Fosfohidrolasas/genética , Humanos , Hibridación in Situ , Ratones , Cadenas Ligeras de Miosina/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Fibras de Estrés/metabolismo , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
20.
Cancer Cell ; 4(1): 67-79, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12892714

RESUMEN

We describe two signaling events downstream of ERK-MAP kinase contributing to cell motility in colon carcinoma cells. The Fos family member Fra-1 is expressed in an ERK-dependent manner. Silencing of Fra-1 expression with short interfering RNAs leads to losses of cell polarization, motility, and invasiveness in vitro. These effects of ablating Fra-1 are a consequence of activation of a RhoA-ROCK pathway by beta1-integrin, leading to an increase in the amount of stress fibers and stabilization of focal adhesions. We propose that Fra-1 promotes cell motility by inactivating beta1-integrin and keeping RhoA activity low. This depression of RhoA activity is necessary to permit a second ERK-dependent signaling event via uPAR, the receptor for urokinase-type plasminogen activator, to activate Rac and to drive motility through polarized lamellipodia extension.


Asunto(s)
Movimiento Celular , Neoplasias del Colon/patología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Polaridad Celular , Colágeno/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Combinación de Medicamentos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Integrina beta1/metabolismo , Laminina/metabolismo , Invasividad Neoplásica , Proteoglicanos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/farmacología , Seudópodos/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Tumorales Cultivadas , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA