Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(18): e2322692121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652744

RESUMEN

Food intake and energy balance are tightly regulated by a group of hypothalamic arcuate neurons expressing the proopiomelanocortin (POMC) gene. In mammals, arcuate-specific POMC expression is driven by two cis-acting transcriptional enhancers known as nPE1 and nPE2. Because mutant mice lacking these two enhancers still showed hypothalamic Pomc mRNA, we searched for additional elements contributing to arcuate Pomc expression. By combining molecular evolution with reporter gene expression in transgenic zebrafish and mice, here, we identified a mammalian arcuate-specific Pomc enhancer that we named nPE3, carrying several binding sites also present in nPE1 and nPE2 for transcription factors known to activate neuronal Pomc expression, such as ISL1, NKX2.1, and ERα. We found that nPE3 originated in the lineage leading to placental mammals and remained under purifying selection in all mammalian orders, although it was lost in Simiiformes (monkeys, apes, and humans) following a unique segmental deletion event. Interestingly, ablation of nPE3 from the mouse genome led to a drastic reduction (>70%) in hypothalamic Pomc mRNA during development and only moderate (<33%) in adult mice. Comparison between double (nPE1 and nPE2) and triple (nPE1, nPE2, and nPE3) enhancer mutants revealed the relative contribution of nPE3 to hypothalamic Pomc expression and its importance in the control of food intake and adiposity in male and female mice. Altogether, these results demonstrate that nPE3 integrates a tripartite cluster of partially redundant enhancers that originated upon a triple convergent evolutionary process in mammals and that is critical for hypothalamic Pomc expression and body weight homeostasis.


Asunto(s)
Peso Corporal , Ingestión de Alimentos , Elementos de Facilitación Genéticos , Hipotálamo , Proopiomelanocortina , Pez Cebra , Animales , Proopiomelanocortina/metabolismo , Proopiomelanocortina/genética , Ratones , Hipotálamo/metabolismo , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Pez Cebra/genética , Pez Cebra/metabolismo , Femenino , Masculino , Ratones Transgénicos , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Mamíferos/metabolismo , Mamíferos/genética
3.
Int J Dev Biol ; 65(4-5-6): 195-205, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32930382

RESUMEN

Rax (Rx) genes encode paired-type homeodomain-containing transcription factors present in virtually all metazoan groups. In vertebrates, studies in fish, amphibian, chick and mouse models have revealed that these genes play important roles in the development of structures located at the anterior portion of the central nervous system, in particular the eyes, the hypothalamus and the pituitary gland. In addition, human patients with eye and brain defects carry mutations in the two human Rax paralogues, RAX and RAX2. Here, we review work done in the last years on Rax genes, focusing especially on the function that mouse Rax and its zebrafish homologue, rx3, play in hypothalamic and pituitary development. Work on both of these model organisms indicate that Rax genes are necessary for the patterning, growth and differentiation of the hypothalamus, in particular the ventro-tuberal and dorso-anterior hypothalamus, where they effect their action by controlling expression of the secreted signalling protein, Sonic hedgehog (Shh). In addition, Rax/rx3 mutations disturb the development of the pituitary gland, mimicking phenotypes observed in human subjects carrying mutations in the RAX gene. Thus, along with their crucial role in eye morphogenesis, Rax genes play a conserved role in the development of the hypothalamus and adjacent structures in the vertebrate clade.


Asunto(s)
Proteínas del Ojo , Proteínas de Homeodominio , Hipotálamo/crecimiento & desarrollo , Hipófisis/crecimiento & desarrollo , Factores de Transcripción , Pez Cebra , Animales , Proteínas del Ojo/fisiología , Proteínas Hedgehog/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Factores de Transcripción/fisiología , Pez Cebra/genética , Pez Cebra/fisiología
4.
J Neurosci ; 39(21): 4023-4035, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-30886014

