Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.214
Filtrar
1.
Physiol Rep ; 12(19): e70057, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39358841

RESUMO

The sense of smell is still considered a fuzzy sensation. Softly wafting aromas can stimulate the appetite and trigger memories; however, there are many unexplored aspects of its underlying mechanisms, and not all of these have been elucidated. Although the final sense of smell takes place in the brain, it is greatly affected during the preliminary stage, when odorants are converted into electrical signals. After signal conversion through ion channels in olfactory cilia, action potentials are generated through other types of ion channels located in the cell body. Spike trains through axons transmit this information as digital signals to the brain, however, before odorants are converted into digital electric signals, such as an action potential, modification of the transduction signal has already occurred. This review focuses on the early stages of olfactory signaling. Modification of signal transduction mechanisms and their effect on the human sense of smell through three characteristics (signal amplification, olfactory adaptation, and olfactory masking) produced by olfactory cilia, which is the site of signal transduction are being addressed in this review.


Assuntos
Cílios , Transdução de Sinais , Olfato , Humanos , Olfato/fisiologia , Animais , Cílios/fisiologia , Cílios/metabolismo , Transdução de Sinais/fisiologia , Neurônios Receptores Olfatórios/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Odorantes
2.
Commun Biol ; 7(1): 1150, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39284959

RESUMO

Measuring neuronal activity is important for understanding neuronal function. Ca2+ imaging by genetically encoded calcium indicators (GECIs) is a powerful way to measure neuronal activity. Although it revealed important aspects of neuronal function, measuring the neuronal membrane voltage is important to understand neuronal function as it triggers neuronal activation. Recent progress of genetically encoded voltage indicators (GEVIs) enabled us fast and precise measurements of neuronal membrane voltage. To clarify the relation of the membrane voltage and intracellular Ca2+, we analyzed neuronal activities of olfactory neuron AWA in Caenorhabditis elegans by GCaMP6f (GECI) and paQuasAr3 (GEVI) responding to odorants. We found that the membrane voltage encodes the stimuli change by the timing and the duration by the weak semi-stable depolarization. However, the change of the intracellular Ca2+ encodes the strength of the stimuli. Furthermore, ODR-3, a G-protein alpha subunit, was shown to be important for stabilizing the membrane voltage. These results suggest that the combination of calcium and voltage imaging provides a deeper understanding of the information in neural circuits.


Assuntos
Caenorhabditis elegans , Cálcio , Animais , Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Cálcio/metabolismo , Potenciais da Membrana/fisiologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Neurônios Receptores Olfatórios/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Odorantes/análise
3.
Cell Rep ; 43(9): 114671, 2024 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-39215999

RESUMO

Recent discoveries have revealed remarkable complexity within olfactory sensory neurons (OSNs), including the existence of two OSN populations based on the expression of Cd36. However, the regulatory mechanisms governing this cellular diversity in the same cell type remain elusive. Here, we show the preferential expression of 79 olfactory receptors in Cd36+ OSNs and the anterior projection characteristics of Cd36+ OSNs, indicating the non-randomness of Cd36 expression. The integrated analysis of single-cell RNA sequencing (scRNA-seq) and scATAC-seq reveals that the differences in Cd36+/- OSNs occur at the immature OSN stage, with Mef2a and Hdac9 being important regulators of developmental divergence. We hypothesize that the absence of Hdac9 may affect the activation of Mef2a, leading to the up-regulation of Mef2a target genes, including teashirt zinc finger family member 1 (Tshz1), in the Cd36+ OSN lineage. We validate that Tshz1 directly promotes Cd36 expression through enhancer bindings. Our study unravels the intricate regulatory landscape and principles governing cellular diversity in the olfactory system.


