Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.141
Filter
1.
Cell Commun Signal ; 22(1): 307, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831315

ABSTRACT

BACKGROUND: Interleukin 24 (IL-24) has been implicated in the nociceptive signaling. However, direct evidence and the precise molecular mechanism underlying IL-24's role in peripheral nociception remain unclear. METHODS: Using patch clamp recording, molecular biological analysis, immunofluorescence labeling, siRNA-mediated knockdown approach and behavior tests, we elucidated the effects of IL-24 on sensory neuronal excitability and peripheral pain sensitivity mediated by T-type Ca2+ channels (T-type channels). RESULTS: IL-24 enhances T-type channel currents (T-currents) in trigeminal ganglion (TG) neurons in a reversible and dose-dependent manner, primarily by activating the interleukin-22 receptor 1 (IL-22R1). Furthermore, we found that the IL-24-induced T-type channel response is mediated through tyrosine-protein kinase Lyn, but not its common downstream target JAK1. IL-24 application significantly activated protein kinase A; this effect was independent of cAMP and prevented by Lyn antagonism. Inhibition of PKA prevented the IL-24-induced T-current response, whereas inhibition of protein kinase C or MAPK kinases had no effect. Functionally, IL-24 increased TG neuronal excitability and enhanced pain sensitivity to mechanical stimuli in mice, both of which were suppressed by blocking T-type channels. In a trigeminal neuropathic pain model induced by chronic constriction injury of the infraorbital nerve, inhibiting IL-22R1 signaling alleviated mechanical allodynia, which was reversed by blocking T-type channels or knocking down Cav3.2. CONCLUSION: Our findings reveal that IL-24 enhances T-currents by stimulating IL-22R1 coupled to Lyn-dependent PKA signaling, leading to TG neuronal hyperexcitability and pain hypersensitivity. Understanding the mechanism of IL-24/IL-22R1 signaling in sensory neurons may pave the way for innovative therapeutic strategies in pain management.


Subject(s)
Calcium Channels, T-Type , Cyclic AMP-Dependent Protein Kinases , Receptors, Interleukin , Sensory Receptor Cells , Signal Transduction , Trigeminal Ganglion , src-Family Kinases , Animals , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , src-Family Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Trigeminal Ganglion/metabolism , Male , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Receptors, Interleukin/metabolism , Mice , Mice, Inbred C57BL , Interleukins/metabolism
2.
Elife ; 122024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896465

ABSTRACT

Spinal pain affects individuals of all ages and is the most common musculoskeletal problem globally. Its clinical management remains a challenge as the underlying mechanisms leading to it are still unclear. Here, we report that significantly increased numbers of senescent osteoclasts (SnOCs) are observed in mouse models of spinal hypersensitivity, like lumbar spine instability (LSI) or aging, compared to controls. The larger population of SnOCs is associated with induced sensory nerve innervation, as well as the growth of H-type vessels, in the porous endplate. We show that deletion of senescent cells by administration of the senolytic drug Navitoclax (ABT263) results in significantly less spinal hypersensitivity, spinal degeneration, porosity of the endplate, sensory nerve innervation, and H-type vessel growth in the endplate. We also show that there is significantly increased SnOC-mediated secretion of Netrin-1 and NGF, two well-established sensory nerve growth factors, compared to non-senescent OCs. These findings suggest that pharmacological elimination of SnOCs may be a potent therapy to treat spinal pain.


Subject(s)
Cellular Senescence , Osteoclasts , Animals , Mice , Osteoclasts/metabolism , Osteoclasts/drug effects , Osteoclasts/physiology , Cellular Senescence/drug effects , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Disease Models, Animal , Male , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Netrin-1/metabolism , Netrin-1/genetics , Mice, Inbred C57BL
3.
Curr Biol ; 34(12): 2756-2763.e2, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38838665

