Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Physiol Rev ; 100(2): 725-803, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31670612

ABSTRACT

The transient receptor potential ankyrin (TRPA) channels are Ca2+-permeable nonselective cation channels remarkably conserved through the animal kingdom. Mammals have only one member, TRPA1, which is widely expressed in sensory neurons and in non-neuronal cells (such as epithelial cells and hair cells). TRPA1 owes its name to the presence of 14 ankyrin repeats located in the NH2 terminus of the channel, an unusual structural feature that may be relevant to its interactions with intracellular components. TRPA1 is primarily involved in the detection of an extremely wide variety of exogenous stimuli that may produce cellular damage. This includes a plethora of electrophilic compounds that interact with nucleophilic amino acid residues in the channel and many other chemically unrelated compounds whose only common feature seems to be their ability to partition in the plasma membrane. TRPA1 has been reported to be activated by cold, heat, and mechanical stimuli, and its function is modulated by multiple factors, including Ca2+, trace metals, pH, and reactive oxygen, nitrogen, and carbonyl species. TRPA1 is involved in acute and chronic pain as well as inflammation, plays key roles in the pathophysiology of nearly all organ systems, and is an attractive target for the treatment of related diseases. Here we review the current knowledge about the mammalian TRPA1 channel, linking its unique structure, widely tuned sensory properties, and complex regulation to its roles in multiple pathophysiological conditions.


Subject(s)
Calcium Signaling , Mechanotransduction, Cellular , Nociception , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel/metabolism , Thermosensing , Animals , Channelopathies/metabolism , Channelopathies/physiopathology , Chemoreceptor Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/physiopathology , Mechanoreceptors/metabolism , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Thermoreceptors/metabolism
2.
Nature ; 555(7694): 98-102, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29466329

ABSTRACT

Circadian clocks coordinate behaviour, physiology and metabolism with Earth's diurnal cycle. These clocks entrain to both light and temperature cycles, and daily environmental temperature oscillations probably contribute to human sleep patterns. However, the neural mechanisms through which circadian clocks monitor environmental temperature and modulate behaviour remain poorly understood. Here we elucidate how the circadian clock neuron network of Drosophila melanogaster processes changes in environmental temperature. In vivo calcium-imaging techniques demonstrate that the posterior dorsal neurons 1 (DN1ps), which are a discrete subset of sleep-promoting clock neurons, constantly monitor modest changes in environmental temperature. We find that these neurons are acutely inhibited by heating and excited by cooling; this is an unexpected result when considering the strong correlation between temperature and light, and the fact that light excites clock neurons. We demonstrate that the DN1ps rely on peripheral thermoreceptors located in the chordotonal organs and the aristae. We also show that the DN1ps and their thermosensory inputs are required for the normal timing of sleep in the presence of naturalistic temperature cycles. These results identify the DN1ps as a major gateway for temperature sensation into the circadian neural network, which continuously integrates temperature changes to coordinate the timing of sleep and activity.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Neurons/physiology , Sleep/physiology , Temperature , Thermosensing/physiology , Animals , Cold Temperature , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Female , Hot Temperature , Locomotion/physiology , Male , Nerve Net/cytology , Nerve Net/physiology , Neural Inhibition , Thermoreceptors/metabolism , Time Factors
3.
Mol Microbiol ; 113(3): 588-592, 2020 03.
Article in English | MEDLINE | ID: mdl-31971637

ABSTRACT

An outstanding question regards the ability of organisms to sense their environments and respond in a suitable way. Pathogenic bacteria in particular exploit host-temperature sensing as a cue for triggering virulence gene expression. This micro-review does not attempt to fully cover the field of bacterial thermosensors and in detail describe each identified case. Instead, the review focus on the time-period at the end of the 1990's and beginning of the 2000's when several key discoveries were made, identifying protein, DNA and RNA as potential thermosensors controlling gene expression in several different bacterial pathogens in general and on the prfA thermosensor of Listeria monocytogenes in particular.