Assuntos
Antígenos CD36 , Neurônios Receptores Olfatórios , Análise de Célula Única , Animais , Neurônios Receptores Olfatórios/metabolismo , Antígenos CD36/metabolismo , Antígenos CD36/genética , Camundongos , Análise de Célula Única/métodos , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Histona Desacetilases/metabolismo , Análise de Sequência de RNA/métodos , Camundongos Endogâmicos C57BL , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , RNA-Seq/métodos , Análise da Expressão Gênica de Célula Única
4.
Cell Tissue Res ; 398(1): 35-62, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39174822

RESUMO

Odor detection in insects is largely mediated by structures on antennae called sensilla, which feature a strongly conserved architecture and repertoire of olfactory sensory neurons (OSNs) and various support cell types. In Drosophila, OSNs are tightly apposed to supporting cells, whose connection with neurons and functional roles in odor detection remain unclear. Coupling mechanisms between these neuronal and non-neuronal cell types have been suggested based on morphological observations, concomitant physiological activity during odor stimulation, and known interactions that occur in other chemosensory systems. For instance, it is not known whether cell-cell coupling via gap junctions between OSNs and neighboring cells exists, or whether hemichannels interconnect cellular and extracellular sensillum compartments. Here, we show that innexins, which form hemichannels and gap junctions in invertebrates, are abundantly expressed in adult drosophilid antennae. By surveying antennal transcriptomes and performing various immunohistochemical stainings in antennal tissues, we discover innexin-specific patterns of expression and localization, with a majority of innexins strongly localizing to glial and non-neuronal cells, likely support and epithelial cells. Finally, by injecting gap junction-permeable dye into a pre-identified sensillum, we observe no dye coupling between neuronal and non-neuronal cells. Together with evidence of non-neuronal innexin localization, we conclude that innexins likely do not conjoin neurons to support cells, but that junctions and hemichannels may instead couple support cells among each other or to their shared sensillum lymph to achieve synchronous activity. We discuss how coupling of sensillum microenvironments or compartments may potentially contribute to facilitate chemosensory functions of odor sensing and sensillum homeostasis.


Assuntos
Antenas de Artrópodes , Conexinas , Proteínas de Drosophila , Junções Comunicantes , Sensilas , Animais , Sensilas/metabolismo , Antenas de Artrópodes/metabolismo , Junções Comunicantes/metabolismo , Proteínas de Drosophila/metabolismo , Conexinas/metabolismo , Drosophila melanogaster/metabolismo , Neurônios Receptores Olfatórios/metabolismo
5.
Cell ; 187(18): 5081-5101.e19, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-38996528

RESUMO

In developing brains, axons exhibit remarkable precision in selecting synaptic partners among many non-partner cells. Evolutionarily conserved teneurins are transmembrane proteins that instruct synaptic partner matching. However, how intracellular signaling pathways execute teneurins' functions is unclear. Here, we use in situ proximity labeling to obtain the intracellular interactome of a teneurin (Ten-m) in the Drosophila brain. Genetic interaction studies using quantitative partner matching assays in both olfactory receptor neurons (ORNs) and projection neurons (PNs) reveal a common pathway: Ten-m binds to and negatively regulates a RhoGAP, thus activating the Rac1 small GTPases to promote synaptic partner matching. Developmental analyses with single-axon resolution identify the cellular mechanism of synaptic partner matching: Ten-m signaling promotes local F-actin levels and stabilizes ORN axon branches that contact partner PN dendrites. Combining spatial proteomics and high-resolution phenotypic analyses, this study advanced our understanding of both cellular and molecular mechanisms of synaptic partner matching.


Assuntos
Axônios , Proteínas de Drosophila , Drosophila melanogaster , Proteínas do Tecido Nervoso , Neurônios Receptores Olfatórios , Transdução de Sinais , Sinapses , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Axônios/metabolismo , Sinapses/metabolismo , Actinas/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Encéfalo/metabolismo , Dendritos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Tenascina , Proteínas rac de Ligação ao GTP
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167347, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-39019092

