Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.195
Filtrar
1.
Sci Adv ; 10(21): eadl2882, 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38781346

RESUMO

Neuromorphic sensors, designed to emulate natural sensory systems, hold the promise of revolutionizing data extraction by facilitating rapid and energy-efficient analysis of extensive datasets. However, a challenge lies in accurately distinguishing specific analytes within mixtures of chemically similar compounds using existing neuromorphic chemical sensors. In this study, we present an artificial olfactory system (AOS), developed through the integration of human olfactory receptors (hORs) and artificial synapses. This AOS is engineered by interfacing an hOR-functionalized extended gate with an organic synaptic device. The AOS generates distinct patterns for odorants and mixtures thereof, at the molecular chain length level, attributed to specific hOR-odorant binding affinities. This approach enables precise pattern recognition via training and inference simulations. These findings establish a foundation for the development of high-performance sensor platforms and artificial sensory systems, which are ideal for applications in wearable and implantable devices.


Assuntos
Odorantes , Receptores Odorantes , Humanos , Receptores Odorantes/metabolismo , Odorantes/análise , Olfato/fisiologia , Sinapses/metabolismo , Reconhecimento Automatizado de Padrão/métodos , Neurônios Receptores Olfatórios/metabolismo , Neurônios Receptores Olfatórios/fisiologia , Técnicas Biossensoriais/métodos
2.
Nat Commun ; 15(1): 3360, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38637611

RESUMO

The mammalian olfactory system detects and discriminates between millions of odorants to elicit appropriate behavioral responses. While much has been learned about how olfactory sensory neurons detect odorants and signal their presence, how specific innate, unlearned behaviors are initiated in response to ethologically relevant odors remains poorly understood. Here, we show that the 4-transmembrane protein CD20, also known as MS4A1, is expressed in a previously uncharacterized subpopulation of olfactory sensory neurons in the main olfactory epithelium of the murine nasal cavity and functions as a mammalian olfactory receptor that recognizes compounds produced by mouse predators. While wildtype mice avoid these predator odorants, mice genetically deleted of CD20 do not appropriately respond. Together, this work reveals a CD20-mediated odor-sensing mechanism in the mammalian olfactory system that triggers innate behaviors critical for organismal survival.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Camundongos , Aprendizagem/fisiologia , Mamíferos/metabolismo , Odorantes , Mucosa Olfatória/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Olfato/fisiologia , Antígenos CD20/metabolismo
3.
Genesis ; 62(2): e23594, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38590146

RESUMO

During development of the nervous system, neurons connect to one another in a precisely organized manner. Sensory systems provide a good example of this organization, whereby the composition of the outside world is represented in the brain by neuronal maps. Establishing correct patterns of neural circuitry is crucial, as inaccurate map formation can lead to severe disruptions in sensory processing. In rodents, olfactory stimuli modulate a wide variety of behaviors essential for survival. The formation of the olfactory glomerular map is dependent on molecular cues that guide olfactory receptor neuron axons to broad regions of the olfactory bulb and on cell adhesion molecules that promote axonal sorting into specific synaptic units in this structure. Here, we demonstrate that the cell adhesion molecule Amigo1 is expressed in a subpopulation of olfactory receptor neurons, and we investigate its role in the precise targeting of olfactory receptor neuron axons to the olfactory bulb using a genetic loss-of-function approach in mice. While ablation of Amigo1 did not lead to alterations in olfactory sensory neuron axonal targeting, our experiments revealed that the presence of a neomycin resistance selection cassette in the Amigo1 locus can lead to off-target effects that are not due to loss of Amigo1 expression, including unexpected altered gene expression in olfactory receptor neurons and reduced glomerular size in the ventral region of the olfactory bulb. Our results demonstrate that insertion of a neomycin selection cassette into the mouse genome can have specific deleterious effects on the development of the olfactory system and highlight the importance of removing antibiotic resistance cassettes from genetic loss-of-function mouse models when studying olfactory system development.