Subject(s)
Bacteria/metabolism , Host Microbial Interactions/physiology , Thermoreceptors/physiology , Bacteria/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Hot Temperature , Listeria monocytogenes/metabolism , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , RNA/genetics , RNA/metabolism , Thermoreceptors/metabolism , Thermosensing/genetics , Thermosensing/physiology , Trans-Activators/metabolism , Virulence/genetics , Virulence Factors/genetics
4.
Nature ; 519(7543): 358-61, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25739506

ABSTRACT

In Drosophila, rapid temperature changes are detected at the periphery by dedicated receptors forming a simple sensory map for hot and cold in the brain. However, flies show a host of complex innate and learned responses to temperature, indicating that they are able to extract a range of information from this simple input. Here we define the anatomical and physiological repertoire for temperature representation in the Drosophila brain. First, we use a photolabelling strategy to trace the connections that relay peripheral thermosensory information to higher brain centres, and show that they largely converge onto three target regions: the mushroom body, the lateral horn (both of which are well known centres for sensory processing) and the posterior lateral protocerebrum, a region we now define as a major site of thermosensory representation. Next, using in vivo calcium imaging, we describe the thermosensory projection neurons selectively activated by hot or cold stimuli. Fast-adapting neurons display transient ON and OFF responses and track rapid temperature shifts remarkably well, while slow-adapting cell responses better reflect the magnitude of simple thermal changes. Unexpectedly, we also find a population of broadly tuned cells that respond to both heating and cooling, and show that they are required for normal behavioural avoidance of both hot and cold in a simple two-choice temperature preference assay. Taken together, our results uncover a coordinated ensemble of neural responses to temperature in the Drosophila brain, demonstrate that a broadly tuned thermal line contributes to rapid avoidance behaviour, and illustrate how stimulus quality, temporal structure, and intensity can be extracted from a simple glomerular map at a single synaptic station.


Subject(s)
Brain/physiology , Drosophila melanogaster/physiology , Neural Pathways , Temperature , Thermosensing/physiology , Animals , Brain/anatomy & histology , Brain/cytology , Brain Mapping , Calcium/analysis , Calcium/metabolism , Drosophila melanogaster/cytology , Mushroom Bodies/innervation , Neurons/metabolism , Synapses/metabolism , Thermoreceptors/metabolism , Time Factors
5.
Int J Mol Sci ; 21(12)2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32580281

ABSTRACT

TRPM8 is the main molecular entity responsible for cold sensing. This polymodal ion channel is activated by cold, cooling compounds such as menthol, voltage, and rises in osmolality. In corneal cold thermoreceptor neurons (CTNs), TRPM8 expression determines not only their sensitivity to cold, but also their role as neural detectors of ocular surface wetness. Several reports suggest that Protein Kinase C (PKC) activation impacts on TRPM8 function; however, the molecular bases of this functional modulation are still poorly understood. We explored PKC-dependent regulation of TRPM8 using Phorbol 12-Myristate 13-Acetate to activate this kinase. Consistently, recombinant TRPM8 channels, cultured trigeminal neurons, and free nerve endings of corneal CTNs revealed a robust reduction of TRPM8-dependent responses under PKC activation. In corneal CTNs, PKC activation decreased ongoing activity, a key parameter in the role of TRPM8-expressing neurons as humidity detectors, and also the maximal cold-evoked response, which were validated by mathematical modeling. Biophysical analysis indicated that PKC-dependent downregulation of TRPM8 is mainly due to a decreased maximal conductance value, and complementary noise analysis revealed a reduced number of functional channels at the cell surface, providing important clues to understanding the molecular mechanisms of how PKC activity modulates TRPM8 channels in CTNs.


