Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38746208

RESUMO

The mammalian olfactory neuronal lineage is regenerative, and accordingly, maintains a population of pluripotent cells that replenish olfactory sensory neurons and other olfactory cell types during the life of the animal. Moreover, in response to acute injury, the early transit amplifying cells along the olfactory sensory neuronal lineage are able to de-differentiate to shift resources in support of tissue restoration. In order to further explore plasticity of various cellular stages along the olfactory sensory neuronal lineage, we challenged the epigenetic stability of two olfactory placode-derived cell lines that model immature olfactory sensory neuronal stages. We found that perturbation of the Ehmt2 chromatin modifier transformed the growth properties, morphology, and gene expression profiles towards states with several stem cell characteristics. This transformation was dependent on continued expression of the large T-antigen, and was enhanced by Sox2 over-expression. These findings may provide momentum for exploring inherent cellular plasticity within early cell types of the olfactory lineage, as well as potentially add to our knowledge of cellular reprogramming. SUMMARY STATEMENT: Discovering how epigenetic modifications influence olfactory neuronal lineage plasticity offers insights into regenerative potential and cellular reprogramming.

2.
Mol Cell Neurosci ; 117: 103681, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34742908

RESUMO

The mammalian olfactory system consists of sensory neurons with specialized odorant-binding capability accomplished by mutually exclusive odorant receptor (OR) expression. Mutually exclusive OR expression is a complex multi-step process regulated by a number of cis and trans factors, including pan-silencing of all OR genes preceding the robust and stable expression of the one OR selected in each sensory neuron. We transfected two olfactory-placode-derived cell lines modeling immature odorant sensory neurons, as well as the GD25 fibroblast cell line, with episomes containing CMV-driven GFP and TK-driven hygromycin reporter genes. We inserted various coding sequences, along with an IRES, immediately upstream of the GFP gene to produce bicistronic mRNAs driven from the local CMV promoter. We found that the presence of several OR coding sequences resulted in significantly diminished episomal expression of GFP in all three cell lines. These findings suggest that OR coding sequences have intrinsic self-silencing capability that might facilitate mutually exclusive OR expression in olfactory sensory neurons by making it less likely that multiple ORs acquire an above-threshold level of expression at once.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Linhagem Celular , Plasmídeos , Receptores Odorantes/genética , Células Receptoras Sensoriais
3.
Chem Senses ; 44(9): 705-720, 2019 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-31529021

RESUMO

Olfactory neuronal function depends on the expression and proper regulation of odorant receptor (OR) genes. Previous studies have identified 54 putative intergenic enhancers within or flanking 40 mouse OR clusters. At least 2 of these putative enhancers have been shown to regulate the expression of a small subset of proximal OR genes. In recognition of the large size of the mouse OR gene family (~1400 OR genes distributed across multiple chromosomal loci), it is likely that there remain many additional not-as-yet discovered OR enhancers. We utilized 23 of the previously identified enhancers as a training set (TS) and designed an algorithm that combines a broad range of epigenetic criteria (histone-3-lysine-4 monomethylation, histone-3-lysine-79 trimethylation, histone-3-lysine-27 acetylation, and DNase hypersensitivity) and genetic criteria (cross-species sequence conservation and transcription-factor binding site enrichment) to more broadly search OR gene clusters for additional candidates. We identified 181 new candidate enhancers located at 58 (of 68) mouse OR loci, including 25 new candidates identified by stringent search criteria whose signal strengths are not significantly different from the 23 previously characterized OR enhancers used as the TS. Additionally, we compared OR enhancer versus generic enhancer features in order to evaluate likelihoods that new enhancer candidates specifically function in OR regulation. We found that features distinguishing OR-specific function are significantly more evident for enhancer candidates located within OR clusters as compared with those in flanking regions.