RESUMO

Intranasal infection is commonly used to establish a SARS-CoV-2 mouse model due to its non-invasive procedures and a minimal effect from the operation itself. However, mice intranasally infected with SARS-CoV-2 have a high mortality rate, which limits the utility of this model for exploring therapeutic strategies and the sequelae of non-fatal COVID-19 cases. To resolve these limitations, an aerosolised viral administration method has been suggested. However, an in-depth pathological analysis comparing the two models is lacking. Here, we show that inhalation and intranasal SARS-CoV-2 (106 PFU) infection models established in K18-hACE2 mice develop unique pathological features in both the respiratory and central nervous systems, which could be directly attributed to the infection method. While the inhalation-infection model exhibited relatively milder pathological parameters, it closely mimicked the prevalent chest CT pattern observed in COVID-19 patients with focal, peripheral lesions and fibrotic scarring in the recuperating lung. We also found the evidence of direct neuron-invasion from the olfactory receptor neurons to the olfactory bulb in the intranasal model and showed the trigeminal nerve as an alternative route of transmission to the brain in inhalation infected mice. Even after viral clearance confirmed at 14 days post-infection, mild lesions were still found in the brain of inhalation-infected mice. These findings suggest that the inhalation-infection model has advantages over the intranasal-infection model in closely mimicking the pathological features of non-fatal symptoms of COVID-19, demonstrating its potential to study the sequelae and possible interventions for long COVID.


Assuntos
COVID-19 , Modelos Animais de Doenças , Pulmão , SARS-CoV-2 , Animais , COVID-19/patologia , COVID-19/virologia , Camundongos , Pulmão/patologia , Pulmão/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Bulbo Olfatório/patologia , Bulbo Olfatório/virologia , Humanos , Administração Intranasal , Feminino , Neurônios Receptores Olfatórios/virologia , Neurônios Receptores Olfatórios/metabolismo
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-39000230

RESUMO

In insect olfaction, sensitization refers to the amplification of a weak olfactory signal when the stimulus is repeated within a specific time window. In the vinegar fly, Drosophila melanogaster, this occurs already at the periphery, at the level of olfactory sensory neurons (OSNs) located in the antenna. In our study, we investigate whether sensitization is a widespread property in a set of seven types of OSNs, as well as the mechanisms involved. First, we characterize and compare the differences in spontaneous activity, response velocity and response dynamics, among the selected OSN types. These express different receptors with distinct tuning properties and behavioral relevance. Second, we show that sensitization is not a general property. Among our selected OSN types, it occurs in those responding to more general food odors, while OSNs involved in very specific detection of highly specific ecological cues like pheromones and warning signals show no sensitization. Moreover, we show that mitochondria play an active role in sensitization by contributing to the increase in intracellular Ca2+ upon weak receptor activation. Thus, by using a combination of single sensillum recordings (SSRs), calcium imaging and pharmacology, we widen the understanding of how the olfactory signal is processed at the periphery.


Assuntos
Drosophila melanogaster , Neurônios Receptores Olfatórios , Olfato , Animais , Neurônios Receptores Olfatórios/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Drosophila melanogaster/fisiologia , Olfato/fisiologia , Odorantes , Cálcio/metabolismo , Plasticidade Neuronal/fisiologia , Receptores Odorantes/metabolismo , Mitocôndrias/metabolismo
8.
Proc Natl Acad Sci U S A ; 121(31): e2402755121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39042673

RESUMO

The precise assembly of a functional nervous system relies on axon guidance cues. Beyond engaging their cognate receptors and initiating signaling cascades that modulate cytoskeletal dynamics, guidance cues also bind components of the extracellular matrix, notably proteoglycans, yet the role and mechanisms of these interactions remain poorly understood. We found that Drosophila secreted semaphorins bind specifically to glycosaminoglycan (GAG) chains of proteoglycans, showing a preference based on the degree of sulfation. Structural analysis of Sema2b unveiled multiple GAG-binding sites positioned outside canonical plexin-binding site, with the highest affinity binding site located at the C-terminal tail, characterized by a lysine-rich helical arrangement that appears to be conserved across secreted semaphorins. In vivo studies revealed a crucial role of the Sema2b C-terminal tail in specifying the trajectory of olfactory receptor neurons. We propose that secreted semaphorins tether to the cell surface through interactions with GAG chains of proteoglycans, facilitating their presentation to cognate receptors on passing axons.