Subject(s)
Cold Temperature , Neurons/metabolism , Protein Kinase C/metabolism , TRPM Cation Channels/metabolism , Thermoreceptors/metabolism , Thermosensing , Trigeminal Nerve/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Sensory Receptor Cells/metabolism , Trigeminal Nerve/cytology
6.
Exp Physiol ; 103(10): 1302-1308, 2018 10.
Article in English | MEDLINE | ID: mdl-30070742

ABSTRACT

NEW FINDINGS: What is the central question of this study? How do gastric stretch and gastric cooling stimuli affect cardiac autonomic control? What is the main finding and its importance? Gastric stretch causes an increase in cardiac sympathetic activity. Stretch combined with cold stimulation result in an elimination of the sympathetic response to stretch and an increase in cardiac parasympathetic activity, in turn resulting in a reduction in heart rate. Gastric cold stimulation causes a shift in sympathovagal balance towards parasympathetic dominance. The cold-induced bradycardia has the potential to decrease cardiac workload, which might be significant in individuals with cardiovascular pathologies. ABSTRACT: Gastric distension increases blood pressure and heart rate in young, healthy humans, but little is known about the effect of gastric stretch combined with cooling. We used a randomized crossover study to assess the cardiovascular responses to drinking 300 ml of ispaghula husk solution at either 6 or 37°C in nine healthy humans (age 24.08 ± 9.36 years) to establish the effect of gastric stretch with and without cooling. The effect of consuming peppermint oil capsules to activate cold thermoreceptors was also investigated. The ECG, respiratory movements and continuous blood pressure were recorded during a 5 min baseline period, followed by a 115 min post-drink period, during which 5 min epochs of data were recorded. Cardiac autonomic activity was assessed using time and frequency domain analyses of respiratory sinus arrhythmia to quantify parasympathetic autonomic activity, and corrected QT (QTc) interval analysis to quantify sympathetic autonomic activity. Gastric stretch only caused a significant reduction in QTc interval lasting up to 15 min, with a concomitant but non-significant increase in heart rate, indicating an increased sympathetic cardiac tone. The additional effect of gastric cold stimulation was significantly to reduce heart rate for up to 15 min, elevate indicators of cardiac parasympathetic tone and eliminate the reduction in QTc interval seen with gastric stretch only. Stimulation of gastric cold thermoreceptors with menthol also caused a significant reduction in heart rate and concomitant increase in the root mean square of successive differences. These findings indicate that gastric cold stimulation causes a shift in the sympathovagal balance of cardiac control towards a more parasympathetic dominant pattern.


Subject(s)
Heart Rate/drug effects , Heart/drug effects , Menthol/administration & dosage , Adult , Autonomic Nervous System/drug effects , Blood Pressure/drug effects , Bradycardia/metabolism , Cold Temperature , Cross-Over Studies , Electrocardiography/drug effects , Healthy Volunteers , Humans , Mentha piperita , Plant Oils/administration & dosage , Psyllium/administration & dosage , Thermoreceptors/metabolism , Young Adult
7.
Somatosens Mot Res ; 34(2): 85-91, 2017 06.
Article in English | MEDLINE | ID: mdl-28325123

ABSTRACT

PURPOSE: Facilitatory and inhibitory responses of spinal motor neurons are influenced by somatosensory input from the skin. The purpose of this study, employing electromyography, was to examine the neuromuscular changes that occur with menthol applied to the skin over the quadriceps muscle. METHODS: Forty-two healthy volunteers performed isometric knee extensions at 35% maximum voluntary contraction (MVC) in three groups (Adult Placebo, Adult Menthol, Older Adult Menthol). Stimulation used was application of 5% menthol gel to the skin. Surface electromyography (sEMG) from the vastus lateralis (VL), vastus medialis (VM), and rectus femoris (RF) was recorded using miniature pair electrodes. RESULTS: Root mean square electromyography (rmsEMG) in VL and VM significantly increased with menthol stimulation both in Adult and Older Adult, but no significant difference was observed between Adult Menthol and Older Adult Menthol. There was a significant decrease in mean power frequency (MPF) in VM with menthol stimulation in Older Adult, but no significant changes were observed in Adult Menthol. CONCLUSION: Neuromuscular modulation was observed with the application of menthol gel at low loads in the present study. These findings could lead to a new method of muscular training that targets the recruitment of fast type muscle safe for older adults.