Assuntos
Neurônios Receptores Olfatórios , Animais , Camundongos , Neurônios Receptores Olfatórios/metabolismo , Mucosa Olfatória , Bulbo Olfatório , Axônios/metabolismo , Expressão Gênica
4.
Genesis ; 62(1): e23586, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38593162

RESUMO

Neural activity influences every aspect of nervous system development. In olfactory systems, sensory neurons expressing the same odorant receptor project their axons to stereotypically positioned glomeruli, forming a spatial map of odorant receptors in the olfactory bulb. As individual odors activate unique combinations of glomeruli, this map forms the basis for encoding olfactory information. The establishment of this stereotypical olfactory map requires coordinated regulation of axon guidance molecules instructed by spontaneous activity. Recent studies show that sensory experiences also modify innervation patterns in the olfactory bulb, especially during a critical period of the olfactory system development. This review examines evidence in the field to suggest potential mechanisms by which various aspects of neural activity regulate axon targeting. We also discuss the precise functions served by neural plasticity during the critical period.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Neurônios Receptores Olfatórios/metabolismo , Bulbo Olfatório/fisiologia , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Axônios/metabolismo , Mamíferos
5.
Genesis ; 62(2): e23593, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38562011

RESUMO

The mammalian sense of smell relies upon a vast array of receptor proteins to detect odorant compounds present in the environment. The proper deployment of these receptor proteins in olfactory sensory neurons is orchestrated by a suite of epigenetic processes that remodel the olfactory genes in differentiating neuronal progenitors. The goal of this review is to elucidate the central role of gene regulatory processes acting in neuronal progenitors of olfactory sensory neurons that lead to a singular expression of an odorant receptor in mature olfactory sensory neurons. We begin by describing the principal features of odorant receptor gene expression in mature olfactory sensory neurons. Next, we delineate our current understanding of how these features emerge from multiple gene regulatory mechanisms acting in neuronal progenitors. Finally, we close by discussing the key gaps in our understanding of how these regulatory mechanisms work and how they interact with each other over the course of differentiation.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Olfato/genética , Regulação da Expressão Gênica , Epigênese Genética , Mamíferos
6.
Sci Rep ; 14(1): 9110, 2024 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-38643298

RESUMO

Critical periods are temporally-restricted, early-life windows when sensory experience remodels synaptic connectivity to optimize environmental input. In the Drosophila juvenile brain, critical period experience drives synapse elimination, which is transiently reversible. Within olfactory sensory neuron (OSN) classes synapsing onto single projection neurons extending to brain learning/memory centers, we find glia mediate experience-dependent pruning of OSN synaptic glomeruli downstream of critical period odorant exposure. We find glial projections infiltrate brain neuropil in response to critical period experience, and use Draper (MEGF10) engulfment receptors to prune synaptic glomeruli. Downstream, we find antagonistic Basket (JNK) and Puckered (DUSP) signaling is required for the experience-dependent translocation of activated Basket into glial nuclei. Dependent on this signaling, we find critical period experience drives expression of the F-actin linking signaling scaffold Cheerio (FLNA), which is absolutely essential for the synaptic glomeruli pruning. We find Cheerio mediates experience-dependent regulation of the glial F-actin cytoskeleton for critical period remodeling. These results define a sequential pathway for experience-dependent brain synaptic glomeruli pruning in a strictly-defined critical period; input experience drives neuropil infiltration of glial projections, Draper/MEGF10 receptors activate a Basket/JNK signaling cascade for transcriptional activation, and Cheerio/FLNA induction regulates the glial actin cytoskeleton to mediate targeted synapse phagocytosis.