Assuntos
Orientação de Axônios , Proteínas de Drosophila , Proteoglicanas , Semaforinas , Transdução de Sinais , Animais , Semaforinas/metabolismo , Semaforinas/genética , Proteoglicanas/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Axônios/metabolismo , Drosophila melanogaster/metabolismo , Glicosaminoglicanos/metabolismo , Sítios de Ligação , Ligação Proteica , Neurônios Receptores Olfatórios/metabolismo
9.
Genesis ; 62(3): e23610, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38874301

RESUMO

The organization of the olfactory glomerular map involves the convergence of olfactory sensory neurons (OSNs) expressing the same odorant receptor (OR) into glomeruli in the olfactory bulb (OB). A remarkable feature of the olfactory glomerular map formation is that the identity of OR instructs the topography of the bulb, resulting in thousands of discrete glomeruli in mice. Several lines of evidence indicate that ORs control the expression levels of various kinds of transmembrane proteins to form glomeruli at appropriate regions of the OB. In this review, we will discuss how the OR identity is decoded by OSNs into gene expression through intracellular regulatory mechanisms.


Assuntos
Bulbo Olfatório , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Camundongos , Bulbo Olfatório/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo
10.
Genesis ; 62(3): e23611, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38888221

RESUMO

Olfactory sensory neurons (OSNs) are one of a few neuron types that are generated continuously throughout life in mammals. The persistence of olfactory sensory neurogenesis beyond early development has long been thought to function simply to replace neurons that are lost or damaged through exposure to environmental insults. The possibility that olfactory sensory neurogenesis may also serve an adaptive function has received relatively little consideration, largely due to the assumption that the generation of new OSNs is stochastic with respect to OSN subtype, as defined by the single odorant receptor gene that each neural precursor stochastically chooses for expression out of hundreds of possibilities. Accordingly, the relative birthrates of different OSN subtypes are predicted to be constant and impervious to olfactory experience. This assumption has been called into question, however, by evidence that the birthrates of specific OSN subtypes can be selectively altered by manipulating olfactory experience through olfactory deprivation, enrichment, and conditioning paradigms. Moreover, studies of recovery of the OSN population following injury provide further evidence that olfactory sensory neurogenesis may not be strictly stochastic with respect to subtype. Here we review this evidence and consider mechanistic and functional implications of the prospect that specific olfactory experiences can regulate olfactory sensory neurogenesis rates in a subtype-selective manner.


Assuntos
Neurogênese , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Neurônios Receptores Olfatórios/fisiologia , Neurogênese/genética , Olfato/fisiologia , Olfato/genética , Humanos
11.
Front Neural Circuits ; 18: 1406218, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903957

RESUMO

The olfactory epithelium (OE) is directly exposed to environmental agents entering the nasal cavity, leaving OSNs prone to injury and degeneration. The causes of olfactory dysfunction are diverse and include head trauma, neurodegenerative diseases, and aging, but the main causes are chronic rhinosinusitis (CRS) and viral infections. In CRS and viral infections, reduced airflow due to local inflammation, inflammatory cytokine production, release of degranulated proteins from eosinophils, and cell injury lead to decreased olfactory function. It is well known that injury-induced loss of mature OSNs in the adult OE causes massive regeneration of new OSNs within a few months through the proliferation and differentiation of progenitor basal cells that are subsequently incorporated into olfactory neural circuits. Although normal olfactory function returns after injury in most cases, prolonged olfactory impairment and lack of improvement in olfactory function in some cases poses a major clinical problem. Persistent inflammation or severe injury in the OE results in morphological changes in the OE and respiratory epithelium and decreases the number of mature OSNs, resulting in irreversible loss of olfactory function. In this review, we discuss the histological structure and distribution of the human OE, and the pathogenesis of olfactory dysfunction associated with CRS and viral infection.