Subject(s)
Isometric Contraction/physiology , Menthol/pharmacology , Motor Neurons/physiology , Quadriceps Muscle/physiology , Thermoreceptors/metabolism , Adult , Aged , Electromyography , Female , Humans , Isometric Contraction/drug effects , Male , Middle Aged , Motor Neurons/drug effects , Quadriceps Muscle/drug effects , Skin/drug effects , Skin/innervation , Thermoreceptors/drug effects , Young Adult
8.
Adv Exp Med Biol ; 1015: 265-277, 2017.
Article in English | MEDLINE | ID: mdl-29080031

ABSTRACT

In primary sensory neurons of the spinal and trigeminal somatosensory system, cold-sensitivity is strongly dependent on the functional balance between TRPM8 channels, the main molecular entity responsible for the cold-activated excitatory current, and Shaker-like Kv1.1-1.2 potassium channels, the molecular counterpart underlying the excitability brake current IKD. This slow-inactivating outward K+ current reduces the excitability of cold thermoreceptor neurons increasing their thermal threshold, and prevents unspecific activation by cold of neurons of other somatosensory modalities. Here we examine the main biophysical properties of this current in primary sensory neurons, its central role in cold thermotransduction, and its contribution to alterations in cold sensitivity triggered by peripheral nerve damage.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/metabolism , Kv1.1 Potassium Channel/metabolism , Sensory Receptor Cells/metabolism , Thermoreceptors/metabolism , Animals , Cold Temperature , TRPM Cation Channels/metabolism
9.
Proc Natl Acad Sci U S A ; 111(41): 14941-6, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25246547

ABSTRACT

Relying almost exclusively on their acute sense of touch, tactile-foraging birds can feed in murky water, but the cellular mechanism is unknown. Mechanical stimuli activate specialized cutaneous end organs in the bill, innervated by trigeminal afferents. We report that trigeminal ganglia (TG) of domestic and wild tactile-foraging ducks exhibit numerical expansion of large-diameter mechanoreceptive neurons expressing the mechano-gated ion channel Piezo2. These features are not found in visually foraging birds. Moreover, in the duck, the expansion of mechanoreceptors occurs at the expense of thermosensors. Direct mechanical stimulation of duck TG neurons evokes high-amplitude depolarizing current with a low threshold of activation, high signal amplification gain, and slow kinetics of inactivation. Together, these factors contribute to efficient conversion of light mechanical stimuli into neuronal excitation. Our results reveal an evolutionary strategy to hone tactile perception in vertebrates at the level of primary afferents.


Subject(s)
Ducks/physiology , Feeding Behavior , Mechanotransduction, Cellular , Neurons/physiology , Touch/physiology , Animals , Down-Regulation , Ion Channel Gating , Ion Channels/metabolism , Sensory Thresholds , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Thermoreceptors/metabolism , Trigeminal Ganglion/physiology , Up-Regulation
10.
EMBO J ; 30(3): 582-93, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21139565

ABSTRACT

The ion-channel TRPV1 is believed to be a major sensor of noxious heat, but surprisingly animals lacking TRPV1 still display marked responses to elevated temperature. In this study, we explored the role of TRPV1-expressing neurons in somatosensation by generating mice wherein this lineage of cells was selectively labelled or ablated. Our data show that TRPV1 is an embryonic marker of many nociceptors including all TRPV1- and TRPM8-neurons as well as many Mrg-expressing neurons. Mutant mice lacking these cells are completely insensitive to hot or cold but in marked contrast retain normal touch and mechanical pain sensation. These animals also exhibit defective body temperature control and lose both itch and pain reactions to potent chemical mediators. Together with previous cell ablation studies, our results define and delimit the roles of TRPV1- and TRPM8-neurons in thermosensation, thermoregulation and nociception, thus significantly extending the concept of labelled lines in somatosensory coding.


Subject(s)
Body Temperature Regulation/physiology , Gene Expression Regulation, Developmental/physiology , Nociceptors/metabolism , TRPV Cation Channels/metabolism , Thermoreceptors/metabolism , Animals , Body Temperature , Body Temperature Regulation/genetics , DNA, Complementary/genetics , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Mice , Mice, Mutant Strains , Models, Biological , Oligonucleotide Array Sequence Analysis , Pain Measurement , Receptors, G-Protein-Coupled/metabolism , Rotarod Performance Test , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics
11.
J Neurosci ; 33(42): 16627-41, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133266