ABSTRACT

Extracellular vesicles (EVs) are submicron membranous structures and key mediators of intercellular communication.1,2 Recent research has highlighted roles for cilia-derived EVs in signal transduction, underscoring their importance as bioactive extracellular organelles containing conserved ciliary signaling proteins.3,4 Members of the transient receptor potential (TRP) channel polycystin-2 (PKD-2) family are found in ciliary EVs of the green algae Chlamydomonas and the nematode Caenorhabditis elegans5,6 and in EVs in the mouse embryonic node and isolated from human urine.7,8 In C. elegans, PKD-2 is expressed in male-specific EV-releasing sensory neurons, which extend ciliary tips to ciliary pore and directly release EVs into the environment.6,9 Males release EVs in a mechanically stimulated manner, regulate EV cargo content in response to mating partners, and deposit PKD-2::GFP-labeled EVs on the vulval cuticle of hermaphrodites during mating.9,10 Combined, our findings suggest that ciliary EV release is a dynamic process. Herein, we identify mechanisms controlling dynamic EV shedding using time-lapse imaging. Cilia can sustain the release of PKD-2-labeled EVs for 2 h. This extended release doesn't require neuronal transmission. Instead, ciliary intrinsic mechanisms regulate PKD-2 ciliary membrane replenishment and dynamic EV release. The kinesin-3 motor kinesin-like protein 6 (KLP-6) is necessary for initial and extended EV release, while the transition zone protein NPHP-4 is required only for sustained EV release. The dynamic replenishment of PKD-2 at the ciliary tip is key to sustained EV release. Our study provides a comprehensive portrait of real-time ciliary EV release and mechanisms supporting cilia as proficient EV release platforms.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cilia , Extracellular Vesicles , Sensory Receptor Cells , TRPP Cation Channels , Animals , Cilia/metabolism , Cilia/physiology , Extracellular Vesicles/metabolism , Extracellular Vesicles/physiology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , TRPP Cation Channels/metabolism , TRPP Cation Channels/genetics , Male
4.
Trends Immunol ; 45(5): 381-396, 2024 May.
Article in English | MEDLINE | ID: mdl-38697871

ABSTRACT

Recent studies have uncovered a new role for sensory neurons in influencing mammalian host immunity, challenging conventional notions of the nervous and immune systems as separate entities. In this review we delve into this groundbreaking paradigm of neuroimmunology and discuss recent scientific evidence for the impact of sensory neurons on host responses against a wide range of pathogens and diseases, encompassing microbial infections and cancers. These valuable insights enhance our understanding of the interactions between the nervous and immune systems, and also pave the way for developing candidate innovative therapeutic interventions in immune-mediated diseases highlighting the importance of this interdisciplinary research field.


Subject(s)
Sensory Receptor Cells , Animals , Humans , Host-Pathogen Interactions , Immunity , Neoplasms/immunology , Neoplasms/therapy , Neuroimmunomodulation , Sensory Receptor Cells/immunology , Sensory Receptor Cells/physiology
5.
PLoS Comput Biol ; 20(5): e1012043, 2024 May.
Article in English | MEDLINE | ID: mdl-38739640

ABSTRACT

Sensory neurons reconstruct the world from action potentials (spikes) impinging on them. To effectively transfer information about the stimulus to the next processing level, a neuron needs to be able to adapt its working range to the properties of the stimulus. Here, we focus on the intrinsic neural properties that influence information transfer in cortical neurons and how tightly their properties need to be tuned to the stimulus statistics for them to be effective. We start by measuring the intrinsic information encoding properties of putative excitatory and inhibitory neurons in L2/3 of the mouse barrel cortex. Excitatory neurons show high thresholds and strong adaptation, making them fire sparsely and resulting in a strong compression of information, whereas inhibitory neurons that favour fast spiking transfer more information. Next, we turn to computational modelling and ask how two properties influence information transfer: 1) spike-frequency adaptation and 2) the shape of the IV-curve. We find that a subthreshold (but not threshold) adaptation, the 'h-current', and a properly tuned leak conductance can increase the information transfer of a neuron, whereas threshold adaptation can increase its working range. Finally, we verify the effect of the IV-curve slope in our experimental recordings and show that excitatory neurons form a more heterogeneous population than inhibitory neurons. These relationships between intrinsic neural features and neural coding that had not been quantified before will aid computational, theoretical and systems neuroscientists in understanding how neuronal populations can alter their coding properties, such as through the impact of neuromodulators. Why the variability of intrinsic properties of excitatory neurons is larger than that of inhibitory ones is an exciting question, for which future research is needed.