Assuntos
Mucosa Olfatória , Humanos , Mucosa Olfatória/patologia , Mucosa Olfatória/metabolismo , Transtornos do Olfato/etiologia , Transtornos do Olfato/fisiopatologia , Transtornos do Olfato/patologia , Neurônios Receptores Olfatórios/fisiologia , Neurônios Receptores Olfatórios/metabolismo , Sinusite/patologia , Sinusite/fisiopatologia , Rinite/patologia , Rinite/fisiopatologia , Rinite/metabolismo , Animais
12.
Int J Mol Sci ; 25(11)2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38892263

RESUMO

The cell surface metalloprotease ADAM17 (a disintegrin and metalloprotease 17) and its binding partners iRhom2 and iRhom1 (inactive Rhomboid-like proteins 1 and 2) modulate cell-cell interactions by mediating the release of membrane proteins such as TNFα (Tumor necrosis factor α) and EGFR (Epidermal growth factor receptor) ligands from the cell surface. Most cell types express both iRhoms, though myeloid cells exclusively express iRhom2, and iRhom1 is the main iRhom in the mouse brain. Here, we report that iRhom2 is uniquely expressed in olfactory sensory neurons (OSNs), highly specialized cells expressing one olfactory receptor (OR) from a repertoire of more than a thousand OR genes in mice. iRhom2-/- mice had no evident morphological defects in the olfactory epithelium (OE), yet RNAseq analysis revealed differential expression of a small subset of ORs. Notably, while the majority of ORs remain unaffected in iRhom2-/- OE, OSNs expressing ORs that are enriched in iRhom2-/- OE showed fewer gene expression changes upon odor environmental changes than the majority of OSNs. Moreover, we discovered an inverse correlation between the expression of iRhom2 compared to OSN activity genes and that odor exposure negatively regulates iRhom2 expression. Given that ORs are specialized G-protein coupled receptors (GPCRs) and many GPCRs activate iRhom2/ADAM17, we investigated if ORs could activate iRhom2/ADAM17. Activation of an olfactory receptor that is ectopically expressed in keratinocytes (OR2AT4) by its agonist Sandalore leads to ERK1/2 phosphorylation, likely via an iRhom2/ADAM17-dependent pathway. Taken together, these findings point to a mechanism by which odor stimulation of OSNs activates iRhom2/ADAM17 catalytic activity, resulting in downstream transcriptional changes to the OR repertoire and activity genes, and driving a negative feedback loop to downregulate iRhom2 expression.


Assuntos
Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Receptores Odorantes/metabolismo , Receptores Odorantes/genética , Camundongos , Neurônios Receptores Olfatórios/metabolismo , Olfato/fisiologia , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Camundongos Knockout , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Mucosa Olfatória/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Humanos
13.
Cell ; 187(15): 3973-3991.e24, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-38897195

RESUMO

The representation of odors in the locust antennal lobe with its >2,000 glomeruli has long remained a perplexing puzzle. We employed the CRISPR-Cas9 system to generate transgenic locusts expressing the genetically encoded calcium indicator GCaMP in olfactory sensory neurons. Using two-photon functional imaging, we mapped the spatial activation patterns representing a wide range of ecologically relevant odors across all six developmental stages. Our findings reveal a functionally ring-shaped organization of the antennal lobe composed of specific glomerular clusters. This configuration establishes an odor-specific chemotopic representation by encoding different chemical classes and ecologically distinct odors in the form of glomerular rings. The ring-shaped glomerular arrangement, which we confirm by selective targeting of OR70a-expressing sensory neurons, occurs throughout development, and the odor-coding pattern within the glomerular population is consistent across developmental stages. Mechanistically, this unconventional spatial olfactory code reflects the locust-specific and multiplexed glomerular innervation pattern of the antennal lobe.


Assuntos
Antenas de Artrópodes , Odorantes , Neurônios Receptores Olfatórios , Animais , Neurônios Receptores Olfatórios/metabolismo , Antenas de Artrópodes/fisiologia , Olfato/fisiologia , Gafanhotos/fisiologia , Animais Geneticamente Modificados , Sistemas CRISPR-Cas/genética , Condutos Olfatórios/fisiologia , Receptores Odorantes/metabolismo , Receptores Odorantes/genética , Locusta migratoria/fisiologia , Cálcio/metabolismo
14.
Sci Adv ; 10(23): eadk9000, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38848359