Assuntos
Proteínas de Drosophila , Neurônios Receptores Olfatórios , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Transdução de Sinais , Encéfalo/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Neuroglia/metabolismo
7.
J Exp Biol ; 227(9)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38511428

RESUMO

Odorants interact with receptors expressed in specialized olfactory neurons, and neurons of the same class send their axons to distinct glomeruli in the brain. The stereotypic spatial glomerular activity map generates recognition and the behavioral response for the odorant. The valence of an odorant changes with concentration, typically becoming aversive at higher concentrations. Interestingly, in Drosophila larvae, the odorant (E)-2-hexenal is aversive at low concentrations and attractive at higher concentrations. We investigated the molecular and neural basis of this phenomenon, focusing on how activities of different olfactory neurons conveying opposing effects dictate behaviors. We identified the repellant neuron in the larvae as one expressing the olfactory receptor Or7a, whose activation alone at low concentrations of (E)-2-hexenal elicits an avoidance response in an Or7a-dependent manner. We demonstrate that avoidance can be overcome at higher concentrations by activation of additional neurons that are known to be attractive, most notably odorants that are known activators of Or42a and Or85c. These findings suggest that in the larval stage, the attraction-conveying neurons can overcome the aversion-conveying channels for (E)-2-hexenal.


Assuntos
Aldeídos , Larva , Odorantes , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Larva/crescimento & desenvolvimento , Larva/fisiologia , Receptores Odorantes/metabolismo , Odorantes/análise , Neurônios Receptores Olfatórios/metabolismo , Neurônios Receptores Olfatórios/fisiologia , Aldeídos/metabolismo , Aldeídos/farmacologia , Drosophila melanogaster/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Olfato/fisiologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Drosophila/fisiologia , Drosophila/metabolismo
8.
Neuron ; 112(9): 1473-1486.e6, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38447577

RESUMO

Phasic (fast) and tonic (sustained) inhibition of γ-aminobutyric acid (GABA) are fundamental for regulating day-to-day activities, neuronal excitability, and plasticity. However, the mechanisms and physiological functions of glial GABA transductions remain poorly understood. Here, we report that the AMsh glia in Caenorhabditis elegans exhibit both phasic and tonic GABAergic signaling, which distinctively regulate olfactory adaptation and neuronal aging. Through genetic screening, we find that GABA permeates through bestrophin-9/-13/-14 anion channels from AMsh glia, which primarily activate the metabolic GABAB receptor GBB-1 in the neighboring ASH sensory neurons. This tonic action of glial GABA regulates the age-associated changes of ASH neurons and olfactory responses via a conserved signaling pathway, inducing neuroprotection. In addition, the calcium-evoked, vesicular glial GABA release acts upon the ionotropic GABAA receptor LGC-38 in ASH neurons to regulate olfactory adaptation. These findings underscore the fundamental significance of glial GABA in maintaining healthy aging and neuronal stability.


Assuntos
Adaptação Fisiológica , Caenorhabditis elegans , Neuroglia , Ácido gama-Aminobutírico , Animais , Ácido gama-Aminobutírico/metabolismo , Neuroglia/metabolismo , Neuroglia/fisiologia , Adaptação Fisiológica/fisiologia , Olfato/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Transdução de Sinais/fisiologia , Senescência Celular/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Envelhecimento/fisiologia , Envelhecimento/metabolismo , Receptores de GABA-A/metabolismo
9.
Int J Mol Sci ; 25(5)2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38474024

RESUMO

Traumatic brain injury (TBI) is defined as an injury to the brain by external forces which can lead to cellular damage and the disruption of normal central nervous system functions. The recently approved blood-based biomarkers GFAP and UCH-L1 can only detect injuries which are detectable on CT, and are not sensitive enough to diagnose milder injuries or concussion. Exosomes are small microvesicles which are released from the cell as a part of extracellular communication in normal as well as diseased states. The objective of this study was to identify the messenger RNA content of the exosomes released by injured neurons to identify new potential blood-based biomarkers for TBI. Human severe traumatic brain injury samples were used for this study. RNA was isolated from neuronal exosomes and total transcriptomic sequencing was performed. RNA sequencing data from neuronal exosomes isolated from serum showed mRNA transcripts of several neuronal genes. In particular, mRNAs of several olfactory receptor genes were present at elevated concentrations in the neuronal exosomes. Some of these genes were OR10A6, OR14A2, OR6F1, OR1B1, and OR1L1. RNA sequencing data from exosomes isolated from CSF showed a similar elevation of these olfactory receptors. We further validated the expression of these samples in serum samples of mild TBI patients, and a similar up-regulation of these olfactory receptors was observed. The data from these experiments suggest that damage to the neurons in the olfactory neuroepithelium as well as in the brain following a TBI may cause the release of mRNA from these receptors in the exosomes. Hence, olfactory receptors can be further explored as biomarkers for the diagnosis of TBI.