Assuntos
Biologia Computacional/métodos , Elementos Facilitadores Genéticos/genética , Receptores Odorantes/genética , Algoritmos , Animais , Loci Gênicos , Camundongos , Família Multigênica , Receptores Odorantes/metabolismo
4.
J Neurosci ; 39(5): 814-832, 2019 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-30530861

RESUMO

Neurons in the murine olfactory epithelium (OE) differ by the olfactory receptor they express as well as other molecular phenotypes that are regionally restricted. These patterns can be precisely regenerated following epithelial injury, suggesting that spatial cues within the tissue can direct neuronal diversification. Nonetheless, the permanency and mechanism of this spatial patterning remain subject to debate. Via transplantation of stem and progenitor cells from dorsal OE into ventral OE, we demonstrate that, in mice of both sexes, nonautonomous spatial cues can direct the spatially circumscribed differentiation of olfactory sensory neurons. The vast majority of dorsal transplant-derived neurons express the ventral marker OCAM (NCAM2) and lose expression of NQO1 to match their new location. Single-cell analysis also demonstrates that OSNs adopt a fate defined by their new position following progenitor cell transplant, such that a ventral olfactory receptor is expressed after stem and progenitor cell engraftment. Thus, spatially constrained differentiation of olfactory sensory neurons is plastic, and any bias toward an epigenetic memory of place can be overcome.SIGNIFICANCE STATEMENT Spatially restricted differentiation of olfactory sensory neurons is both key to normal olfactory function and a challenging example of biological specificity. That the stem cells of the olfactory epithelium reproduce the organization of the olfactory periphery to a very close approximation during lesion-induced regeneration begs the question of whether stem cell-autonomous genomic architecture or environmental cues are responsible. The plasticity demonstrated after transfer to a novel location suggests that cues external to the transplanted stem and progenitor cells confer neuronal identity. Thus, a necessary prerequisite is satisfied for using engraftment of olfactory stem and progenitor cells as a cellular therapeutic intervention to reinvigorate neurogenesis whose exhaustion contributes to the waning of olfaction with age.


Assuntos
Mucosa Olfatória/citologia , Neurônios Receptores Olfatórios/fisiologia , Animais , Diferenciação Celular/fisiologia , Sinais (Psicologia) , Epigênese Genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Moléculas de Adesão de Célula Nervosa/biossíntese , Moléculas de Adesão de Célula Nervosa/genética , Células-Tronco Neurais , Neurogênese/fisiologia , Plasticidade Neuronal , Transplante de Células-Tronco
5.
PLoS One ; 13(9): e0204604, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30256852

RESUMO

We previously characterized a clonal olfactory placode-derived cell line (OP6) as a model system for studying odorant receptor (OR) choice, where individual OP6 cells, similar to olfactory sensory neurons in vivo, transcribe one allele ("monoallelic") of one OR gene ("monogenic"). The OP6 cell line provides a unique opportunity to investigate intrinsic properties of OR regulation that cannot easily be investigated in vivo. First, whereas OR-expressing cells in vivo are post-mitotic, OP6 cells are immortalized, raising interesting questions about the stability of epigenetic states associated with OR selection/silencing as OP6 cells progress through the cell cycle. Second, OP6 cells have been isolated away from extrinsic developmental cues, and therefore, any long-term OR selection biases are likely to arise from intrinsic epigenetic states that persist in the absence of developmental context. In this study, we investigated OR re-selection frequency and selection biases within clonal OP6 cell populations. We found no evidence of OR stability through the cell cycle: our results were most consistent with OR re-selection events transpiring at least once per cell division, suggesting that chromatin states associated with OR selection in this system might not be maintained in the subsequent generation. In contrast, we found strong evidence for OR selection biases maintained over prolonged culturing across a diverse set of OP6 cell lineages, suggesting the persistence of intrinsic epigenetic states that advantage some OR loci over others. Together, our data suggest that in the absence of instructive cues, intrinsic epigenetic states influencing OR eligibility, but not those determining OR choice, might persist through the cell cycle.


Assuntos
Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Animais , Ciclo Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Epigênese Genética , Expressão Gênica , Perfilação da Expressão Gênica , Hibridização in Situ Fluorescente , Camundongos , Modelos Genéticos , Neurônios Receptores Olfatórios/citologia , Reação em Cadeia da Polimerase , RNA/genética , Processos Estocásticos
6.
Mol Cell Neurosci ; 82: 1-11, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28414096

RESUMO

Function of the mammalian olfactory system depends on specialized olfactory sensory neurons (OSNs) that each express only one allele ("monoallelic") of one odorant receptor (OR) gene ("monogenic"). The lysine-specific demethylase-1 (LSD1) protein removes activating H3K4 or silencing H3K9 methylation marks in a variety of developmental contexts, and is thought to be important for proper OR regulation. Most of the focus in the field has been on a potential "activating" function for LSD1; e.g., in the demethylation of H3K9 associated with the expressed OR allele. Here we show that depletion of LSD1 in an immortalized olfactory-placode-derived cell line (OP6) results in multigenic and multiallelic OR transcription per cell, while not seemingly disrupting the ability of these cells to activate new OR genes during clonal expansion. These results are consistent with LSD1 having a role in silencing additional OR alleles, as opposed to being required for the activation of OR alleles, within the OP6 cellular context.


Assuntos
Histona Desmetilases/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Animais , Linhagem Celular , Expressão Gênica/genética , Histona Desmetilases/deficiência , Histonas/metabolismo , Lisina/genética , Lisina/metabolismo , Metilação , Camundongos , Olfato/genética
7.
Mol Cell Neurosci ; 74: 58-70, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26947098

RESUMO

Mammalian olfaction depends on the development of specialized olfactory sensory neurons (OSNs) that each express one odorant receptor (OR) protein from a large family of OR genes encoded in the genome. The lysine-specific demethylase-1 (LSD1) protein removes activating H3K4 or silencing H3K9 methylation marks at gene promoters and is required for proper OR regulation. We show that LSD1 protein exhibits variable organization within nuclei of developing OSNs, and tends to consolidate into a single dominant compartment at the edges of chromocenters within nuclei of early post-mitotic cells of the mouse olfactory epithelium (MOE). Using an immortalized cell line derived from developing olfactory placode, we show that consolidation of LSD1 appears to be cell-cycle regulated, with a peak occurrence in early G1. LSD1 co-compartmentalizes with CoREST, a protein known to collaborate with LSD1 to carry out a variety of chromatin-modifying functions. We show that LSD1 compartments co-localize with 1-3 OR loci at the exclusion of most OR genes, and commonly associate with Lhx2, a transcription factor involved in OR regulation. Together, our data suggests that LSD1 is sequestered into a distinct nuclear space that might restrict a histone-modifying function to a narrow developmental time window and/or range of OR gene targets.


Assuntos
Cromatina/metabolismo , Fase G1 , Histona Desmetilases/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Animais , Linhagem Celular , Linhagem da Célula , Núcleo Celular/metabolismo , Proteínas Correpressoras , Histona Desmetilases/genética , Histonas/metabolismo , Camundongos , Mitose , Proteínas do Tecido Nervoso/metabolismo , Neurônios Receptores Olfatórios/citologia , Ligação Proteica , Receptores Odorantes/genética , Proteínas Repressoras/metabolismo
8.
Nucleus ; 5(4): 318-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25482121

RESUMO

Mouse olfaction depends on specialized olfactory sensory neurons (OSNs) that each express only one olfactory receptor protein from among a family of >1000 olfactory receptor (OR) genes encoded in the genome. To investigate epigenetic mechanisms underlying monogenic OR expression, we characterized the nuclear organization of OR loci in an olfactory placode-derived cell line (OP6) derived from a pre-neuronal cell along the OSN lineage. OR loci are significantly enriched within nuclear chromocenters in these cells as compared with control loci tested. However, we observe variability in chromocenter occupancy among different OR loci and from cell-to-cell, suggesting that these associations are transient or context dependent. The lamin B receptor (LBR), whose downregulation is necessary for aggregation of chromocenters and OR genes in mature OSNs, exhibits an unusual non-peripheral expression pattern in OP6 nuclei; upon further OP6 cell differentiation, LBR expression is lost and chromocenters begin to aggregate. However, neither undifferentiated nor differentiated OP6 cells sequester OR genes within the chromocenters, despite the establishment of monogenic OR expression in these cells. These results indicate that sequestration of competing OR loci is not a requirement for monogenic OR expression in OP6 cells, and could indicate that the initial establishment of monogenic OR expression during OSN differentiation in vivo occurs prior to recruitment of OR genes into chromocenters.


Assuntos
Receptores Odorantes/metabolismo , Animais , Linhagem Celular , Histonas/metabolismo , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptor de Lamina B
9.
Genome Res ; 21(8): 1249-59, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21705439

RESUMO

The olfactory receptor (OR) genes are the largest mammalian gene family and are expressed in a monogenic and monoallelic fashion in olfactory neurons. Using a high-throughput approach, we mapped the transcription start sites of 1085 of the 1400 murine OR genes and performed computational analysis that revealed potential transcription factor binding sites shared by the majority of these promoters. Our analysis produced a hierarchical model for OR promoter recognition in which unusually high AT content, a unique epigenetic signature, and a stereotypically positioned O/E site distinguish OR promoters from the rest of the murine promoters. Our computations revealed an intriguing correlation between promoter AT content and evolutionary plasticity, as the most AT-rich promoters regulate rapidly evolving gene families. Within the AT-rich promoter category the position of the TATA-box does not correlate with the transcription start site. Instead, a spike in GC composition might define the exact location of the TSS, introducing the concept of "genomic contrast" in transcriptional regulation. Finally, our experiments show that genomic neighborhood rather than promoter sequence correlates with the probability of different OR genes to be expressed in the same olfactory cell.


Assuntos
Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Receptores Odorantes/genética , Animais , Sítios de Ligação , Camundongos , Camundongos Endogâmicos C57BL , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
BMC Genomics ; 10: 74, 2009 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-19203383

RESUMO

BACKGROUND: The mammalian vomeronasal organ (VNO) expresses two G-protein coupled receptor gene families that mediate pheromone responses, the V1R and V2R receptor genes. In rodents, there are ~150 V1R genes comprising 12 subfamilies organized in gene clusters at multiple chromosomal locations. Previously, we showed that several of these subfamilies had been extensively modulated by gene duplications, deletions, and gene conversions around the time of the evolutionary split of the mouse and rat lineages, consistent with the hypothesis that V1R repertoires might be involved in reinforcing speciation events. Here, we generated genome sequence for one large cluster containing two V1R subfamilies in Mus spretus, a closely related and sympatric species to Mus musculus, and investigated evolutionary change in these repertoires along the two mouse lineages. RESULTS: We describe a comparison of spretus and musculus with respect to genome organization and synteny, as well as V1R gene content and phylogeny, with reference to previous observations made between mouse and rat. Unlike the mouse-rat comparisons, synteny seems to be largely conserved between the two mouse species. Disruption of local synteny is generally associated with differences in repeat content, although these differences appear to arise more from deletion than new integrations. Even though unambiguous V1R orthology is evident, we observe dynamic modulation of the functional repertoires, with two of seven V1Rb and one of eleven V1Ra genes lost in spretus, two V1Ra genes becoming pseudogenes in musculus, two additional orthologous pairs apparently subject to strong adaptive selection, and another divergent orthologous pair that apparently was subjected to gene conversion. CONCLUSION: Therefore, eight of the 18 (~44%) presumptive V1Ra/V1Rb genes in the musculus-spretus ancestor appear to have undergone functional modulation since these two species diverged. As compared to the rat-mouse split, where modulation is evident by independent expansions of these two V1R subfamilies, divergence between musculus and spretus has arisen more by mutations within coding sequences. These results support the hypothesis that adaptive changes in functional V1R repertoires contribute to the delineation of very closely related species.


Assuntos
Fatores Quimiotáticos/genética , Evolução Molecular , Camundongos/genética , Receptores de Feromônios/genética , Animais , Mapeamento Cromossômico , Cromossomos Artificiais Bacterianos , Cromossomos de Mamíferos , Especiação Genética , Genoma , Elementos Nucleotídeos Longos e Dispersos , Família Multigênica , Seleção Genética , Análise de Sequência de DNA , Especificidade da Espécie , Sintenia , Órgão Vomeronasal/fisiologia
11.
J Mol Evol ; 68(2): 145-59, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19153790

RESUMO

The mammalian olfactory system utilizes three large receptor families: the olfactory receptors (ORs) of the main nose and the vomeronasal type-1 and type-2 receptor genes (V1Rs and V2Rs) of the vomeronasal organ. We find that these loci are among the most long interspersed nuclear element (LINE)-dense regions of mammalian genomes. We investigate two evolutionary models to account for this cohabitation. First, we investigate an adaptive selection model, in which LINEs have contributed to expansions of mouse V1R repertoires. We find that even evolutionarily stable V1R loci are exceptionally LINE-rich compared to other genome loci, including loci containing other large gene clusters. Also, a more detailed analysis of specific V1R duplications does not reveal LINE patterns predicted by common LINE-mediated duplication mechanisms. Next, we investigate neutral models, in which LINEs were tolerated by, but not advantageous for, surrounding V1R genes. We find that V1R loci are exceptionally LINE-rich compared to other regions of similar AT base composition, and that duplicated V1R gene blocks are generally depleted of LINE elements, suggesting that these loci did not become densely populated with LINEs simply as a consequence of targeted integration or passive multiplication along with the genes. Finally, we show that individual LINE repeats of a given age at V1R, V2R, and OR loci exhibit a significantly longer average length than at other autosomal loci, suggesting a reduced tendency for these LINEs to be disrupted. We speculate that LINEs at V1R, V2R, and OR loci might be selectively retained because they contribute to allelic regulation of these three gene families.


Assuntos
Evolução Molecular , Elementos Nucleotídeos Longos e Dispersos/genética , Mamíferos/genética , Modelos Genéticos , Receptores Odorantes/genética , Receptores de Feromônios/genética , Algoritmos , Animais , Bovinos , Cromossomos de Mamíferos/genética , Análise por Conglomerados , Cães , Mecanismo Genético de Compensação de Dose , Duplicação Gênica , Camundongos , Gambás , Ratos , Estatísticas não Paramétricas , Cromossomo X/genética
12.
J Neurochem ; 108(2): 486-97, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19012738

RESUMO

We have investigated two clonal mouse olfactory placode (OP) cell lines as a model system for studying endogenous odorant receptor (OR) regulation. Both lines can be differentiated into bipolar neurons with transcriptional profiles consistent with mature sensory neurons. We show that single cells exhibit monogenic OR expression like sensory neurons in vivo. Monogenic OR expression is established in undifferentiated cells and persists through differentiation, but OR gene choice is not a clonal property of either cell line. Interestingly, OR RNA shifts from predominantly nuclear to cytoplasma during differentiation of both cell lines. Finally, our data indicate that a restricted subset of OR genes and OR clusters are over-represented in cell populations, suggesting either a pre-existing intrinsic bias in OP founder cells or extrinsic influences arising from culture conditions.


Assuntos
Diferenciação Celular/fisiologia , Núcleo Celular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios Receptores Olfatórios/citologia , RNA Mensageiro/metabolismo , Receptores Odorantes/genética , Actinas/genética , Actinas/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Embrião não Mamífero , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Proteína de Marcador Olfatório/genética , Proteína de Marcador Olfatório/metabolismo , Mucosa Olfatória/embriologia , Mucosa Olfatória/metabolismo , Receptores Odorantes/classificação
13.
BMC Neurosci ; 8 Suppl 3: S2, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17903278

RESUMO

The olfactory system meets niche- and species-specific demands by an accelerated evolution of its odorant receptor repertoires. In this review, we describe evolutionary processes that have shaped olfactory and vomeronasal receptor gene families in vertebrate genomes. We emphasize three important periods in the evolution of the olfactory system evident by comparative genomics: the adaptation to land in amphibian ancestors, the decline of olfaction in primates, and the delineation of putative pheromone receptors concurrent with rodent speciation. The rapid evolution of odorant receptor genes, the sheer size of the repertoire, as well as their wide distribution in the genome, presents a developmental challenge: how are these ever-changing odorant receptor repertoires coordinated within the olfactory system? A central organizing principle in olfaction is the specialization of sensory neurons resulting from each sensory neuron expressing only ~one odorant receptor allele. In this review, we also discuss this mutually exclusive expression of odorant receptor genes. We have considered several models to account for co-regulation of odorant receptor repertoires, as well as discussed a new hypothesis that invokes important epigenetic properties of the system.


Assuntos
Evolução Molecular , Mucosa Olfatória/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/biossíntese , Receptores Odorantes/genética , Animais , Humanos , Mucosa Olfatória/fisiologia , Neurônios Receptores Olfatórios/química , Neurônios Receptores Olfatórios/fisiologia , Receptores Odorantes/fisiologia
14.
BMC Genomics ; 8: 253, 2007 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-17651493

RESUMO

BACKGROUND: The mouse vomeronasal organ (VNO) processes chemosensory information, including pheromone signals that influence reproductive behaviors. The sensory neurons of the VNO express two types of chemosensory receptors, V1R and V2R. There are ~165 V1R genes in the mouse genome that have been classified into ~12 divergent subfamilies. Each sensory neuron of the apical compartment of the VNO transcribes only one of the repertoire of V1R genes. A model for mutually exclusive V1R transcription in these cells has been proposed in which each V1R gene might compete stochastically for a single transcriptional complex. This model predicts that the large repertoire of divergent V1R genes in the mouse genome contains common regulatory elements. In this study, we have characterized V1R promoter regions by comparative genomics and by mapping transcription start sites. RESULTS: We find that transcription is initiated from ~1 kb promoter regions that are well conserved within V1R subfamilies. While cross-subfamily homology is not evident by traditional methods, we developed a heuristic motif-searching tool, LogoAlign, and applied this tool to identify motifs shared within the promoters of all V1R genes. Our motif-searching tool exhibits rapid convergence to a relatively small number of non-redundant solutions (97% convergence). We also find that the best motifs contain significantly more information than those identified in controls, and that these motifs are more likely to be found in the immediate vicinity of transcription start sites than elsewhere in gene blocks. The best motifs occur near transcription start sites of ~90% of all V1R genes and across all of the divergent subfamilies. Therefore, these motifs are candidate binding sites for transcription factors involved in V1R co-regulation. CONCLUSION: Our analyses show that V1R subfamilies have broad and well conserved promoter regions from which transcription is initiated. Results from a new motif-finding algorithm, LogoAlign, designed for this context and more generally for searching large, hierarchical datasets, suggest the existence of common information-rich regulatory motifs that are shared across otherwise divergent V1R subfamilies.


Assuntos
Fatores Quimiotáticos/genética , Regiões Promotoras Genéticas/genética , Receptores Odorantes/genética , Órgão Vomeronasal/metabolismo , Algoritmos , Animais , Mapeamento Cromossômico , Cromossomos de Mamíferos/genética , Biologia Computacional/métodos , Sequência Conservada , Bases de Dados Genéticas , Genoma , Camundongos , Família Multigênica , Ratos , Sequências Reguladoras de Ácido Nucleico , Sítio de Iniciação de Transcrição , Transcrição Gênica
15.
Genome Res ; 15(2): 231-40, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15653832

RESUMO

The V1R gene family comprises one of two types of putative pheromone receptors expressed in the mammalian vomeronasal organ (VNO). We searched the most recent mouse, rat, dog, chimpanzee, and human genome sequence assemblies to compile a near-complete repertoire of V1R genes for each species. Dog, human, and chimpanzee have very few intact V1Rs (8, 2, and 0, respectively) compared to more than a hundred intact V1Rs in each of the rat (106) and mouse (165) genomes. We also provide the first description of the diversity of V1R pseudogenes in these species. We identify at least 165 pseudogenes in mouse, 110 in rat, 102 in chimpanzee, 115 in human, and 54 in dog. Primate and dog pseudogenes are distributed among almost all V1R subfamilies seen in rodents, indicating that the common ancestor of these species had a diverse V1R repertoire. We find that V1R genes were subject to strikingly different fates in different species and in different subfamilies. In rodents, some subfamilies remained relatively stable or underwent roughly equivalent expansion in mouse and rat; other subfamilies expanded in one species but not the other. The small number of intact V1Rs in the dog genome is unexpected given the presumption that dogs, like rodents, have a functional VNO, and a complex system of pheromone-based behaviors. We identify an intact transient receptor potential channel 2beta in the dog genome, consistent with a functional VNO in dogs. The diminished V1R repertoire in dogs raises questions about the relative contributions of V1Rs versus other candidate pheromone receptor genes in the establishment of complex pheromone systems in mammals.


Assuntos
Amplificação de Genes/genética , Variação Genética/genética , Pan troglodytes/genética , Receptores Odorantes/genética , Animais , Fatores Quimiotáticos/genética , Mapeamento Cromossômico , Cães , Evolução Molecular , Feminino , Marcadores Genéticos/genética , Genoma , Genoma Humano , Humanos , Camundongos , Pseudogenes/genética , Ratos , Sintenia/genética , Órgão Vomeronasal/química , Órgão Vomeronasal/metabolismo
16.
Genome Res ; 14(4): 603-8, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15060001

RESUMO

The mouse V1R putative pheromone receptor gene family consists of at least 137 intact genes clustered at multiple chromosomal locations in the genome. Species-specific pheromone receptor repertoires may partly explain species-specific social behavior. We conducted a genomic analysis of an orthologous pair of mouse and rat V1R gene clusters to test for species specificity in rodent pheromone systems. Mouse and rat have lineage-specific V1R repertoires in each of three major subfamilies at these loci as a result of postspeciation duplications, gene loss, and gene conversions. The onset of this diversification roughly coincides with a wave of Line1 (L1) retrotranspositions into the two loci. We propose that L1 activity has facilitated postspeciation V1R duplications and gene conversions. In addition, we find extensive homology among putative V1R promoter regions in both species. We propose a regulatory model in which promoter homogenization could ensure that V1R genes are equally competitive for a limiting transcriptional structure to account for mutually exclusive V1R expression in vomeronasal neurons.


Assuntos
Família Multigênica/genética , Receptores de Feromônios/genética , Animais , Mapeamento Cromossômico/métodos , Biologia Computacional/estatística & dados numéricos , Bases de Dados Genéticas , Funções Verossimilhança , Camundongos , Filogenia , Ratos , Especificidade da Espécie , Sintenia/genética
17.
Genome Biol ; 4(11): R71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14611657

RESUMO

BACKGROUND: The olfactory receptor gene family is one of the largest in the mammalian genome. Previous computational analyses have identified approximately 1,500 mouse olfactory receptors, but experimental evidence confirming olfactory function is available for very few olfactory receptors. We therefore screened a mouse olfactory epithelium cDNA library to obtain olfactory receptor expressed sequence tags, providing evidence of olfactory function for many additional olfactory receptors, as well as identifying gene structure and putative promoter regions. RESULTS: We identified more than 1,200 odorant receptor cDNAs representing more than 400 genes. Using real-time PCR to confirm expression level differences suggested by our screen, we find that transcript levels in the olfactory epithelium can differ between olfactory receptors by up to 300-fold. Differences for one gene pair are apparently due to both unequal numbers of expressing cells and unequal transcript levels per expressing cell. At least two-thirds of olfactory receptors exhibit multiple transcriptional variants, with alternative isoforms of both 5' and 3' untranslated regions. Some transcripts (5%) utilize splice sites within the coding region, contrary to the stereotyped olfactory receptor gene structure. Most atypical transcripts encode nonfunctional olfactory receptors, but can occasionally increase receptor diversity. CONCLUSIONS: Our cDNA collection confirms olfactory function of over one-third of the intact mouse olfactory receptors. Most of these genes were previously annotated as olfactory receptors based solely on sequence similarity. Our finding that different olfactory receptors have different expression levels is intriguing given the one-neuron, one-gene expression regime of olfactory receptors. We provide 5' untranslated region sequences and candidate promoter regions for more than 300 olfactory receptors, valuable resources for computational regulatory motif searches and for designing olfactory receptor microarrays and other experimental probes.


Assuntos
Processamento Alternativo , Etiquetas de Sequências Expressas , Mucosa Olfatória/metabolismo , Receptores Odorantes/genética , Animais , DNA/química , DNA/genética , DNA Complementar/química , DNA Complementar/genética , Feminino , Perfilação da Expressão Gênica , Biblioteca Gênica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Filogenia , Pseudogenes/genética , Receptores Odorantes/classificação , Análise de Sequência de DNA
18.
Genome Res ; 12(1): 81-7, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11779833

RESUMO

We have conducted a comparative genomic analysis of several olfactory receptor (OR) genes that lie immediately 5' to the V-alpha gene segments at the mouse and human T-cell receptor (TCR) alpha/delta loci. Five OR genes are identified in the human cluster. The murine cluster has at least six OR genes; the first five are orthologous to the human genes. The sixth mouse gene has arisen since mouse-human divergence by a duplication of a approximately 10-kb block. One pair of OR paralogs found at the mouse and human loci are more similar to each other than to their corresponding orthologs. This paralogous "twinning" appears to be under selection, perhaps to increase sensitivity to particular odorants or to resolve structurally-similar odorants. The promoter regions of the mouse OR genes were identified by RACE-PCR. Orthologs share extensive 5' UTR homology, but we find no significant similarity among paralogs. These findings extend previous observations that suggest that OR genes do not share local significant regulatory homology despite having a common regulatory agenda. We also identified a diverged TCR-alpha gene segment that uses a divergent recombination signal sequence (RSS) to initiate recombination in T-cells from within the OR region. We explored the hypothesis that OR genes may use DNA recombination in expressing neurons, e.g., to recombine ORs into a transcriptionally active locus. We searched the mouse sequence for OR-flanking RSS motifs, but did not find evidence to suggest that these OR genes use TCR-like recombination target sequences.


Assuntos
Genes Codificadores da Cadeia alfa de Receptores de Linfócitos T/genética , Genes Codificadores da Cadeia delta de Receptores de Linfócitos T/genética , Receptores Odorantes/genética , Animais , Marcadores Genéticos/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Análise de Sequência de DNA/métodos
19.
Proc Natl Acad Sci U S A ; 99(1): 291-6, 2002 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-11752409

RESUMO

We have analyzed the organization and sequence of 73 V1R genes encoding putative pheromone receptors to identify regulatory features and characterize the evolutionary history of the V1R family. The 73 V1Rs arose from seven ancestral genes around the time of mouse-rat speciation through large local duplications, and this expansion may contribute to speciation events. Orthologous V1R genes appear to have been lost during primate evolution. Exceptional noncoding homology is observed across four V1R subfamilies at one cluster and thus may be important for locus-specific transcriptional regulation.


Assuntos
Células Quimiorreceptoras/fisiologia , Família Multigênica , Regiões Promotoras Genéticas , Órgão Vomeronasal/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , DNA Complementar/metabolismo , Éxons , Biblioteca Gênica , Genoma , Genoma Humano , Humanos , Camundongos , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Ratos , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...