RESUMO

Olfaction is essential for complex social behavior in insects. To discriminate complex social cues, ants evolved an expanded number of odorant receptor (Or) genes. Mutations in the obligate odorant co-receptor gene orco lead to the loss of ~80% of the antennal lobe glomeruli in the jumping ant Harpegnathos saltator. However, the cellular mechanism remains unclear. Here, we demonstrate massive apoptosis of odorant receptor neurons (ORNs) in the mid to late stages of pupal development, possibly due to ER stress in the absence of Orco. Further bulk and single-nucleus transcriptome analysis shows that, although most orco-expressing ORNs die in orco mutants, a small proportion of them survive: They express ionotropic receptor (Ir) genes that form IR complexes. In addition, we found that some Or genes are expressed in mechanosensory neurons and nonneuronal cells, possibly due to leaky regulation from nearby non-Or genes. Our findings provide a comprehensive overview of ORN development and Or expression in H. saltator.


Assuntos
Formigas , Neurônios Receptores Olfatórios , Receptores Odorantes , Animais , Neurônios Receptores Olfatórios/metabolismo , Formigas/genética , Formigas/metabolismo , Formigas/fisiologia , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Apoptose , Mutação , Sobrevivência Celular , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Perfilação da Expressão Gênica , Transcriptoma
15.
Biochem Biophys Res Commun ; 719: 150062, 2024 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-38740002

RESUMO

Pluripotent stem cells, such as embryonic stem cells and induced pluripotent stem cells (iPSCs), can differentiate into almost all cell types and are anticipated to have significant applications in the field of regenerative medicine. However, there are no reports of successfully directing iPSCs to become functional olfactory sensory neurons (OSNs) capable of selectively receiving odorant compounds. In this study, we employed dual SMAD inhibition and fibroblast growth factor 8 (FGF-8, reported to dictate olfactory fates) along with N-2 and B-27 supplements in the culture medium to efficiently induce the differentiation of iPSCs into neuronal cells with olfactory function through olfactory placode. Temporal gene expression and expression of OSN-specific markers during differentiation indicated that the expression of olfactory marker proteins and various olfactory receptors (ORs), which are markers of mature OSNs, was observed after approximately one month of differentiation culture, irrespective of the differentiation cues, suggesting differentiation into OSNs. Cells that exhibited specific responses to odorant compounds were identified after administering odorant compounds to differentiated iPSC-derived OSNs. This suggests the spontaneous generation of functional OSNs expressing diverse ORs that respond to odorant compounds from iPSCs.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Odorantes , Neurônios Receptores Olfatórios , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Humanos , Neurônios Receptores Olfatórios/metabolismo , Neurônios Receptores Olfatórios/citologia , Odorantes/análise , Células Cultivadas , Receptores Odorantes/genética , Receptores Odorantes/metabolismo
16.
Cell Signal ; 120: 111224, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38740233

RESUMO

Early-life critical periods allow initial sensory experience to remodel brain circuitry so that synaptic connectivity can be optimized to environmental input. In the Drosophila juvenile brain, olfactory sensory neuron (OSN) synaptic glomeruli are pruned by glial phagocytosis in dose-dependent response to early odor experience during a well-defined critical period. Extracellular signal-regulated kinase (ERK) separation of phases-based activity reporter of kinase (SPARK) biosensors reveal experience-dependent signaling in glia during this critical period. Glial ERK-SPARK signaling is depressed by removal of Draper receptors orchestrating glial phagocytosis. Cell-targeted genetic knockdown of glial ERK signaling reduces olfactory experience-dependent glial pruning of the OSN synaptic glomeruli in a dose-dependent mechanism. Noonan Syndrome is caused by gain-of-function mutations in protein tyrosine phosphatase non-receptor type 11 (PTPN11) inhibiting ERK signaling, and a glial-targeted patient-derived mutation increases experience-dependent glial ERK signaling and impairs experience-dependent glial pruning of the OSN synaptic glomeruli. We conclude that critical period experience drives glial ERK signaling that is required for dose-dependent pruning of brain synaptic glomeruli, and that altered glial ERK signaling impairs this critical period mechanism in a Noonan Syndrome disease model.