Assuntos
Concussão Encefálica , Lesões Encefálicas Traumáticas , Lesões Encefálicas , Vesículas Extracelulares , Neurônios Receptores Olfatórios , Receptores Odorantes , Humanos , Lesões Encefálicas Traumáticas/metabolismo , Vesículas Extracelulares/metabolismo , Neurônios Receptores Olfatórios/metabolismo , RNA , Biomarcadores , RNA Mensageiro , Perfilação da Expressão Gênica
10.
Dev Neurobiol ; 84(2): 59-73, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38439531

RESUMO

In contrast to other S100 protein members, the function of S100 calcium-binding protein Z (S100Z) remains largely uncharacterized. It is expressed in the olfactory epithelium of fish, and it is closely associated with the vomeronasal organ (VNO) in mammals. In this study, we analyzed the expression pattern of S100Z in the olfactory system of the anuran amphibian Xenopus laevis. Using immunohistochemistry in whole mount and slice preparations of the larval olfactory system, we found exclusive S100Z expression in a subpopulation of olfactory receptor neurons (ORNs) of the main olfactory epithelium (MOE). S100Z expression was not co-localized with TP63 and cytokeratin type II, ruling out basal cell and supporting cell identity. The distribution of S100Z-expressing ORNs was laterally biased, and their average number was significantly increased in the lateral half of the olfactory epithelium. The axons of S100Z-positive neurons projected exclusively into the lateral and intermediate glomerular clusters of the main olfactory bulb (OB). Even after metamorphic restructuring of the olfactory system, S100Z expression was restricted to a neuronal subpopulation of the MOE, which was then located in the newly formed middle cavity. An axonal projection into the ventro-lateral OB persisted also in postmetamorphic frogs. In summary, S100Z is exclusively associated with the main olfactory system in the amphibian Xenopus and not with the VNO as in mammals, despite the presence of a separate accessory olfactory system in both classes.


Assuntos
Neurônios Receptores Olfatórios , Proteínas S100 , Órgão Vomeronasal , Animais , Bulbo Olfatório/metabolismo , Mucosa Olfatória , Neurônios Receptores Olfatórios/metabolismo , Proteínas S100/metabolismo , Órgão Vomeronasal/metabolismo , Xenopus laevis/metabolismo
11.
Genesis ; 62(2): e23587, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38454646

RESUMO

The sense of smell is intricately linked to essential animal behaviors necessary for individual survival and species preservation. During vertebrate evolution, odorant receptors (ORs), responsible for detecting odor molecules, have evolved to adapt to changing environments, transitioning from aquatic to terrestrial habitats and accommodating increasing complex chemical environments. These evolutionary pressures have given rise to the largest gene family in vertebrate genomes. Vertebrate ORs are phylogenetically divided into two major classes; class I and class II. Class I OR genes, initially identified in fish and frog, have persisted across vertebrate species. On the other hand, class II OR genes are unique to terrestrial animals, accounting for ~90% of mammalian OR genes. In mice, each olfactory sensory neuron (OSN) expresses a single functional allele of a single OR gene from either the class I or class II OR repertoire. This one neuron-one receptor rule is established through two sequential steps: specification of OR class and subsequent exclusive OR expression from the corresponding OR class. Consequently, OSNs acquire diverse neuronal identities during the process of OSN differentiation, enabling animals to detect a wide array of odor molecules. This review provides an overview of the OSN differentiation process through which OSN diversity is achieved, primarily using the mouse as a model animal.