ABSTRACT

Topically applied camphor elicits a sensation of cool, but nothing is known about how it affects cold temperature sensing. We found that camphor sensitizes a subpopulation of menthol-sensitive native cutaneous nociceptors in the mouse to cold, but desensitizes and partially blocks heterologously expressed TRPM8 (transient receptor potential cation channel subfamily M member 8). In contrast, camphor reduces potassium outward currents in cultured sensory neurons and, in cold nociceptors, the cold-sensitizing effects of camphor and menthol are additive. Using a membrane potential dye-based screening assay and heterologously expressed potassium channels, we found that the effects of camphor are mediated by inhibition of Kv7.2/3 channels subtypes that generate the M-current in neurons. In line with this finding, the specific M-current blocker XE991 reproduced the cold-sensitizing effect of camphor in nociceptors. However, the M-channel blocking effects of XE991 and camphor are not sufficient to initiate cold transduction but require a cold-activated inward current generated by TRPM8. The cold-sensitizing effects of XE991 and camphor are largest in high-threshold cold nociceptors. Low-threshold corneal cold thermoreceptors that express high levels of TRPM8 and lack potassium channels are not affected by camphor. We also found that menthol--like camphor--potently inhibits Kv7.2/3 channels. The apparent functional synergism arising from TRPM8 activation and M-current block can improve the effectiveness of topical coolants and cooling lotions, and may also enhance TRPM8-mediated analgesia.


Subject(s)
Nociceptors/physiology , Signal Transduction/physiology , TRPM Cation Channels/metabolism , Thermoreceptors/physiology , Thermosensing/physiology , Animals , Camphor/pharmacology , Cold Temperature , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Menthol/pharmacology , Mice , Mice, Inbred C57BL , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/metabolism , Nociceptors/metabolism , Signal Transduction/drug effects , TRPM Cation Channels/genetics , Thermoreceptors/metabolism , Thermosensing/drug effects
12.
J Biol Chem ; 287(22): 18218-29, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22493431

ABSTRACT

TRPM8 is a member of the transient receptor potential ion channel superfamily, which is expressed in sensory neurons and is activated by cold and cooling compounds, such as menthol. Activation of TRPM8 by agonists takes place through shifts in its voltage activation curve, allowing channel opening at physiological membrane potentials. Here, we studied the role of the N-glycosylation occurring at the pore loop of TRPM8 on the function of the channel. Using heterologous expression of recombinant channels in HEK293 cells we found that the unglycosylated TRPM8 mutant (N934Q) displays marked functional differences compared with the wild type channel. These differences include a shift in the threshold of temperature activation and a reduced response to menthol and cold stimuli. Biophysical analysis indicated that these modifications are due to a shift in the voltage dependence of TRPM8 activation toward more positive potentials. By using tunicamycin, a drug that prevents N-glycosylation of proteins, we also evaluated the effect of the N-glycosylation on the responses of trigeminal sensory neurons expressing TRPM8. These experiments showed that the lack of N-glycosylation affects the function of native TRPM8 ion channels in a similar way to heterologously expressed ones, causing an important shift of the temperature threshold of cold-sensitive thermoreceptor neurons. Altogether, these results indicate that post-translational modification of TRPM8 is an important mechanism modulating cold thermoreceptor function, explaining the marked differences in temperature sensitivity observed between recombinant and native TRPM8 ion channels.


Subject(s)
Cold Temperature , Neurons/physiology , TRPM Cation Channels/metabolism , Thermoreceptors/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Glycosylation , Humans , Mice , Protein Processing, Post-Translational , TRPM Cation Channels/physiology , Thermoreceptors/physiology
13.
J Biol Chem ; 287(5): 2963-70, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22128172

ABSTRACT

Transient receptor potential (TRP) channels couple various environmental factors to changes in membrane potential, calcium influx, and cell signaling. They also integrate multiple stimuli through their typically polymodal activation. Thus, although the TRPM8 channel has been extensively investigated as the major neuronal cold sensor, it is also regulated by various chemicals, as well as by several short channel isoforms. Mechanistic understanding of such complex regulation is facilitated by quantitative single-channel analysis. We have recently proposed a single-channel mechanism of TRPM8 regulation by voltage and temperature. Using this gating mechanism, we now investigate TRPM8 inhibition in cell-attached patches using HEK293 cells expressing TRPM8 alone or coexpressed with its short sM8-6 isoform. This is compared with inhibition by the chemicals N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)piperazine-1-carboxamide (BCTC) and clotrimazole or by elevated temperature. We found that within the seven-state single-channel gating mechanism, inhibition of TRPM8 by short sM8-6 isoforms closely resembles inhibition by increased temperature. In contrast, inhibition by BCTC and that by clotrimazole share a different set of common features.


Subject(s)
Antifungal Agents/pharmacology , Clotrimazole/pharmacology , Hot Temperature , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Pyrazines/pharmacology , Pyridines/pharmacology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , HEK293 Cells , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , TRPM Cation Channels/genetics , Thermoreceptors/metabolism
15.
J Inorg Biochem ; 229: 111745, 2022 04.
Article in English | MEDLINE | ID: mdl-35121188

ABSTRACT

A conservative characteristic of manganese superoxide dismutase is the rapid formation of product inhibition at high temperatures. At lower temperatures, the enzyme is less inhibited and undergoes more catalytic fast cycles before being product-inhibited. The temperature-dependent kinetics could be rationalized by the temperature-dependent coordination in the conserved center of manganese superoxide dismutase. As temperature decreases, a water molecule (WAT2) approaches or even coordinates Mn as the sixth ligand to interfere with O2•--Mn coordination and reduce product inhibition, so the dismutation should mainly proceed in the fast outer-sphere pathway at low temperatures. Cold-activation is an adaptive response to low temperature rather than a passive adaptation to excess superoxide levels since the cold-activated dismutase activity significantly exceeds the amount of superoxide in the cell or mitochondria. Physiologically speaking, cold activation of manganese superoxide dismutase mediates cold stress signaling and transduces temperature (physical signal) degree into H2O2 fluxes (chemical signal), which in turn may act as a second messenger to induce a series of physiological responses such as cold shock.


Subject(s)
Superoxide Dismutase/metabolism , Thermoreceptors/metabolism , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cold Temperature , Cold-Shock Response/physiology , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungi/enzymology , Humans , Hydrogen Peroxide/metabolism , Manganese/chemistry , Oxidative Stress/physiology , Protein Conformation , Signal Transduction/physiology , Superoxide Dismutase/chemistry , Superoxides/chemistry , Superoxides/metabolism , Thermoreceptors/chemistry
16.
Pharmacol Ther ; 240: 108297, 2022 12.
Article in English | MEDLINE | ID: mdl-36202261

ABSTRACT

Chronic pain is a major burden for the society and remains more prevalent and severe in females. The presence of chronic pain is linked to persistent alterations in the peripheral and the central nervous system. One of the main types of peripheral pain transducers are the transient receptor potential channels (TRP), also known as thermoTRP channels, which intervene in the perception of hot and cold external stimuli. These channels, and especially TRPV1, TRPA1 and TRPM8, have been subjected to profound investigation because of their role as thermosensors and also because of their implication in acute and chronic pain. Surprisingly, their sensitivity to endogenous signaling has been far less studied. Cumulative evidence suggests that the function of these channels may be differently modulated in males and females, in part through sexual hormones, and this could constitute a significant contributor to the sex differences in chronic pain. Here, we review the exciting advances in thermoTRP pharmacology for males and females in two paradigmatic types of chronic pain with a strong peripheral component: chronic migraine and chemotherapy-induced peripheral neuropathy (CIPN). The possibilities of peripheral druggability offered by these channels and the differential exploitation for men and women represent a development opportunity that will lead to a significant increment of the armamentarium of analgesic medicines for personalized chronic pain treatment.


Subject(s)
Chronic Pain , Migraine Disorders , Peripheral Nervous System Diseases , Thermoreceptors , Transient Receptor Potential Channels , Female , Humans , Male , Analgesics/therapeutic use , Chronic Pain/drug therapy , Migraine Disorders/drug therapy , Sex Characteristics , Transient Receptor Potential Channels/metabolism , Antineoplastic Agents/adverse effects , Thermoreceptors/metabolism
17.
Fiziol Cheloveka ; 37(2): 71-6, 2011.
Article in Russian | MEDLINE | ID: mdl-21542321

ABSTRACT

The examination of people belonging to the Russian ethnic group revealed that 20.3% of subjects had heterozygous genotype, containing the C-allele in single nucleotide polymorphism rs11562975, located in exon 7 of the gene encoding the temperature-sensitive ion channel TRPM8. Functional differences, associated with sensitivity to cold and menthol were identified between subjects with different genotypes of the polymorphism rs11562975 (GG and GC). Subjects with heterozygous genotype GC were characterized by increased sensitivity to cold and reduced sensitivity to menthol, agonist of the ion channel TRPM8, compared with subjects with homozygous genotype GG.


Subject(s)
Cold Temperature , Menthol , Polymorphism, Single Nucleotide , TRPM Cation Channels/genetics , Thermosensing/genetics , Adult , DNA/analysis , Female , Heterozygote , Homozygote , Humans , Male , Menthol/pharmacology , Polymerase Chain Reaction , Skin/metabolism , Skin Temperature/genetics , Skin Temperature/physiology , TRPM Cation Channels/agonists , Thermoreceptors/metabolism , Young Adult
18.
J Laryngol Otol ; 135(2): 104-109, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33612130

ABSTRACT

BACKGROUND: The mechanism of nasal airflow sensation is poorly understood. This study aimed to examine the role of nasal mucosal temperature change in the subjective perception of nasal patency and the methods by which it can be quantified. METHOD: Medline and PubMed database searches were performed to retrieve literature relevant to the topic. RESULTS: The primary mechanism producing the sensation of nasal patency is thought to be the activation of transient receptor potential melastatin family member 8 ('TRPM8'), a thermoreceptor that is activated by nasal mucosal cooling. Computational fluid dynamics studies have demonstrated that increased airflow and heat flux are correlated with better patient-reported outcome measure scores. Similarly, physical measurements of the nasal cavity using temperature probes have shown a correlation between lower nasal mucosal temperatures and better patient-reported outcome measure scores. CONCLUSION: Nasal mucosal temperature change may be correlated with the perception of improved nasal patency. Future research should quantify the impact of mucosal cooling on the perception of nasal airway obstruction.


Subject(s)
Cold Temperature/adverse effects , Nasal Mucosa/physiology , Nasal Obstruction/psychology , Perception/physiology , Airway Resistance/physiology , Computer Simulation , Humans , Hydrodynamics , Nasal Cavity/anatomy & histology , Nasal Cavity/diagnostic imaging , Nasal Cavity/physiology , Nasal Mucosa/metabolism , Nasal Obstruction/diagnosis , Nasal Obstruction/physiopathology , Nasal Obstruction/surgery , Patient Reported Outcome Measures , Pulmonary Ventilation/physiology , TRPM Cation Channels/metabolism , Temperature , Thermoreceptors/metabolism
20.
Mol Cell Endocrinol ; 518: 110986, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32835783

ABSTRACT

Thermoregulation is critical for survival and animals therefore employ strategies to keep their body temperature within a physiological range. As ectotherms, fish exclusively rely on behavioral strategies for thermoregulation. Different species of fish seek out their specific optimal temperatures through thermal navigation by biasing behavioral output based on experienced environmental temperatures. Like other vertebrates, fish sense water temperature using thermoreceptors in trigeminal and dorsal root ganglia neurons that innervate the skin. Recent research in larval zebrafish has revealed how neural circuits subsequently transform this sensation of temperature into thermoregulatory behaviors. Across fish species, thermoregulatory strategies rely on a modulation of swim vigor based on current temperature and a modulation of turning based on temperature change. Interestingly, temperature preferences are not fixed but depend on other environmental cues and internal states. The following review is intended as an overview on the current knowledge as well as open questions in fish thermoregulation.


Subject(s)
Fishes/physiology , Thermoreceptors/metabolism , Animals , Behavior, Animal , Body Temperature Regulation
SELECTION OF CITATIONS
SEARCH DETAIL