RESUMEN

Food intake is tightly regulated by a group of neurons present in the arcuate nucleus of the hypothalamus, which release Pomc-encoded melanocortins, the absence of which induces marked hyperphagia and early-onset obesity. Although the relevance of hypothalamic POMC neurons in the regulation of body weight and energy balance is well appreciated, little is known about the transcription factors that establish the melanocortin neuron identity during brain development and its phenotypic maintenance in postnatal life. Here, we report that the transcription factor NKX2.1 is present in mouse hypothalamic POMC neurons from early development to adulthood. Electromobility shift assays showed that NKX2.1 binds in vitro to NKX binding motifs present in the neuronal Pomc enhancers nPE1 and nPE2 and chromatin immunoprecipitation assays detected in vivo binding of NKX2.1 to nPE1 and nPE2 in mouse hypothalamic extracts. Transgenic and mutant studies performed in mouse embryos of either sex and adult males showed that the NKX motifs present in nPE1 and nPE2 are essential for their transcriptional enhancer activity. The conditional early inactivation of Nkx2.1 in the ventral hypothalamus prevented the onset of Pomc expression. Selective Nkx2.1 ablation from POMC neurons decreased Pomc expression in adult males and mildly increased their body weight and adiposity. Our results demonstrate that NKX2.1 is necessary to activate Pomc expression by binding to conserved canonical NKX motifs present in nPE1 and nPE2. Therefore, NKX2.1 plays a critical role in the early establishment of hypothalamic melanocortin neuron identity and participates in the maintenance of Pomc expression levels during adulthood.SIGNIFICANCE STATEMENT Food intake and body weight regulation depend on hypothalamic neurons that release satiety-inducing neuropeptides, known as melanocortins. Central melanocortins are encoded byPomc, and Pomc mutations may lead to hyperphagia and severe obesity. Although the importance of central melanocortins is well appreciated, the genetic program that establishes and maintains fully functional POMC neurons remains to be explored. Here, we combined molecular, genetic, developmental, and functional studies that led to the discovery of NKX2.1, a transcription factor that participates in the early morphogenesis of the developing hypothalamus, as a key player in establishing the early identity of melanocortin neurons by activating Pomc expression. Thus, Nkx2.1 adds to the growing list of genes that participate in body weight regulation and adiposity.


Asunto(s)
Melanocortinas/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Factor Nuclear Tiroideo 1/metabolismo , Animales , Peso Corporal/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipotálamo/embriología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
5.
PLoS One ; 12(5): e0177573, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28494025

RESUMEN

MicroRNAs (miRNAs) are short, single stranded RNA molecules that regulate the stability and translation of messenger RNAs in diverse eukaryotic groups. Several miRNA genes are of ancient origin and have been maintained in the genomes of animal and plant taxa for hundreds of millions of years, playing key roles in development and physiology. In the last decade, genome and small RNA (sRNA) sequencing of several plant species have helped unveil the evolutionary history of land plants. Among these, the fern group (monilophytes) occupies a key phylogenetic position, as it represents the closest extant cousin taxon of seed plants, i.e. gymno- and angiosperms. However, in spite of their evolutionary, economic and ecological importance, no fern genome has been sequenced yet and few genomic resources are available for this group. Here, we sequenced the small RNA fraction of an epiphytic South American fern, Pleopeltis minima (Polypodiaceae), and compared it to plant miRNA databases, allowing for the identification of miRNA families that are shared by all land plants, shared by all vascular plants (tracheophytes) or shared by euphyllophytes (ferns and seed plants) only. Using the recently described transcriptome of another fern, Lygodium japonicum, we also estimated the degree of conservation of fern miRNA targets in relation to other plant groups. Our results pinpoint the origin of several miRNA families in the land plant evolutionary tree with more precision and are a resource for future genomic and functional studies of fern miRNAs.


Asunto(s)
Evolución Molecular , Helechos/genética , MicroARNs/genética , ARN de Planta/genética , Análisis de Secuencia de ARN/métodos , Secuencia de Bases , Secuencia Conservada/genética , MicroARNs/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Planta/metabolismo
6.
Mech Dev ; 144(Pt B): 163-170, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27838261

RESUMEN

Rax proteins comprise a small family of paired-type, homeodomain-containing transcription factors with essential functions in eye and forebrain development. While invertebrates possess only one Rax gene, vertebrates can have several Rax paralogue genes, but the evolutionary history of the members of the family has not been studied in detail. Here, we present a thorough analysis of the evolutionary relationships between vertebrate Rax genes and proteins available in diverse genomic databases. Phylogenetic and synteny analyses indicate that Rax genes went through a duplication in an ancestor of all jawed vertebrates (Gnathostomata), giving rise to the ancestral vertebrate Rax1 and Rax2 genes. This duplication event is likely related to the proposed polyploidisations that occurred during early vertebrate evolution. Subsequent genome-wide duplications in the lineage of ray-finned fish (Actinopterygii) originated new Rax2 paralogues in the genomes of teleosts. In the lobe-finned fish lineage (Sarcopterygii), the N-terminal octapeptide domain of Rax2 was lost in a common ancestor of tetrapods, giving rise to a shorter version of Rax2 in this lineage. Within placental mammals, the Rax2 gene was lost altogether in an ancestor of rodents and lagomorphs (Glires). Finally, we discuss the scientific literature in the light of Rax gene evolution and propose new avenues of research on the function of this important family of transcriptional regulators.


Asunto(s)
Evolución Molecular , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Filogenia , Sintenía , Vertebrados/genética
7.
Dev Biol ; 416(1): 212-224, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27212025

RESUMEN

The hypothalamus is a region of the anterior forebrain that controls basic aspects of vertebrate physiology, but the genes involved in its development are still poorly understood. Here, we investigate the function of the homeobox gene Rax/Rx in early hypothalamic development using a conditional targeted inactivation strategy in the mouse. We found that lack of Rax expression prior to embryonic day 8.5 (E8.5) caused a general underdevelopment of the hypothalamic neuroepithelium, while inactivation at later timepoints had little effect. The early absence of Rax impaired neurogenesis and prevented the expression of molecular markers of the dorsomedial hypothalamus, including neuropeptides Proopiomelanocortin and Somatostatin. Interestingly, the expression domains of genes expressed in the ventromedial hypothalamus and infundibulum invaded dorsal hypothalamic territory, showing that Rax is needed for the proper dorsoventral patterning of the developing medial hypothalamus. The phenotypes caused by the early loss of Rax are similar to those of eliminating the expression of the morphogen Sonic hedgehog (Shh) specifically from the hypothalamus. Consistent with this similarity in phenotypes, we observed that Shh and Rax are coexpressed in the rostral forebrain at late head fold stages and that loss of Rax caused a downregulation of Shh expression in the dorsomedial portion of the hypothalamus.


Asunto(s)
Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Hipotálamo/embriología , Factores de Transcripción/fisiología , Animales , Tipificación del Cuerpo , Desarrollo Embrionario/genética , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Noqueados , Factores de Transcripción/genética
8.
Sci Rep ; 6: 19036, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26750147

RESUMEN

The plastid organelle comprises a high proportion of nucleus-encoded proteins that were acquired from different prokaryotic donors via independent horizontal gene transfers following its primary endosymbiotic origin. What forces drove the targeting of these alien proteins to the plastid remains an unresolved evolutionary question. To better understand this process we screened for suitable candidate proteins to recapitulate their prokaryote-to-eukaryote transition. Here we identify the ancient horizontal transfer of a bacterial polyphenol oxidase (PPO) gene to the nuclear genome of an early land plant ancestor and infer the possible mechanism behind the plastidial localization of the encoded enzyme. Arabidopsis plants expressing PPO versions either lacking or harbouring a plastid-targeting signal allowed examining fitness consequences associated with its subcellular localization. Markedly, a deleterious effect on plant growth was highly correlated with PPO activity only when producing the non-targeted enzyme, suggesting that selection favoured the fixation of plastid-targeted protein versions. Our results reveal a possible evolutionary mechanism of how selection against heterologous genes encoding cytosolic proteins contributed in incrementing plastid proteome complexity from non-endosymbiotic gene sources, a process that may also impact mitochondrial evolution.


Asunto(s)
Arabidopsis/genética , Evolución Biológica , Catecol Oxidasa/genética , Transferencia de Gen Horizontal , Genoma de Planta , Plastidios/genética , Arabidopsis/clasificación , Arabidopsis/enzimología , Bacterias/clasificación , Bacterias/enzimología , Bacterias/genética , Catecol Oxidasa/metabolismo , Núcleo Celular/enzimología , Núcleo Celular/genética , Chlorophyta/clasificación , Chlorophyta/enzimología , Chlorophyta/genética , Células Eucariotas/citología , Hongos/clasificación , Hongos/enzimología , Hongos/genética , Expresión Génica , Modelos Moleculares , Filogenia , Plastidios/enzimología , Células Procariotas/citología , Células Procariotas/enzimología , Señales de Clasificación de Proteína , Transporte de Proteínas , Selección Genética , Simbiosis/fisiología
9.
Proc Natl Acad Sci U S A ; 112(15): E1861-70, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825735

RESUMEN

Food intake and body weight regulation depend on proper expression of the proopiomelanocortin gene (Pomc) in a group of neurons located in the mediobasal hypothalamus of all vertebrates. These neurons release POMC-encoded melanocortins, which are potent anorexigenic neuropeptides, and their absence from mice or humans leads to hyperphagia and severe obesity. Although the pathophysiology of hypothalamic POMC neurons is well understood, the genetic program that establishes the neuronal melanocortinergic phenotype and maintains a fully functional neuronal POMC phenotype throughout adulthood remains unknown. Here, we report that the early expression of the LIM-homeodomain transcription factor Islet 1 (ISL1) in the developing hypothalamus promotes the terminal differentiation of melanocortinergic neurons and is essential for hypothalamic Pomc expression since its initial onset and throughout the entire lifetime. We detected ISL1 in the prospective hypothalamus just before the onset of Pomc expression and, from then on, Pomc and Isl1 coexpress. ISL1 binds in vitro and in vivo to critical homeodomain binding DNA motifs present in the neuronal Pomc enhancers nPE1 and nPE2, and mutations of these sites completely disrupt the ability of these enhancers to drive reporter gene expression to hypothalamic POMC neurons in transgenic mice and zebrafish. ISL1 is necessary for hypothalamic Pomc expression during mouse and zebrafish embryogenesis. Furthermore, conditional Isl1 inactivation from POMC neurons impairs Pomc expression, leading to hyperphagia and obesity. Our results demonstrate that ISL1 specifies the identity of hypothalamic melanocortin neurons and is required for melanocortin-induced satiety and normal adiposity throughout the entire lifespan.


Asunto(s)
Adiposidad/fisiología , Ingestión de Alimentos/fisiología , Proteínas con Homeodominio LIM/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Factores de Transcripción/metabolismo , Adiposidad/genética , Animales , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Ingestión de Alimentos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hiperfagia/genética , Hiperfagia/fisiopatología , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/metabolismo , Proteínas con Homeodominio LIM/genética , Masculino , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Datos de Secuencia Molecular , Neuronas/citología , Obesidad/genética , Obesidad/fisiopatología , Proopiomelanocortina/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/genética , Pez Cebra/embriología , Pez Cebra/metabolismo
10.
PLoS Genet ; 11(2): e1004935, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25671638

RESUMEN

Cell-specific expression of many genes is conveyed by multiple enhancers, with each individual enhancer controlling a particular expression domain. In contrast, multiple enhancers drive similar expression patterns of some genes involved in embryonic development, suggesting regulatory redundancy. Work in Drosophila has indicated that functionally overlapping enhancers canalize development by buffering gene expression against environmental and genetic disturbances. However, little is known about regulatory redundancy in vertebrates and in genes mainly expressed during adulthood. Here we study nPE1 and nPE2, two phylogenetically conserved mammalian enhancers that drive expression of the proopiomelanocortin gene (Pomc) to the same set of hypothalamic neurons. The simultaneous deletion of both enhancers abolished Pomc expression at all ages and induced a profound metabolic dysfunction including early-onset extreme obesity. Targeted inactivation of either nPE1 or nPE2 led to very low levels of Pomc expression during early embryonic development indicating that both enhancers function synergistically. In adult mice, however, Pomc expression is controlled additively by both enhancers, with nPE1 being responsible for ∼80% and nPE2 for ∼20% of Pomc transcription. Consequently, nPE1 knockout mice exhibit mild obesity whereas nPE2-deficient mice maintain a normal body weight. These results suggest that nPE2-driven Pomc expression is compensated by nPE1 at later stages of development, essentially rescuing the earlier phenotype of nPE2 deficiency. Together, these results reveal that cooperative interactions between the enhancers confer robustness of Pomc expression against gene regulatory disturbances and preclude deleterious metabolic phenotypes caused by Pomc deficiency in adulthood. Thus, our study demonstrates that enhancer redundancy can be used by genes that control adult physiology in mammals and underlines the potential significance of regulatory sequence mutations in common diseases.


Asunto(s)
Desarrollo Embrionario/genética , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Proopiomelanocortina/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Secuencia Conservada , Femenino , Regulación del Desarrollo de la Expresión Génica , Mamíferos/genética , Ratones , Neuronas/metabolismo , Filogenia , Embarazo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética
11.
Philos Trans R Soc Lond B Biol Sci ; 368(1632): 20130017, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24218630

RESUMEN

Deciphering the genetic bases that drive animal diversity is one of the major challenges of modern biology. Although four decades ago it was proposed that animal evolution was mainly driven by changes in cis-regulatory DNA elements controlling gene expression rather than in protein-coding sequences, only now are powerful bioinformatics and experimental approaches available to accelerate studies into how the evolution of transcriptional enhancers contributes to novel forms and functions. In the introduction to this Theme Issue, we start by defining the general properties of transcriptional enhancers, such as modularity and the coexistence of tight sequence conservation with transcription factor-binding site shuffling as different mechanisms that maintain the enhancer grammar over evolutionary time. We discuss past and current methods used to identify cell-type-specific enhancers and provide examples of how enhancers originate de novo, change and are lost in particular lineages. We then focus in the central part of this Theme Issue on analysing examples of how the molecular evolution of enhancers may change form and function. Throughout this introduction, we present the main findings of the articles, reviews and perspectives contributed to this Theme Issue that together illustrate some of the great advances and current frontiers in the field.


Asunto(s)
Biodiversidad , Biología Computacional/métodos , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Animales , Sitios de Unión/genética , Biología Computacional/tendencias , Secuencia Conservada/genética , Humanos
12.
Philos Trans R Soc Lond B Biol Sci ; 368(1632): 20130027, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24218639

RESUMEN

Mutations in regulatory regions including enhancers are an important source of variation and innovation during evolution. Enhancers can evolve by changes in the sequence, arrangement and repertoire of transcription factor binding sites, but whole enhancers can also be lost or gained in certain lineages in a process of turnover. The proopiomelanocortin gene (Pomc), which encodes a prohormone, is expressed in the pituitary and hypothalamus of all jawed vertebrates. We have previously described that hypothalamic Pomc expression in mammals is controlled by two enhancers-nPE1 and nPE2-that are derived from transposable elements and that presumably replaced the ancestral neuronal Pomc regulatory regions. Here, we show that nPE1 and nPE2, even though they are mammalian novelties with no homologous counterpart in other vertebrates, nevertheless can drive gene expression specifically to POMC neurons in the hypothalamus of larval and adult transgenic zebrafish. This indicates that when neuronal Pomc enhancers originated de novo during early mammalian evolution, the newly created cis- and trans-codes were similar to the ancestral ones. We also identify the neuronal regulatory region of zebrafish pomca and confirm that it is not homologous to the mammalian enhancers. Our work sheds light on the process of gene regulatory evolution by showing how a locus can undergo enhancer turnover and nevertheless maintain the ancestral transcriptional output.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Evolución Molecular , Regulación de la Expresión Génica/genética , Proopiomelanocortina/genética , Vertebrados/genética , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Biología Computacional , Secuencia Conservada/genética , Inmunohistoquímica , Hibridación in Situ , Mutación/genética , Neuronas/metabolismo , Pez Cebra
13.
Mol Biol Evol ; 30(6): 1239-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23486611

RESUMEN

Transposable elements (TEs) are mobile genetic sequences that can jump around the genome from one location to another, behaving as genomic parasites. TEs have been particularly effective in colonizing mammalian genomes, and such heavy TE load is expected to have conditioned genome evolution. Indeed, studies conducted both at the gene and genome levels have uncovered TE insertions that seem to have been co-opted--or exapted--by providing transcription factor binding sites (TFBSs) that serve as promoters and enhancers, leading to the hypothesis that TE exaptation is a major factor in the evolution of gene regulation. Here, we critically review the evidence for exaptation of TE-derived sequences as TFBSs, promoters, enhancers, and silencers/insulators both at the gene and genome levels. We classify the functional impact attributed to TE insertions into four categories of increasing complexity and argue that so far very few studies have conclusively demonstrated exaptation of TEs as transcriptional regulatory regions. We also contend that many genome-wide studies dealing with TE exaptation in recent lineages of mammals are still inconclusive and that the hypothesis of rapid transcriptional regulatory rewiring mediated by TE mobilization must be taken with caution. Finally, we suggest experimental approaches that may help attributing higher-order functions to candidate exapted TEs.


Asunto(s)
Elementos Transponibles de ADN , Regulación de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Línea Celular , Embrión de Mamíferos , Elementos de Facilitación Genéticos , Humanos , Ratones , Filogenia
14.
J Clin Invest ; 122(11): 4203-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23093774

RESUMEN

Obesity is a chronic metabolic disorder affecting half a billion people worldwide. Major difficulties in managing obesity are the cessation of continued weight loss in patients after an initial period of responsiveness and rebound to pretreatment weight. It is conceivable that chronic weight gain unrelated to physiological needs induces an allostatic regulatory state that defends a supranormal adipose mass despite its maladaptive consequences. To challenge this hypothesis, we generated a reversible genetic mouse model of early-onset hyperphagia and severe obesity by selectively blocking the expression of the proopiomelanocortin gene (Pomc) in hypothalamic neurons. Eutopic reactivation of central POMC transmission at different stages of overweight progression normalized or greatly reduced food intake in these obesity-programmed mice. Hypothalamic Pomc rescue also attenuated comorbidities such as hyperglycemia, hyperinsulinemia, and hepatic steatosis and normalized locomotor activity. However, effectiveness of treatment to normalize body weight and adiposity declined progressively as the level of obesity at the time of Pomc induction increased. Thus, our study using a novel reversible monogenic obesity model reveals the critical importance of early intervention for the prevention of subsequent allostatic overload that auto-perpetuates obesity.


Asunto(s)
Tejido Adiposo/fisiopatología , Adiposidad , Ingestión de Alimentos , Hipotálamo/fisiopatología , Obesidad/prevención & control , Obesidad/fisiopatología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Modelos Animales de Enfermedad , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/patología , Hiperfagia/fisiopatología , Hiperfagia/prevención & control , Hipotálamo/metabolismo , Hipotálamo/patología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Obesidad/genética , Obesidad/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo
15.
Mob Genet Elements ; 2(2): 106-109, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22934245

RESUMEN

The proopiomelanocortin (Pomc) gene encodes a prepropeptide with essential functions in the response to stress and energy balance, which is expressed in the pituitary and hypothalamus of vertebrate animals. Neuronal expression of Pomc is controlled by two distal enhancers named nPE1 and nPE2. Using transgenic mice, we observed that both enhancers drive identical expression patterns in the mammalian hypothalamus, starting at embryonic day 10.5, when endogenous Pomc expression commences. This overlapping enhancer activity is maintained throughout hypothalamic development and into adulthood. We also found that nPE1 and nPE2 were exapted as neuronal enhancers into the POMC locus after the sequential insertion of two unrelated retroposons. Thus, nPE1 and nPE2 are functional analogs and represent an authentic first example of convergent molecular evolution of cell-specific transcriptional enhancers. In this Commentary we discuss the following questions that remain unanswered: (1) how does transcriptional control of POMC operate in hypothalamic neurons of non-mammalian vertebrates? (2) What evolutionary forces are maintaining two discrete neuronal POMC enhancers under purifying selection for the last ~100 million years in all placental mammals? (3) What is the contribution of MaLRs to genome evolution?

16.
Eur J Pharmacol ; 660(1): 181-7, 2011 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-21211522

RESUMEN

The gene encoding the prohormone proopiomelanocortin (POMC) is mainly expressed in two regions in vertebrates, namely corticotrophs and melanotrophs in the pituitary and a small population of neurons in the arcuate nucleus of the hypothalamus. In this latter region, POMC-derived peptides participate in the control of energy balance and sensitivity to pain. Neuronal expression of POMC is conferred by two enhancers, nPE1 and nPE2, which are conserved in most mammals, but no transcription factors are yet known to bind to these enhancers. In this work, by means of a one-hybrid screening, we identify that nPE2 possesses an element recognized by transcription factors of the nuclear receptor superfamily. This element, named NRBE, is conserved in all known nPE2 enhancers and is necessary to confer full enhancer strength to nPE2-driven reporter gene expression in transgenic mice assays, indicating that the phylogenetic conservation of the element is indicative of its functional importance. In a search for candidate nuclear receptors that might control POMC we observed that estrogen receptor alpha (ESR1) - a known regulator of energy balance at the hypothalamic level - can bind to the NRBE element in vitro. In addition we observed by immunofluorescence that ESR1 is coexpressed with POMC in around 25-30% of hypothalamic neurons of males and females during late embryonic stages and adulthood. Thus, our results indicate that hypothalamic expression of POMC is controlled by nuclear receptors and establish ESR1 as a candidate regulator of POMC.


Asunto(s)
Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Receptor alfa de Estrógeno/metabolismo , Hipotálamo/citología , Neuronas/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Animales , Secuencia de Bases , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Unión Proteica , Transporte de Proteínas
17.
J Assoc Res Otolaryngol ; 10(3): 397-406, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19452222

RESUMEN

Efferent inhibition of cochlear hair cells is mediated by alpha9alpha10 nicotinic cholinergic receptors (nAChRs) functionally coupled to calcium-activated, small conductance (SK2) potassium channels. Before the onset of hearing, efferent fibers transiently make functional cholinergic synapses with inner hair cells (IHCs). The retraction of these fibers after the onset of hearing correlates with the cessation of transcription of the Chrna10 (but not the Chrna9) gene in IHCs. To further analyze this developmental change, we generated a transgenic mice whose IHCs constitutively express alpha10 into adulthood by expressing the alpha10 cDNA under the control of the Pou4f3 gene promoter. In situ hybridization showed that the alpha10 mRNA is expressed in IHCs of 8-week-old transgenic mice, but not in wild-type mice. Moreover, this mRNA is translated into a functional protein, since IHCs from P8-P10 alpha10 transgenic mice backcrossed to a Chrna10(-/-) background (whose IHCs have no cholinergic function) displayed normal synaptic and acetylcholine (ACh)-evoked currents in patch-clamp recordings. Thus, the alpha10 transgene restored nAChR function. However, in the alpha10 transgenic mice, no synaptic or ACh-evoked currents were observed in P16-18 IHCs, indicating developmental down-regulation of functional nAChRs after the onset of hearing, as normally observed in wild-type mice. The lack of functional ACh currents correlated with the lack of SK2 currents. These results indicate that multiple features of the efferent postsynaptic complex to IHCs, in addition to the nAChR subunits, are down-regulated in synchrony after the onset of hearing, leading to lack of responses to ACh.


Asunto(s)
Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/metabolismo , Audición/fisiología , Receptores Nicotínicos/metabolismo , Acetilcolina/farmacología , Animales , Colinérgicos/farmacología , Células Ciliadas Auditivas Internas/efectos de los fármacos , Audición/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Modelos Animales , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Factor de Transcripción Brn-3C/genética , Factor de Transcripción Brn-3C/metabolismo
18.
PLoS Genet ; 3(10): 1813-26, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17922573

RESUMEN

The proopiomelanocortin gene (POMC) is expressed in the pituitary gland and the ventral hypothalamus of all jawed vertebrates, producing several bioactive peptides that function as peripheral hormones or central neuropeptides, respectively. We have recently determined that mouse and human POMC expression in the hypothalamus is conferred by the action of two 5' distal and unrelated enhancers, nPE1 and nPE2. To investigate the evolutionary origin of the neuronal enhancer nPE2, we searched available vertebrate genome databases and determined that nPE2 is a highly conserved element in placentals, marsupials, and monotremes, whereas it is absent in nonmammalian vertebrates. Following an in silico paleogenomic strategy based on genome-wide searches for paralog sequences, we discovered that opossum and wallaby nPE2 sequences are highly similar to members of the superfamily of CORE-short interspersed nucleotide element (SINE) retroposons, in particular to MAR1 retroposons that are widely present in marsupial genomes. Thus, the neuronal enhancer nPE2 originated from the exaptation of a CORE-SINE retroposon in the lineage leading to mammals and remained under purifying selection in all mammalian orders for the last 170 million years. Expression studies performed in transgenic mice showed that two nonadjacent nPE2 subregions are essential to drive reporter gene expression into POMC hypothalamic neurons, providing the first functional example of an exapted enhancer derived from an ancient CORE-SINE retroposon. In addition, we found that this CORE-SINE family of retroposons is likely to still be active in American and Australian marsupial genomes and that several highly conserved exonic, intronic and intergenic sequences in the human genome originated from the exaptation of CORE-SINE retroposons. Together, our results provide clear evidence of the functional novelties that transposed elements contributed to their host genomes throughout evolution.


Asunto(s)
Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Mamíferos/genética , Neuronas/metabolismo , Proopiomelanocortina/genética , Elementos de Nucleótido Esparcido Corto/genética , Animales , Secuencia de Bases , Biología Computacional , Secuencia de Consenso , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Genómica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Zarigüeyas/genética , Paleontología , Proopiomelanocortina/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Eliminación de Secuencia
19.
Mol Endocrinol ; 21(11): 2738-49, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17698954

RESUMEN

The stress response involves complex physiological mechanisms that maximize behavioral efficacy during attack or defense and is highly conserved in all vertebrates. Key mediators of the stress response are pituitary hormones encoded by the proopiomelanocortin gene (POMC). Despite conservation of physiological function and expression pattern of POMC in all vertebrates, phylogenetic footprinting analyses at the POMC locus across vertebrates failed to detect conserved noncoding sequences with potential regulatory function. To investigate whether ortholog POMC promoters from extremely distant vertebrates are functionally conserved, we used 5'-flanking sequences of the teleost fish Tetraodon nigroviridis POMCalpha gene to produce transgenic mice. Tetraodon POMCalpha promoter targeted reporter gene expression exclusively to mouse pituitary cells that normally express Pomc. Importantly, transgenic expression in mouse corticotrophs was increased after adrenalectomy. To understand how conservation of precise gene expression mechanisms coexists with great sequence divergence, we investigated whether very short elements are still conserved in all vertebrate POMC promoters. Multiple local sequence alignments that consider phylogenetic relationships of ortholog regions identified a unique 10-bp motif GTGCTAA(T/G)CC that is usually present in two copies in POMC 5'-flanking sequences of all vertebrates. Underlined nucleotides represent totally conserved sequences. Deletion of these paired motifs from Tetraodon POMCalpha promoter markedly reduced its transcriptional activity in a mouse corticotropic cell line and in pituitary POMC cells of transgenic mice. In mammals, the conserved motifs correspond to reported binding sites for pituitary-specific nuclear proteins that participate in POMC transcriptional regulation. Together, these results demonstrate that mechanisms that participate in pituitary-specific and hormonally regulated expression of POMC have been preserved since mammals and teleosts diverged from a common ancestor 450 million years ago despite great promoter sequence divergence.


Asunto(s)
Regulación de la Expresión Génica , Hipófisis/metabolismo , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/genética , Regiones Promotoras Genéticas , Transcripción Genética , Secuencias de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Peces , Ratones , Ratones Transgénicos , Filogenia , Especificidad de la Especie
20.
Mol Biol Evol ; 22(12): 2417-27, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16093565

RESUMEN

The proopiomelanocortin gene (POMC) encodes several bioactive peptides, including adrenocorticotropin hormone, alpha-, beta-, and gamma-melanocyte-stimulating hormone, and the opioid peptide beta-endorphin, which play key roles in vertebrate physiology. In the human, mouse, and chicken genomes, there is only one POMC gene. By searching public genome projects, we have found that Tetraodon (Tetraodon nigroviridis), Fugu (Takifugu rubripes), and zebrafish (Danio rerio) possess two POMC genes, which we called POMCalpha and POMCbeta, and we present phylogenetic and mapping evidence that these paralogue genes originated in the whole-genome duplication specific to the teleost lineage over 300 MYA. In addition, we present evidence for two types of subfunction partitioning between the paralogues. First, in situ hybridization experiments indicate that the expression domains of the ancestral POMC gene have been subfunctionalized in Tetraodon, with POMCalpha expressed in the nucleus lateralis tuberis of the hypothalamus, as well as in the rostral pars distalis and pars intermedia (PI) of the pituitary, whereas POMCbeta is expressed in the preoptic area of the brain and weakly in the pituitary PI. Second, POMCbeta genes have a beta-endorphin segment that lacks the consensus opioid signal and seems to be under neutral evolution in tetraodontids, whereas POMCalpha genes possess well-conserved peptide regions. Thus, POMC paralogues have experienced subfunctionalization of both expression and peptide domains during teleost evolution. The study of regulatory regions of fish POMC genes might shed light on the mechanisms of enhancer partitioning between duplicate genes, as well as the roles of POMC-derived peptides in fish physiology.


Asunto(s)
Duplicación de Gen , Filogenia , Proopiomelanocortina/genética , Tetraodontiformes/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Peces/genética , Datos de Secuencia Molecular , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...