Assuntos
Neurônios Receptores Olfatórios , Animais , Camundongos , Neurônios Receptores Olfatórios/metabolismo , Olfato/fisiologia , Odorantes , Células Receptoras Sensoriais , Mamíferos
12.
Trends Neurosci ; 47(3): 167-169, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38378395

RESUMO

In a recent study, Pourmorady and colleagues uncovered a noncoding role for olfactory receptor (OR)-coding mRNA in mediating nuclear architecture and singular OR choice. The OR mRNAs reinforce the prevailing enhancer hub and inhibit other competitors, facilitating transition from polygenic to singular OR expression.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Humanos , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neurônios Receptores Olfatórios/metabolismo
13.
Cell Tissue Res ; 396(1): 95-102, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38347202

RESUMO

The odor space of aquatic organisms is by necessity quite different from that of air-breathing animals. The recognized odor classes in teleost fish include amino acids, bile acids, reproductive hormones, nucleotides, and a limited number of polyamines. Conversely, a significant portion of the fish olfactory receptor repertoire is composed of trace amine-associated receptors, generally assumed to be responsible for detecting amines. Zebrafish possess over one hundred of these receptors, but the responses of olfactory sensory neurons to amines have not been known so far. Here we examined odor responses of zebrafish olfactory epithelial explants at the cellular level, employing calcium imaging. We report that amines elicit strong responses in olfactory sensory neurons, with a time course characteristically different from that of ATP-responsive (basal) cells. A quantitative analysis of the laminar height distribution shows amine-responsive cells undistinguishable from ciliated neurons positive for olfactory marker protein. This distribution is significantly different from those measured for microvillous neurons positive for transient receptor potential channel 2 and basal cells positive for proliferating cell nuclear antigen. Our results suggest amines as an important odor class for teleost fish.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Peixe-Zebra/metabolismo , Cálcio/metabolismo , Aminas/metabolismo , Odorantes , Mucosa Olfatória/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo
14.
Insect Biochem Mol Biol ; 167: 104069, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38220070

RESUMO

The host-seeking behavior of mosquitoes have long been established to be primarily odor-mediated. In this process, olfactory receptors (Ors) play a critical role. 1-Octen-3-ol is a common volatile compound that is attractive to hematophagous arthropods such as mosquitos. The olfactory receptor 8 (AaOr8) on the tip of the stylet and maxillary palp of Aedes aegypti is tuned to 1-octen-3-ol, which is required for mosquitoes to quickly find blood vessels from a vertebrate host. However, little is known about the interaction of AaOr8 with 1-octen-3-ol which was studied in vivo and in silico in this study. The molecular binding poses and energies between ligands and the receptor were investigated. Three mutants of AaOr8 were cloned and compared with in vivo calcium imaging utilizing heterologous expression systems. As a result, our findings imply that a genetic disruption including targeted modification of Ors genes may be used to reduce mosquito bites.


Assuntos
Aedes , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Aedes/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Octanóis/química
15.
Proc Biol Sci ; 291(2015): 20232578, 2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38228178

RESUMO

In the silkmoth Bombyx mori, the role of male sensilla trichodea in pheromone detection is well established. Here we study the corresponding female sensilla, which contain two olfactory sensory neurons (OSNs) and come in two lengths, each representing a single physiological type. Only OSNs in medium trichoids respond to the scent of mulberry, the silkworm's exclusive host plant, and are more sensitive in mated females, suggesting a role in oviposition. In long trichoids, one OSN is tuned to (+)-linalool and the other to benzaldehyde and isovaleric acid, both odours emitted by silkworm faeces. While the significance of (+)-linalool detection remains unclear, isovaleric acid repels mated females and may therefore play a role in avoiding crowded oviposition sites. When we examined the underlying molecular components of neurons in female trichoids, we found non-canonical co-expression of Ir8a, the co-receptor for acid responses, and ORco, the co-receptor of odorant receptors, in long trichoids, and the unexpected expression of a specific odorant receptor in both trichoid sensillum types. In addition to elucidating the function of female trichoids, our results suggest that some accepted organizational principles of the insect olfactory system may not apply to the predominant sensilla on the antenna of female B. mori.