Subject(s)
Action Potentials , Adaptation, Physiological , Models, Neurological , Animals , Mice , Action Potentials/physiology , Adaptation, Physiological/physiology , Computational Biology , Computer Simulation , Neurons/physiology , Sensory Receptor Cells/physiology , Somatosensory Cortex/physiology
6.
J Comp Neurol ; 532(6): e25627, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38813969

ABSTRACT

During development, cell-intrinsic and cell-extrinsic factors play important roles in neuronal differentiation; however, the underlying mechanisms in nonmammalian species remain largely unknown. We here investigated the mechanisms responsible for the differentiation of sensory input neurons in the chick entopallium, which receives its primary visual input via the tectofugal pathway from the nucleus rotundus. The results obtained revealed that input neurons in the entopallium expressed Potassium Voltage-Gated Channel Subfamily H Member 5 (KCNH5/EAG2) mRNA from embryonic day (E) 11. On the other hand, the onset of protein expression was E20, which was 1 day before hatching. We confirm that entopallium input neurons in chicks were generated during early neurogenesis in the lateral and ventral ventricular zones. Notably, neurons derived from the lateral (LP) and ventral pallium (VP) exhibited a spatially distinct distribution along the rostro-caudal axis. We further demonstrated that the expression of EAG2 was directly regulated by input activity from thalamic axons. Collectively, the present results reveal that thalamic input activity is essential for specifying input neurons among LP- and VP-derived early-generated neurons in the developing chick entopallium.


Subject(s)
Neurogenesis , Thalamus , Animals , Chick Embryo , Neurogenesis/physiology , Thalamus/embryology , Thalamus/cytology , Thalamus/metabolism , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/cytology , Chickens , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology
7.
Trends Immunol ; 45(5): 371-380, 2024 May.
Article in English | MEDLINE | ID: mdl-38653601

ABSTRACT

Peripheral sensory neurons recognize diverse noxious stimuli, including microbial products and allergens traditionally thought to be targets of the mammalian immune system. Activation of sensory neurons by these stimuli leads to pain and itch responses as well as the release of neuropeptides that interact with their cognate receptors expressed on immune cells, such as dendritic cells (DCs). Neuronal control of immune cell function through neuropeptide release not only affects local inflammatory responses but can impact adaptive immune responses through downstream effects on T cell priming. Numerous neuropeptide receptors are expressed by DCs but only a few have been characterized, presenting opportunities for further investigation of the pathways by which cutaneous neuroimmune interactions modulate host immunity.


Subject(s)
Sensory Receptor Cells , Skin , Humans , Animals , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Skin/immunology , Neuropeptides/metabolism , Neuropeptides/immunology , Dendritic Cells/immunology , Neuroimmunomodulation , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/immunology
8.
eNeuro ; 11(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38627062

ABSTRACT

Proprioception, the sense of limb and body position, is required to produce accurate and precise movements. Proprioceptive sensory neurons transmit muscle length and tension information to the spinal cord. The function of excitatory neurons in the intermediate spinal cord, which receive this proprioceptive information, remains poorly understood. Using genetic labeling strategies and patch-clamp techniques in acute spinal cord preparations in mice, we set out to uncover how two sets of spinal neurons, Clarke's column (CC) and Atoh1-lineage neurons, respond to electrical activity and how their inputs are organized. Both sets of neurons are located in close proximity in laminae V-VII of the thoracolumbar spinal cord and have been described to receive proprioceptive signals. We find that a majority of CC neurons have a tonic-firing type and express a distinctive hyperpolarization-activated current (Ih). Atoh1-lineage neurons, which cluster into two spatially distinct populations, are mostly a fading-firing type and display similar electrophysiological properties to each other, possibly due to their common developmental lineage. Finally, we find that CC neurons respond to stimulation of lumbar dorsal roots, consistent with prior knowledge that CC neurons receive hindlimb proprioceptive information. In contrast, using a combination of electrical stimulation, optogenetic stimulation, and transsynaptic rabies virus tracing, we find that Atoh1-lineage neurons receive heterogeneous, predominantly local thoracic inputs that include parvalbumin-lineage sensory afferents and local interneuron presynaptic inputs. Altogether, we find that CC and Atoh1-lineage neurons have distinct membrane properties and sensory input organization, representing different subcircuit modes of proprioceptive information processing.