Assuntos
Proteínas de Drosophila , Sistema de Sinalização das MAP Quinases , Neuroglia , Animais , Neuroglia/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Sinapses/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fagocitose
17.
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
18.
Chem Senses ; 492024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38771102

RESUMO

Toxic puffers accumulate tetrodotoxin (TTX), a well-known neurotoxin, by feeding on TTX-bearing organisms and using it to defend themselves from predators. Our previous studies have demonstrated that toxic puffers are attracted to 5,6,11-trideoxytetrodotoxin (TDT), a nontoxic TTX analog that is simultaneously accumulated with TTX in toxic puffers and their prey. In addition, activity labeling using immunohistochemistry targeting neuronal activity marker suggests that TDT activates crypt olfactory sensory neurons (OSN) of the green spotted puffer. However, it remains to be determined whether individual crypt OSNs can physiologically respond to TDT. By employing electroporation to express GCaMP6s in OSNs, we successfully identified a distinct group of oval OSNs that exhibited a specific calcium response when exposed to TDT in green spotted puffers. These oval OSNs showed no response to amino acids (AAs), which serve as food odor cues for teleosts. Furthermore, oval morphology and surface positioning of TDT-sensitive OSNs in the olfactory epithelium closely resemble that of crypt OSNs. These findings further substantiate that TDT is specifically detected by crypt OSNs in green spotted puffer. The TDT odor may act as a chemoattractant for finding conspecific toxic puffers and for feeding TTX-bearing organisms for effective toxification.


Assuntos
Odorantes , Neurônios Receptores Olfatórios , Tetraodontiformes , Tetrodotoxina , Animais , Tetrodotoxina/farmacologia , Neurônios Receptores Olfatórios/efeitos dos fármacos , Neurônios Receptores Olfatórios/metabolismo , Odorantes/análise , Cálcio/metabolismo
19.
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
20.
Mol Neurobiol ; 61(11): 8921-8927, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38570429

RESUMO

The biological substrate of persistent post-COVID-19 hyposmia is still unclear. However, as many neurodegenerative diseases present with smell impairment at onset, it may theoretically reflect degeneration within the central olfactory circuits. However, no data still exist regarding the post-COVID-19 patients. As the olfactory neurons (ONs) mirror pathological changes in the brain, allowing for tracking the underlying molecular events, here, we performed a broad analysis of ONs from patients with persistent post-COVID-19 OD to identify traces of potential neurodegeneration. ONs were collected through the non-invasive brushing of the olfactory mucosa from ten patients with persistent post-COVID-19 hyposmia (lasting > 6 months after infection) and ten age/sex-matched controls. Immunofluorescence staining for protein quantification and RT-PCR for gene expression levels were combined to measure ONs markers of α-synuclein, amyloid-ß, and tau pathology, axonal injury, and mitochondrial network. Patients and controls had similar ONs levels of oligomeric α-synuclein, amyloid-ß peptide, tau protein, neurofilament light chain (NfL), cytochrome C oxidase subunit 3 (COX3), and the heat shock protein 60 (HSP60). Our findings thus did not provide evidence for synucleinopathy and amyloid-ß mismetabolism or gross traces of neuronal injury and mitochondrial dysfunction within the olfactory system in the early phase of persistent post-COVID-19 hyposmia.


Assuntos
Peptídeos beta-Amiloides , Anosmia , Biomarcadores , COVID-19 , Condutos Olfatórios , Proteínas tau , Humanos , COVID-19/complicações , Feminino , Masculino , Anosmia/metabolismo , Pessoa de Meia-Idade , Peptídeos beta-Amiloides/metabolismo , Biomarcadores/metabolismo , Condutos Olfatórios/metabolismo , Condutos Olfatórios/patologia , Proteínas tau/metabolismo , Idoso , alfa-Sinucleína/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/complicações , Mucosa Olfatória/metabolismo , Mucosa Olfatória/patologia , Mucosa Olfatória/virologia , Proteínas de Neurofilamentos/metabolismo , Neurônios Receptores Olfatórios/metabolismo , Adulto , SARS-CoV-2 , Transtornos do Olfato/etiologia , Transtornos do Olfato/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...