Assuntos
Monoterpenos Acíclicos , Bombyx , Hemiterpenos , Neurônios Receptores Olfatórios , Ácidos Pentanoicos , Receptores Odorantes , Animais , Feminino , Bombyx/metabolismo , Sensilas/fisiologia , Olfato , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo , Feromônios/metabolismo
16.
Insect Biochem Mol Biol ; 164: 104046, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38043913

RESUMO

In insect antenna, following the activation of olfactory sensory neurons, odorant molecules are inactivated by enzymes in the sensillum lymph. How the inactivation products are cleared from the sensillum lymph is presently unknown. Here we studied the role of support cells (SCs) and the so-called sensory neuron membrane protein 2 (SNMP2), a member of the CD36 family of lipid transporters abundantly expressed in SCs, in sensillum lymph clearance processes in the moths Heliothis virescens and Bombyx mori. In these species, the sex pheromone components are inactivated to long-chain fatty acids. To approach a role of SNMP2 in the removal of such inactivation products, we analyzed the uptake of a fluorescent long-chain fatty acid analog into a newly generated HvirSNMP2-expressing cell line. We found an increased uptake of the analog into SNMP2-cells compared to control cells, which could be blocked by the CD36 protein inhibitor, SSO. Furthermore, analyses of sensilla from antenna treated with the fatty acid analog indicated that SNMP2-expressing SCs are able to take up fatty acids from the sensillum lymph. In addition, sensilla from SSO-pretreated antenna of B. mori showed reduced removal of the fluorescent analog from the sensillum lymph. Finally, we revealed that SSO pretreatment of male silkmoth antenna significantly prolonged the duration of the female pheromone-induced wing-fluttering behavior, possibly as a result of impaired lymph clearance processes. Together our findings in H. virescens and B. mori support a pivotal role of olfactory SCs in sensillum lymph maintenance processes and suggest an integral role of SNMP2 in the removal of lipophilic "waste products" such as fatty acids resulting from sex pheromone inactivation.


Assuntos
Bombyx , Mariposas , Neurônios Receptores Olfatórios , Atrativos Sexuais , Masculino , Feminino , Animais , Mariposas/metabolismo , Sensilas/metabolismo , Feromônios/metabolismo , Atrativos Sexuais/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Bombyx/metabolismo , Células Receptoras Sensoriais/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Ácidos Graxos/metabolismo
17.
Nucleic Acids Res ; 52(D1): D1370-D1379, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37870437

RESUMO

Mammalian sense of smell is triggered by interaction between odorant molecules and a class of proteins, called olfactory receptors (ORs). These receptors, expressed at the surface of olfactory sensory neurons, encode myriad of distinct odors via a sophisticated activation pattern. However, determining the molecular recognition spectrum of ORs remains a major challenge. The Molecule to Olfactory Receptor database (M2OR, https://m2or.chemsensim.fr/) provides curated data that allows an easy exploration of the current state of the research on OR-molecule interaction. We have gathered a database of 75,050 bioassay experiments for 51 395 distinct OR-molecule pairs. Drawn from published literature and public databases, M2OR contains information about OR responses to molecules and their mixtures, receptor sequences and experimental details. Users can obtain information on the activity of a chosen molecule or a group of molecules, or search for agonists for a specific OR or a group of ORs. Advanced search allows for fine-grained queries using various metadata such as species or experimental assay system, and the database can be queried by multiple inputs via a batch search. Finally, for a given search query, users can access and download a curated aggregation of the experimental data into a binarized combinatorial code of olfaction.