Subject(s)
Proprioception , Spinal Cord , Animals , Proprioception/physiology , Spinal Cord/physiology , Spinal Cord/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice, Transgenic , Mice , Male , Female , Action Potentials/physiology , Sensory Receptor Cells/physiology , Patch-Clamp Techniques , Mice, Inbred C57BL , Thoracic Vertebrae
9.
Science ; 384(6693): 295-301, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38669574

ABSTRACT

Airway neuroendocrine (NE) cells have been proposed to serve as specialized sensory epithelial cells that modulate respiratory behavior by communicating with nearby nerve endings. However, their functional properties and physiological roles in the healthy lung, trachea, and larynx remain largely unknown. In this work, we show that murine NE cells in these compartments have distinct biophysical properties but share sensitivity to two commonly aspirated noxious stimuli, water and acid. Moreover, we found that tracheal and laryngeal NE cells protect the airways by releasing adenosine 5'-triphosphate (ATP) to activate purinoreceptive sensory neurons that initiate swallowing and expiratory reflexes. Our work uncovers the broad molecular and biophysical diversity of NE cells across the airways and reveals mechanisms by which these specialized excitable cells serve as sentinels for activating protective responses.


Subject(s)
Adenosine Triphosphate , Larynx , Neuroendocrine Cells , Reflex , Trachea , Animals , Mice , Neuroendocrine Cells/metabolism , Larynx/physiology , Adenosine Triphosphate/metabolism , Reflex/physiology , Trachea/innervation , Trachea/cytology , Deglutition , Lung/physiology , Exhalation/physiology , Water/metabolism , Sensory Receptor Cells/physiology , Mice, Inbred C57BL
10.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38548333

ABSTRACT

PVD neuron of Caenorhabditis elegans is a highly polarized cell with well-defined axonal, and dendritic compartments. PVD neuron operates in multiple sensory modalities including the control of both nociceptive touch sensation and body posture. Although both the axon and dendrites of this neuron show a regeneration response following laser-assisted injury, it is rather unclear how the behavior associated with this neuron is affected by the loss of these structures. It is also unclear whether neurite regrowth would lead to functional restoration in these neurons. Upon axotomy, using a femtosecond laser, we saw that harsh touch response was specifically affected leaving the body posture unperturbed. Subsequently, recovery in the touch response is highly correlated to the axon regrowth, which was dependent on DLK-1/MLK-1 MAP Kinase. Dendrotomy of both major and minor primary dendrites affected the wavelength and amplitude of sinusoidal movement without any apparent effect on harsh touch response. We further correlated the recovery in posture behavior to the type of dendrite regeneration events. We found that dendrite regeneration through the fusion and reconnection between the proximal and distal branches of the injured dendrite corresponded to improved recovery in posture. Our data revealed that the axons and dendrites of PVD neurons regulate the nociception and proprioception in worms, respectively. It also revealed that dendrite and axon regeneration lead to the restoration of these differential sensory modalities.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Dendrites , Nerve Regeneration , Animals , Caenorhabditis elegans/physiology , Dendrites/physiology , Nerve Regeneration/physiology , Caenorhabditis elegans Proteins/metabolism , Recovery of Function/physiology , Sensory Receptor Cells/physiology , Axotomy , Touch/physiology , Animals, Genetically Modified , Axons/physiology , MAP Kinase Kinase Kinases
11.
J Invest Dermatol ; 144(5): 950-953, 2024 May.
Article in English | MEDLINE | ID: mdl-38430083

ABSTRACT

Pruritus or itch is a defining symptom of atopic dermatitis (AD). The origins of itch are complex, and it is considered both a defense mechanism and a cause of disease that leads to inflammation and psychological stress. Considerable progress has been made in understanding the processes that trigger itch, particularly the pruritoceptive origins that are generated in the skin. This perspective review discusses the implications of a recent observation that the V8 protease expressed by Staphylococcus aureus can directly trigger sensory neurons in the skin through activation of protease-activated receptor 1. This may be a key to understanding why itch is so common in AD because S. aureus commonly overgrows in this disease owing to deficient antimicrobial defense from both the epidermis and the cutaneous microbiome. Increased understanding of the role of microbes in AD provides increased opportunities for safely improving the treatment of this disorder.


Subject(s)
Dermatitis, Atopic , Pruritus , Staphylococcus aureus , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/complications , Dermatitis, Atopic/immunology , Humans , Pruritus/microbiology , Pruritus/immunology , Animals , Skin/microbiology , Skin/pathology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Receptors, Proteinase-Activated/metabolism , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology
12.
Blood ; 143(20): 2037-2052, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38427938

ABSTRACT

ABSTRACT: Individuals living with sickle cell disease (SCD) experience severe recurrent acute and chronic pain. Challenges to gaining mechanistic insight into pathogenic SCD pain processes include differential gene expression and function of sensory neurons between humans and mice with SCD, and extremely limited availability of neuronal tissues from patients with SCD. Here, we used induced pluripotent stem cells (iPSCs), derived from patients with SCD, differentiated into sensory neurons (SCD iSNs) to begin to overcome these challenges. We characterize key gene expression and function of SCD iSNs to establish a model to investigate intrinsic and extrinsic factors that may contribute to SCD pain. Despite similarities in receptor gene expression, SCD iSNs show pronounced excitability using patch clamp electrophysiology. Furthermore, we find that plasma taken from patients with SCD during acute pain associated with a vaso-occlusive event increases the calcium responses to the nociceptive stimulus capsaicin in SCD iSNs compared with those treated with paired plasma from patients with SCD at steady state baseline or healthy control plasma samples. We identified high levels of the polyamine spermine in baseline and acute pain states of plasma from patients with SCD, which sensitizes SCD iSNs to subthreshold concentrations of capsaicin. Together, these data identify potential intrinsic mechanisms within SCD iSNs that may extend beyond a blood-based pathology.


Subject(s)
Anemia, Sickle Cell , Induced Pluripotent Stem Cells , Sensory Receptor Cells , Humans , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Sensory Receptor Cells/pathology , Cell Differentiation , Capsaicin/pharmacology , Male , Female , Plasma/metabolism
13.
Pflugers Arch ; 476(6): 975-992, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38538988

ABSTRACT

Human-induced pluripotent stem cells (iPS cells) are efficiently differentiated into sensory neurons. These cells express the voltage-gated sodium channel NaV1.7, which is a validated pain target. NaV1.7 deficiency leads to pain insensitivity, whereas NaV1.7 gain-of-function mutants are associated with chronic pain. During differentiation, the sensory neurons start spontaneous action potential firing around day 22, with increasing firing rate until day 40. Here, we used CRISPR/Cas9 genome editing to generate a HA-tag NaV1.7 to follow its expression during differentiation. We used two protocols to generate sensory neurons: the classical small molecule approach and a directed differentiation methodology and assessed surface NaV1.7 expression by Airyscan high-resolution microscopy. Our results show that maturation of at least 49 days is necessary to observe robust NaV1.7 surface expression in both protocols. Electric activity of the sensory neurons precedes NaV1.7 surface expression. A clinically effective NaV1.7 blocker is still missing, and we expect this iPS cell model system to be useful for drug discovery and disease modeling.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , NAV1.7 Voltage-Gated Sodium Channel , Sensory Receptor Cells , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Action Potentials , CRISPR-Cas Systems
14.
Nat Commun ; 15(1): 1660, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38396085

ABSTRACT

Animals must sense and acclimatize to environmental temperatures for survival, yet their thermosensing mechanisms other than transient receptor potential (TRP) channels remain poorly understood. We identify a trimeric G protein-coupled receptor (GPCR), SRH-40, which confers thermosensitivity in sensory neurons regulating temperature acclimatization in Caenorhabditis elegans. Systematic knockdown of 1000 GPCRs by RNAi reveals GPCRs involved in temperature acclimatization, among which srh-40 is highly expressed in the ADL sensory neuron, a temperature-responsive chemosensory neuron, where TRP channels act as accessorial thermoreceptors. In vivo Ca2+ imaging demonstrates that an srh-40 mutation reduced the temperature sensitivity of ADL, resulting in supranormal temperature acclimatization. Ectopically expressing SRH-40 in a non-warmth-sensing gustatory neuron confers temperature responses. Moreover, temperature-dependent SRH-40 activation is reconstituted in Drosophila S2R+ cells. Overall, SRH-40 may be involved in thermosensory signaling underlying temperature acclimatization. We propose a dual thermosensing machinery through a GPCR and TRP channels in a single sensory neuron.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Temperature , Sensory Receptor Cells/physiology , Caenorhabditis elegans Proteins/genetics , Acclimatization/genetics , Receptors, G-Protein-Coupled/genetics
15.
J Comp Neurol ; 532(2): e25575, 2024 02.
Article in English | MEDLINE | ID: mdl-38335058

ABSTRACT

The distinct organization of Kv2 voltage-gated potassium channels on and near the cell body of brain neurons enables their regulation of action potentials and specialized membrane contact sites. Somatosensory neurons have a pseudounipolar morphology and transmit action potentials from peripheral nerve endings through axons that bifurcate to the spinal cord and the cell body within ganglia including the dorsal root ganglia (DRG). Kv2 channels regulate action potentials in somatosensory neurons, yet little is known about where Kv2 channels are located. Here, we define the cellular and subcellular localization of the Kv2 paralogs, Kv2.1 and Kv2.2, in DRG somatosensory neurons with a panel of antibodies, cell markers, and genetically modified mice. We find that relative to spinal cord neurons, DRG neurons have similar levels of detectable Kv2.1 and higher levels of Kv2.2. In older mice, detectable Kv2.2 remains similar, while detectable Kv2.1 decreases. Both Kv2 subtypes adopt clustered subcellular patterns that are distinct from central neurons. Most DRG neurons co-express Kv2.1 and Kv2.2, although neuron subpopulations show preferential expression of Kv2.1 or Kv2.2. We find that Kv2 protein expression and subcellular localization are similar between mouse and human DRG neurons. We conclude that the organization of both Kv2 channels is consistent with physiological roles in the somata and stem axons of DRG neurons. The general prevalence of Kv2.2 in DRG as compared to central neurons and the enrichment of Kv2.2 relative to detectable Kv2.1 in older mice, proprioceptors, and axons suggest more widespread roles for Kv2.2 in DRG neurons.


Subject(s)
Axons , Ganglia, Spinal , Mice , Humans , Animals , Action Potentials , Sensory Receptor Cells/physiology
16.
J Neurosci ; 44(16)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38423763

ABSTRACT

Peripheral sensory neurons are a critical part of the nervous system that transmit a multitude of sensory stimuli to the central nervous system. During larval and juvenile stages in zebrafish, this function is mediated by Rohon-Beard somatosensory neurons (RBs). RBs are optically accessible and amenable to experimental manipulation, making them a powerful system for mechanistic investigation of sensory neurons. Previous studies provided evidence that RBs fall into multiple subclasses; however, the number and molecular makeup of these potential RB subtypes have not been well defined. Using a single-cell RNA sequencing (scRNA-seq) approach, we demonstrate that larval RBs in zebrafish fall into three, largely nonoverlapping classes of neurons. We also show that RBs are molecularly distinct from trigeminal neurons in zebrafish. Cross-species transcriptional analysis indicates that one RB subclass is similar to a mammalian group of A-fiber sensory neurons. Another RB subclass is predicted to sense multiple modalities, including mechanical stimulation and chemical irritants. We leveraged our scRNA-seq data to determine that the fibroblast growth factor (Fgf) pathway is active in RBs. Pharmacological and genetic inhibition of this pathway led to defects in axon maintenance and RB cell death. Moreover, this can be phenocopied by treatment with dovitinib, an FDA-approved Fgf inhibitor with a common side effect of peripheral neuropathy. Importantly, dovitinib-mediated axon loss can be suppressed by loss of Sarm1, a positive regulator of neuronal cell death and axonal injury. This offers a molecular target for future clinical intervention to fight neurotoxic effects of this drug.


Subject(s)
Sensory Receptor Cells , Zebrafish , Animals , Zebrafish/metabolism , Animals, Genetically Modified , Cell Survival , Sensory Receptor Cells/physiology , Axons/physiology , Single-Cell Analysis , Mammals
17.
Cell Host Microbe ; 32(1): 3-4, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38211562

ABSTRACT

In a recent issue of Cell, Deng et al. show that S. aureus serine protease V8 triggers itch, independent of inflammation, by activating sensory neurons through PAR1. This study presents mechanistic insights into pruritogenic bacteria and their interactions with sensory neurons while providing a possible approach for treating itch-related diseases.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Humans , Sensory Receptor Cells/physiology , Pruritus , Inflammation
18.
J Invest Dermatol ; 144(7): 1449-1453, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38206270

ABSTRACT

Mechanical itch, which is defined as an itch sensation caused by innocuous mechanical force, may warn of the potential risk in the skin. The increased mechanosensitivity in sensory neurons may cause scratch-induced itch and promote the transition from acute itch to chronic itch. Recent studies have not only expanded our knowledge about the neuronal circuits in the CNS but have also highlighted the importance of the peripheral epithelia-immune-neuronal crosstalk in the development of mechanical itch. In this review, we will summarize related findings about the molecular and cellular mechanisms of mechanical itch in the skin.


Subject(s)
Pruritus , Sensory Receptor Cells , Skin , Pruritus/immunology , Pruritus/physiopathology , Pruritus/etiology , Humans , Sensory Receptor Cells/physiology , Animals , Skin/immunology , Skin/pathology , Mechanotransduction, Cellular
19.
Ocul Surf ; 32: 60-70, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38242319

ABSTRACT

PURPOSE: Sensory nerve terminals are highly distributed in the cornea, and regulate ocular surface sensation and homeostasis in response to various endogenous and exogenous stimuli. However, little is known about mediators regulating the physiological and pathophysiological activities of corneal sensory nerves. The aim of this study was to investigate the presence of cholinergic regulation in sensory nerves in the cornea. METHODS: Localization of choline acetyltransferase (ChAT) and vesicular acetylcholine transporter (vAChT) was evaluated using western blotting and immunohistochemical analysis. The synthesis and liberation of acetylcholine from the cornea were assessed using corneal segments pre-incubated with [3H]choline. The responsiveness of corneal neurons and nerves to cholinergic drugs was explored using calcium imaging with primary cultures of trigeminal ganglion neurons and extracellular recording from corneal preparations in guinea pigs. RESULTS: ChAT, but not vAChT, was highly distributed in the corneal epithelium. In corneal segments, [3H] acetylcholine was synthesized from [3H]choline, and was also released in response to electrical stimuli. In cultured corneal neurons, the population sensitive to a transient receptor potential melastatin 8 (TRPM8) agonist exhibited high probability of responding to nicotine in a calcium imaging experiment. The firing frequency of cold-sensitive corneal nerves was increased by the application of nicotine, but diminished by an α4 nicotinic acetylcholine receptor antagonist. CONCLUSIONS: The corneal epithelium can synthesize and release acetylcholine. Corneal acetylcholine can excite sensory nerves via nicotinic receptors containing the α4 subunit. Therefore, corneal acetylcholine may be one of the important regulators of corneal nerve activity arranging ocular surface condition and sensation.


Subject(s)
Acetylcholine , Cornea , Receptors, Nicotinic , Animals , Acetylcholine/metabolism , Acetylcholine/pharmacology , Cornea/innervation , Cornea/metabolism , Guinea Pigs , Receptors, Nicotinic/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Blotting, Western , Cells, Cultured , Male , Trigeminal Ganglion/metabolism , Immunohistochemistry , Choline O-Acetyltransferase/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
20.
Exp Physiol ; 109(1): 45-54, 2024 01.
Article in English | MEDLINE | ID: mdl-37417654

ABSTRACT

Proprioceptors are non-nociceptive low-threshold mechanoreceptors. However, recent studies have shown that proprioceptors are acid-sensitive and express a variety of proton-sensing ion channels and receptors. Accordingly, although proprioceptors are commonly known as mechanosensing neurons that monitor muscle contraction status and body position, they may have a role in the development of pain associated with tissue acidosis. In clinical practice, proprioception training is beneficial for pain relief. Here we summarize the current evidence to sketch a different role of proprioceptors in 'non-nociceptive pain' with a focus on their acid-sensing properties.


Subject(s)
Musculoskeletal Pain , Humans , Acid Sensing Ion Channels/physiology , Sensory Receptor Cells/physiology , Mechanoreceptors , Proprioception/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...