Assuntos
Bases de Dados de Proteínas , Receptores Odorantes , Animais , Mamíferos/metabolismo , Odorantes , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Olfato
18.
Insect Sci ; 31(2): 469-488, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38105530

RESUMO

The olfactory system of adult lepidopterans is among the best described neuronal circuits. However, comparatively little is known about the organization of the olfactory system in the larval stage of these insects. Here, we explore the expression of olfactory receptors and the organization of olfactory sensory neurons in caterpillars of Pieris brassicae, a significant pest species in Europe and a well-studied species for its chemical ecology. To describe the larval olfactory system in this species, we first analyzed the head transcriptome of third-instar larvae (L3) and identified 16 odorant receptors (ORs) including the OR coreceptor (Orco), 13 ionotropic receptors (IRs), and 8 gustatory receptors (GRs). We then quantified the expression of these 16 ORs in different life stages, using qPCR, and found that the majority of ORs had significantly higher expression in the L4 stage than in the L3 and L5 stages, indicating that the larval olfactory system is not static throughout caterpillar development. Using an Orco-specific antibody, we identified all olfactory receptor neurons (ORNs) expressing the Orco protein in L3, L4, and L5 caterpillars and found a total of 34 Orco-positive ORNs, distributed among three sensilla on the antenna. The number of Orco-positive ORNs did not differ among the three larval instars. Finally, we used retrograde axon tracing of the antennal nerve and identified a mean of 15 glomeruli in the larval antennal center (LAC), suggesting that the caterpillar olfactory system follows a similar design as the adult olfactory system, although with a lower numerical redundancy. Taken together, our results provide a detailed analysis of the larval olfactory neurons in P. brassicae, highlighting both the differences as well as the commonalities with the adult olfactory system. These findings contribute to a better understanding of the development of the olfactory system in insects and its life-stage-specific adaptations.


Assuntos
Lepidópteros , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Neurônios Receptores Olfatórios/metabolismo , Insetos/fisiologia , Larva/metabolismo , Sensilas/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo
19.
Nature ; 625(7993): 181-188, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38123679

RESUMO

Olfactory receptor (OR) choice provides an extreme example of allelic competition for transcriptional dominance, where every olfactory neuron stably transcribes one of approximately 2,000 or more OR alleles1,2. OR gene choice is mediated by a multichromosomal enhancer hub that activates transcription at a single OR3,4, followed by OR-translation-dependent feedback that stabilizes this choice5,6. Here, using single-cell genomics, we show formation of many competing hubs with variable enhancer composition, only one of which retains euchromatic features and transcriptional competence. Furthermore, we provide evidence that OR transcription recruits enhancers and reinforces enhancer hub activity locally, whereas OR RNA inhibits transcription of competing ORs over distance, promoting transition to transcriptional singularity. Whereas OR transcription is sufficient to break the symmetry between equipotent enhancer hubs, OR translation stabilizes transcription at the prevailing hub, indicating that there may be sequential non-coding and coding mechanisms that are implemented by OR alleles for transcriptional prevalence. We propose that coding OR mRNAs possess non-coding functions that influence nuclear architecture, enhance their own transcription and inhibit transcription from their competitors, with generalizable implications for probabilistic cell fate decisions.


Assuntos
Neurônios Receptores Olfatórios , RNA , Receptores Odorantes , Alelos , Linhagem da Célula , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , RNA/genética , Transcrição Gênica , Genômica , Análise de Célula Única
20.
Curr Biol ; 33(24): 5427-5438.e5, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38070506

RESUMO

Olfactory coding, from insects to humans, is canonically considered to involve considerable across-fiber coding already at the peripheral level, thereby allowing recognition of vast numbers of odor compounds. We show that the migratory locust has evolved an alternative strategy built on highly specific odorant receptors feeding into a complex primary processing center in the brain. By collecting odors from food and different life stages of the locust, we identified 205 ecologically relevant odorants, which we used to deorphanize 48 locust olfactory receptors via ectopic expression in Drosophila. Contrary to the often broadly tuned olfactory receptors of other insects, almost all locust receptors were found to be narrowly tuned to one or very few ligands. Knocking out a single receptor using CRISPR abolished physiological and behavioral responses to the corresponding ligand. We conclude that the locust olfactory system, with most olfactory receptors being narrowly tuned, differs from the so-far described olfactory systems.


Assuntos
Gafanhotos , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Humanos , Odorantes , Olfato/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/metabolismo , Insetos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA