Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.464
Filter
Add more filters

Publication year range
1.
Cell ; 178(6): 1375-1386.e11, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474366

ABSTRACT

In search of the molecular identities of cold-sensing receptors, we carried out an unbiased genetic screen for cold-sensing mutants in C. elegans and isolated a mutant allele of glr-3 gene that encodes a kainate-type glutamate receptor. While glutamate receptors are best known to transmit chemical synaptic signals in the CNS, we show that GLR-3 senses cold in the peripheral sensory neuron ASER to trigger cold-avoidance behavior. GLR-3 transmits cold signals via G protein signaling independently of its glutamate-gated channel function, suggesting GLR-3 as a metabotropic cold receptor. The vertebrate GLR-3 homolog GluK2 from zebrafish, mouse, and human can all function as a cold receptor in heterologous systems. Mouse DRG sensory neurons express GluK2, and GluK2 knockdown in these neurons suppresses their sensitivity to cold but not cool temperatures. Our study identifies an evolutionarily conserved cold receptor, revealing that a central chemical receptor unexpectedly functions as a thermal receptor in the periphery.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/genetics , Receptors, Glutamate/physiology , Receptors, Kainic Acid/physiology , Receptors, Metabotropic Glutamate/physiology , Thermosensing/physiology , Animals , CHO Cells , Caenorhabditis elegans Proteins/genetics , Cold Temperature , Cricetulus , Humans , Mice , Neurons/metabolism , Receptors, Glutamate/genetics , Receptors, Kainic Acid/genetics , Receptors, Metabotropic Glutamate/genetics , Thermosensing/genetics
2.
Nature ; 628(8009): 826-834, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538787

ABSTRACT

Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.


Subject(s)
Brain Stem , Ependymoglial Cells , Feeding Behavior , Hot Temperature , Hypothalamus , Neural Pathways , Neurons , Animals , Female , Male , Mice , Agouti-Related Protein/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/cytology , Brain Stem/cytology , Brain Stem/physiology , Dopamine/metabolism , Eating/physiology , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Feeding Behavior/physiology , Glutamic Acid/metabolism , Hypothalamus/cytology , Hypothalamus/physiology , Neural Pathways/metabolism , Neurons/metabolism , Parabrachial Nucleus/cytology , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Thermosensing/physiology , Time Factors , Vascular Endothelial Growth Factor A/cerebrospinal fluid , Vascular Endothelial Growth Factor A/metabolism
3.
Nature ; 629(8014): 1126-1132, 2024 May.
Article in English | MEDLINE | ID: mdl-38750356

ABSTRACT

Plants exposed to incidences of excessive temperatures activate heat-stress responses to cope with the physiological challenge and stimulate long-term acclimation1,2. The mechanism that senses cellular temperature for inducing thermotolerance is still unclear3. Here we show that TWA1 is a temperature-sensing transcriptional co-regulator that is needed for basal and acquired thermotolerance in Arabidopsis thaliana. At elevated temperatures, TWA1 changes its conformation and allows physical interaction with JASMONATE-ASSOCIATED MYC-LIKE (JAM) transcription factors and TOPLESS (TPL) and TOPLESS-RELATED (TPR) proteins for repressor complex assembly. TWA1 is a predicted intrinsically disordered protein that has a key thermosensory role functioning through an amino-terminal highly variable region. At elevated temperatures, TWA1 accumulates in nuclear subdomains, and physical interactions with JAM2 and TPL appear to be restricted to these nuclear subdomains. The transcriptional upregulation of the heat shock transcription factor A2 (HSFA2) and heat shock proteins depended on TWA1, and TWA1 orthologues provided different temperature thresholds, consistent with the sensor function in early signalling of heat stress. The identification of the plant thermosensors offers a molecular tool for adjusting thermal acclimation responses of crops by breeding and biotechnology, and a sensitive temperature switch for thermogenetics.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Intrinsically Disordered Proteins , Temperature , Thermosensing , Thermotolerance , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation, Plant , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Repressor Proteins/metabolism , Thermosensing/genetics , Thermosensing/physiology , Thermotolerance/genetics , Thermotolerance/physiology , Transcription Factors/metabolism , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 121(25): e2322475121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857404

ABSTRACT

Low temperatures and cooling agents like menthol induce cold sensation by activating the peripheral cold receptors TRPM8 and TRPA1, cation channels belonging to the TRP channel family, while the reduction of potassium currents provides an additional and/or synergistic mechanism of cold sensation. Despite extensive studies over the past decades to identify the molecular receptors that mediate thermosensation, cold sensation is still not fully understood and many cold-sensitive peripheral neurons do not express the well-established cold sensor TRPM8. We found that the voltage-gated potassium channel KCNQ1 (Kv7.1), which is defective in cardiac LQT1 syndrome, is, in addition to its known function in the heart, a highly relevant and sex-specific sensor of moderately cold temperatures. We found that KCNQ1 is expressed in skin and dorsal root ganglion neurons, is sensitive to menthol and cooling agents, and is highly sensitive to moderately cold temperatures, in a temperature range at which TRPM8 is not thermosensitive. C-fiber recordings from KCNQ1-/- mice displayed altered action potential firing properties. Strikingly, only male KCNQ1-/- mice showed substantial deficits in cold avoidance at moderately cold temperatures, with a strength of the phenotype similar to that observed in TRPM8-/- animals. While sex-dependent differences in thermal sensitivity have been well documented in humans and mice, KCNQ1 is the first gene reported to play a role in sex-specific temperature sensation. Moreover, we propose that KCNQ1, together with TRPM8, is a key instrumentalist that orchestrates the range and intensity of cold sensation.


Subject(s)
Cold Temperature , KCNQ1 Potassium Channel , Animals , Male , Female , Mice , KCNQ1 Potassium Channel/metabolism , KCNQ1 Potassium Channel/genetics , Mice, Knockout , Ganglia, Spinal/metabolism , Thermosensing/physiology , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Mice, Inbred C57BL , Action Potentials/physiology , Sex Characteristics , Menthol/pharmacology
5.
Nature ; 583(7818): 825-829, 2020 07.
Article in English | MEDLINE | ID: mdl-32669706

ABSTRACT

Temperature is a key factor in the growth and development of all organisms1,2. Plants have to interpret temperature fluctuations, over hourly to monthly timescales, to align their growth and development with the seasons. Much is known about how plants respond to acute thermal stresses3,4, but the mechanisms that integrate long-term temperature exposure remain unknown. The slow, winter-long upregulation of VERNALIZATION INSENSITIVE 3 (VIN3)5-7, a PHD protein that functions with Polycomb repressive complex 2 to epigenetically silence FLOWERING LOCUS C (FLC) during vernalization, is central to plants interpreting winter progression5,6,8-11. Here, by a forward genetic screen, we identify two dominant mutations of the transcription factor NTL8 that constitutively activate VIN3 expression and alter the slow VIN3 cold induction profile. In the wild type, the NTL8 protein accumulates slowly in the cold, and directly upregulates VIN3 transcription. Through combining computational simulation and experimental validation, we show that a major contributor to this slow accumulation is reduced NTL8 dilution due to slow growth at low temperatures. Temperature-dependent growth is thus exploited through protein dilution to provide the long-term thermosensory information for VIN3 upregulation. Indirect mechanisms involving temperature-dependent growth, in addition to direct thermosensing, may be widely relevant in long-term biological sensing of naturally fluctuating temperatures.


Subject(s)
Arabidopsis/growth & development , Cold Temperature , Thermosensing/physiology , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MADS Domain Proteins/genetics , Models, Biological , Plant Roots/metabolism , Thermosensing/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Bioessays ; 46(7): e2400047, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769699

ABSTRACT

Recent insights reveal the significant role of TRPV3 in warmth sensation. A novel finding elucidated how thermosensation is affected by TRPV3 membrane abundance that is modulated by the transmembrane protein TMEM79. TRPV3 is a warmth-sensitive ion channel predominantly expressed in epithelial cells, particularly skin keratinocytes. Multiple studies investigated the roles of TRPV3 in cutaneous physiology and pathophysiology. TRPV3 activation by innocuous warm temperatures in keratinocytes highlights its significance in temperature sensation, but whether TRPV3 directly contributes to warmth sensations in vivo remains controversial. This review explores the electrophysiological and structural properties of TRPV3 and how modulators affect its intricate regulatory mechanisms. Moreover, we discuss the multifaceted involvement of TRPV3 in skin physiology and pathology, including barrier formation, hair growth, inflammation, and itching. Finally, we examine the potential of TRPV3 as a therapeutic target for skin diseases and highlight its diverse role in maintaining skin homeostasis.


Subject(s)
Homeostasis , Keratinocytes , Skin , TRPV Cation Channels , TRPV Cation Channels/metabolism , Humans , Animals , Skin/metabolism , Keratinocytes/metabolism , Thermosensing/physiology , Skin Diseases/metabolism , Skin Diseases/drug therapy
7.
J Neurosci ; 44(11)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38316563

ABSTRACT

Cooling sensations arise inside the mouth during ingestive and homeostasis behaviors. Oral presence of cooling temperature engages the cold and menthol receptor TRPM8 (transient receptor potential melastatin 8) on trigeminal afferents. Yet, how TRPM8 influences brain and behavioral responses to oral temperature is undefined. Here we used in vivo neurophysiology to record action potentials stimulated by cooling and warming of oral tissues from trigeminal nucleus caudalis neurons in female and male wild-type and TRPM8 gene deficient mice. Using these lines, we also measured orobehavioral licking responses to cool and warm water in a novel, temperature-controlled fluid choice test. Capture of antidromic electrophysiological responses to thalamic stimulation identified that wild-type central trigeminal neurons showed diverse responses to oral cooling. Some neurons displayed relatively strong excitation to cold <10°C (COLD neurons) while others responded to only a segment of mild cool temperatures below 30°C (COOL neurons). Notably, TRPM8 deficient mice retained COLD-type but lacked COOL cells. This deficit impaired population responses to mild cooling temperatures below 30°C and allowed warmth-like (≥35°C) neural activity to pervade the normally innocuous cool temperature range, predicting TRPM8 deficient mice would show anomalously similar orobehavioral responses to warm and cool temperatures. Accordingly, TRPM8 deficient mice avoided both warm (35°C) and mild cool (≤30°C) water and sought colder temperatures in fluid licking tests, whereas control mice avoided warm but were indifferent to mild cool and colder water. Results imply TRPM8 input separates cool from warm temperature sensing and suggest other thermoreceptors also participate in oral cooling sensation.


Subject(s)
TRPM Cation Channels , Mice , Male , Animals , Female , TRPM Cation Channels/genetics , Cold Temperature , Neurons , Temperature , Thermosensing/physiology , Water
8.
Physiology (Bethesda) ; 39(4): 0, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38536114

ABSTRACT

Whether it is the dramatic suffocating sensation from a heat wave in the summer or the positive reinforcement arising from a hot drink on a cold day; we can certainly agree that our thermal environment underlies our daily rhythms of sensation. Extensive research has focused on deciphering the central circuits responsible for conveying the impact of thermogenesis on mammalian behavior. Here, we revise the recent literature responsible for defining the behavioral correlates that arise from thermogenic fluctuations in mammals. We transition from the physiological significance of thermosensation to the circuitry responsible for the autonomic or behavioral responses associated with it. Subsequently, we delve into the positive and negative valence encoded by thermoregulatory processes. Importantly, we emphasize the crucial junctures where reward, pain, and thermoregulation intersect, unveiling a complex interplay within these neural circuits. Finally, we briefly outline fundamental questions that are pending to be addressed in the field. Fully deciphering the thermoregulatory circuitry in mammals will have far-reaching medical implications. For instance, it may lead to the identification of novel targets to overcome thermal pain or allow the maintenance of our core temperature in prolonged surgeries.


Subject(s)
Body Temperature Regulation , Brain , Cues , Thermosensing , Humans , Animals , Thermosensing/physiology , Brain/physiology , Body Temperature Regulation/physiology , Pain/physiopathology , Thermogenesis/physiology
9.
Proc Natl Acad Sci U S A ; 119(21): e2201349119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35594403

ABSTRACT

To cope with temperature fluctuations, molecular thermosensors in animals play a pivotal role in accurately sensing ambient temperature. Transient receptor potential melastatin 8 (TRPM8) is the most established cold sensor. In order to understand how the evolutionary forces bestowed TRPM8 with cold sensitivity, insights into both emergence of cold sensing during evolution and the thermodynamic basis of cold activation are needed. Here, we show that the trpm8 gene evolved by forming and regulating two domains (MHR1-3 and pore domains), thus determining distinct cold-sensitive properties among vertebrate TRPM8 orthologs. The young trpm8 gene without function can be observed in the closest living relatives of tetrapods (lobe-finned fishes), while the mature MHR1-3 domain with independent cold sensitivity has formed in TRPM8s of amphibians and reptiles to enable channel activation by cold. Furthermore, positive selection in the TRPM8 pore domain that tuned the efficacy of cold activation appeared late among more advanced terrestrial tetrapods. Interestingly, the mature MHR1-3 domain is necessary for the regulatory mechanism of the pore domain in TRPM8 cold activation. Our results reveal the domain-based evolution for TRPM8 functions and suggest that the acquisition of cold sensitivity in TRPM8 facilitated terrestrial adaptation during the water-to-land transition.


Subject(s)
TRPM Cation Channels , Transient Receptor Potential Channels , Cold Temperature , TRPM Cation Channels/chemistry , TRPM Cation Channels/genetics , Thermosensing/physiology
10.
Annu Rev Physiol ; 83: 205-230, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33085927

ABSTRACT

Temperature is a universal cue and regulates many essential processes ranging from enzymatic reactions to species migration. Due to the profound impact of temperature on physiology and behavior, animals and humans have evolved sophisticated mechanisms to detect temperature changes. Studies from animal models, such as mouse, Drosophila, and C. elegans, have revealed many exciting principles of thermosensation. For example, conserved molecular thermosensors, including thermosensitive channels and receptors, act as the initial detectors of temperature changes across taxa. Additionally, thermosensory neurons and circuits in different species appear to adopt similar logic to transduce and process temperature information. Here, we present the current understanding of thermosensation at the molecular and cellular levels. We also discuss the fundamental coding strategies of thermosensation at the circuit level. A thorough understanding of thermosensation not only provides key insights into sensory biology but also builds a foundation for developing better treatments for various sensory disorders.


Subject(s)
Neurons/physiology , Thermosensing/physiology , Animals , Humans , Temperature
11.
Biophys J ; 123(8): 947-956, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38449311

ABSTRACT

The ability to perceive temperature is crucial for most animals. It enables them to maintain their body temperature and swiftly react to noxiously cold or hot objects. Caenorhabditis elegans is a powerful genetic model for the study of thermosensation as its simple nervous system is well characterized and its transparent body is suited for in vivo functional imaging of neurons. The behavior triggered by experience-dependent thermosensation has been well studied in C. elegans under temperature-gradient environments. However, how C. elegans senses temperature via its nervous system is not well understood due to the limitations of currently available technologies. One major bottleneck is the difficulty in creating fast temperature changes, especially cold stimuli. Here, we developed a microfluidic-based platform that allowed the in vivo functional imaging of C. elegans responding to well-controlled temporally varying temperature stimulation by rapidly switching fluid streams at different temperatures. We used computational models to enable rational design and optimization of experimental conditions. We validated the design and utility of our system with studies of the functional role of thermosensory neurons. We showed that the responses of PVD polymodal nociceptor neurons observed in previous studies can be recapitulated. Further, we highlighted how this platform may be used to dissect neuronal circuits with an example of activity recording in PVC interneurons. Both of these neuron types show sensitization phenotypes. We envision that both the engineered system and the findings in this work will spur further studies of molecular and cellular mechanisms underlying cold-sensing through the nervous system.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Temperature , Caenorhabditis elegans/genetics , Microfluidics , Thermosensing/physiology , Cold Temperature , Caenorhabditis elegans Proteins/genetics
12.
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
13.
Nature ; 555(7698): 662-666, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29539642

ABSTRACT

Acute pain represents a crucial alarm signal to protect us from injury. Whereas the nociceptive neurons that convey pain signals were described more than a century ago, the molecular sensors that detect noxious thermal or mechanical insults have yet to be fully identified. Here we show that acute noxious heat sensing in mice depends on a triad of transient receptor potential (TRP) ion channels: TRPM3, TRPV1, and TRPA1. We found that robust somatosensory heat responsiveness at the cellular and behavioural levels is observed only if at least one of these TRP channels is functional. However, combined genetic or pharmacological elimination of all three channels largely and selectively prevents heat responses in both isolated sensory neurons and rapidly firing C and Aδ sensory nerve fibres that innervate the skin. Strikingly, Trpv1-/-Trpm3-/-Trpa1-/- triple knockout (TKO) mice lack the acute withdrawal response to noxious heat that is necessary to avoid burn injury, while showing normal nociceptive responses to cold or mechanical stimuli and a preserved preference for moderate temperatures. These findings indicate that the initiation of the acute heat-evoked pain response in sensory nerve endings relies on three functionally redundant TRP channels, representing a fault-tolerant mechanism to avoid burn injury.


Subject(s)
Hot Temperature/adverse effects , Nociceptive Pain/physiopathology , TRPA1 Cation Channel/metabolism , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Thermosensing/physiology , Animals , Burns/physiopathology , Burns/prevention & control , Cold Temperature/adverse effects , Female , Male , Mice , Mice, Knockout , Nerve Endings/physiology , Nerve Fibers/physiology , Nociception/physiology , Sensory Receptor Cells/physiology , Skin/innervation , Skin/physiopathology , TRPA1 Cation Channel/deficiency , TRPA1 Cation Channel/genetics , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Thermosensing/genetics
14.
Int J Biometeorol ; 68(2): 289-304, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38047941

ABSTRACT

Passenger thermal comfort in high-speed train (HST) carriages presents unique challenges due to factors such as extensive operational areas, longer travel durations, larger spaces, and higher passenger capacities. This study aims to propose a new prediction model to better understand and address thermal comfort in HST carriages. The proposed prediction model incorporates skin wettedness, vertical skin temperature difference (ΔTd), and skin temperature as parameters to predict the thermal sensation vote (TSV) of HST passengers. The experiments were conducted with 65 subjects, evenly distributed throughout the HST compartment. Thermal environmental conditions and physiological signals were measured to capture the subjects' thermal responses. The study also investigated regional and overall thermal sensations experienced by the subjects. Results revealed significant regional differences in skin temperature between upper and lower body parts. By analyzing data from 45 subjects, We analyzed the effect of 25 variables on TSV by partial least squares (PLS), from which we singled out 3 key factors. And the optimal multiple regression equation was derived to predict the TSV of HST occupants. Validation with an additional 20 subjects demonstrated a strong linear correlation (0.965) between the actual TSV and the predicted values, confirming the feasibility and accuracy of the developed prediction model. By integrating skin wettedness and ΔTd with skin temperature, the model provides a comprehensive approach to predicting thermal comfort in HST environments. This research contributes to advancing thermal comfort analysis in HST and offers valuable insights for optimizing HST system design and operation to meet passengers' comfort requirements.


Subject(s)
Air Conditioning , Skin Temperature , Humans , Air Conditioning/methods , Thermosensing/physiology , Temperature
15.
Proc Natl Acad Sci U S A ; 117(11): 6178-6188, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123108

ABSTRACT

The nervous system evaluates environmental cues and adjusts motor output to ensure navigation toward a preferred environment. The nematode Caenorhabditis elegans navigates in the thermal environment and migrates toward its cultivation temperature by moving up or down thermal gradients depending not only on absolute temperature but on relative difference between current and previously experienced cultivation temperature. Although previous studies showed that such thermal context-dependent opposing migration is mediated by bias in frequency and direction of reorientation behavior, the complete neural pathways-from sensory to motor neurons-and their circuit logics underlying the opposing behavioral bias remain elusive. By conducting comprehensive cell ablation, high-resolution behavioral analyses, and computational modeling, we identified multiple neural pathways regulating behavioral components important for thermotaxis, and demonstrate that distinct sets of neurons are required for opposing bias of even single behavioral components. Furthermore, our imaging analyses show that the context-dependent operation is evident in sensory neurons, very early in the neural pathway, and manifested by bidirectional responses of a first-layer interneuron AIB under different thermal contexts. Our results suggest that the contextual differences are encoded among sensory neurons and a first-layer interneuron, processed among different downstream neurons, and lead to the flexible execution of context-dependent behavior.


Subject(s)
Behavior, Animal/physiology , Caenorhabditis elegans/physiology , Interneurons/physiology , Spatial Navigation/physiology , Thermoreceptors/physiology , Animals , Behavior Observation Techniques , Locomotion/physiology , Neural Pathways/physiology , Temperature , Thermosensing/physiology
16.
Int J Biometeorol ; 67(12): 1957-1964, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37833565

ABSTRACT

The ClimApp smartphone application was developed to merge meteorological forecast data with personal information for individualized and improved thermal warning during heat and cold stress and for indoor comfort in buildings. For cold environments, ClimApp predicts the personal thermal stress and strain by the use of the Insulation REQuired model that combines weather and personal physiological data with additional consideration of the Wind Chill index based on the local weather forecast. In this study, we validated the individualized ClimApp index relative to measurements and compared it with the Universal Temperature Climate Index (UTCI). To this aim, 55 participants (27 females) were exposed to at least 1 h in an outdoor environment of 10 °C or below (average 1.4 °C air temperature, 74.9% relative humidity, and 4.7 m/s air velocity) inputting their activity level and clothing insulation as instructed by ClimApp. The UTCI and ClimApp indices were calculated and compared to the participants' perceived thermal sensation. The ClimApp index root mean square deviation (RMSD) was below the standard deviation of the perceived thermal sensation which indicates a valid prediction and the UTCI RMSD was higher than the standard deviation which indicates an invalid prediction. The correlation of ClimApp and UTCI to the perceived thermal sensation was statistically significant for both models.


Subject(s)
Climate , Smartphone , Female , Humans , Temperature , Weather , Wind , Thermosensing/physiology
17.
J Therm Biol ; 112: 103447, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36796902

ABSTRACT

Temperature step change is the typical transient thermal environment. The purpose of this study was to explore the association of subjective and objective parameters in a step-change environment, including thermal sensation vote (TSV), thermal comfort vote (TCV), mean skin temperature (MST) and endogenous dopamine (DA). Three temperature step changes defined as I3 (15 °C-18 °C to 15 °C), I9 (15 °C-24 °C to 15 °C) and I15 (15 °C-30 °C to 15 °C) were designed for this experiment. Eight male and eight female healthy subjects who participated in the experiment reported thermal perception (TSV and TCV). Skin temperatures of six body parts and DA were measured. Results show that the inverted U-shaped in TSV and TCV was deviated by seasonal factors of the experiment. The deviation direction of TSV in winter was to the warm sensation side, which was opposite to the inherent cold and hot impression of people in winter and summer. The association between dimensionless dopamine (DA*), TSV and MST were described as follows: DA* was the U-shaped change with exposure times when MST was not greater than 31 °C, and TSV was at -2 and -1, and DA* increased with exposure times when MST was greater than 31 °C, and TSV was at 0, 1 and 2. The changes in the body heat storage and autonomous thermal regulation under temperature step changes may potentially be related to the concentration of DA. The human state on thermal nonequilibrium and stronger thermal regulation would correspond to a higher concentration of DA. This work is conducive to exploring the human regulation mechanism in a transient environment.


Subject(s)
Dopamine , Skin Temperature , Humans , Male , Female , Seasons , Cold Temperature , Hot Temperature , Thermosensing/physiology , Temperature
18.
J Therm Biol ; 112: 103488, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36796929

ABSTRACT

When exposed to ambient temperatures that cause thermal discomfort, a human's behavioral responses are more effective than autonomic ones at compensating for thermal imbalance. These behavioral thermal responses are typically directed by an individual's perception of the thermal environment. Perception of the environment is a holistic amalgamation of human senses, and in some circumstances, humans prioritize visual information. Existing research has considered this in the specific case of thermal perception, and this review investigates the state of the literature examining this effect. We identify the frameworks, research rationales, and potential mechanisms that underpin the evidence base in this area. Our review identified 31 experiments, comprising 1392 participants that met the inclusion criteria. Methodological heterogeneity was observed in the assessment of thermal perception, and a variety of methods were employed to manipulate the visual environment. However, the majority of the included experiments (80%) reported a difference in thermal perception after the visual environment was manipulated. There was limited research exploring any effects on physiological variables (e.g. skin and core temperature). This review has wide-ranging implications for the broad discipline of (thermo)physiology, psychology, psychophysiology, neuroscience, ergonomics, and behavior.


Subject(s)
Body Temperature Regulation , Perception , Humans , Body Temperature Regulation/physiology , Skin , Thermosensing/physiology , Autonomic Nervous System
19.
J Therm Biol ; 112: 103430, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36796887

ABSTRACT

BACKGROUND: Severe thermal discomfort may increase risk of drowning due to hypothermia or hyperthermia from prolonged exposure to noxious water temperatures. The importance of using a behavioral thermoregulation model with thermal sensation may predict the thermal load that the human body receives when exposed to various immersive water conditions. However, there is no thermal sensation "gold standard" model specific for water immersion. This scoping review aims to present a comprehensive overview regarding human physiological and behavioral thermoregulation during whole-body water immersion and explore the feasibility for an accepted defined sensation scale for cold and hot water immersion. METHODS: A standard literary search was performed on PubMed, Google Scholar, and SCOPUS. The words "Water Immersion," "Thermoregulation," "Cardiovascular responses" were used either as independent searched terms and MeSH terms (Medical Subject Headings) or in combination with other text words. The inclusion criteria for clinical trials terms to thermoregulatory measurements (core or skin temperature), whole-body immersion, 18-60 years old and healthy individuals. The prementioned data were analyzed narratively to achieve the overall study objective. RESULTS: Twenty-three published articles fulfilled the review inclusion/exclusion criteria (with nine measured behavioral responses). Our outcomes illustrated a homogenous thermal sensation in a variety of water temperatures ranges, that was strongly associated with thermal balance, and observed different thermoregulatory responses. This scoping review highlights the impact of water immersion duration on human thermoneutral zone, thermal comfort zone, and thermal sensation. CONCLUSION: Our findings enlighten the significance of thermal sensation as a health indicator for establishing a behavioral thermal model applicable for water immersion. This scoping review provides insight for the needed development of subjective thermal model of thermal sensation in relation to human thermal physiology specific to immersive water temperature ranges within and outside the thermal neutral and comfort zone.


Subject(s)
Immersion , Water , Humans , Adolescent , Young Adult , Adult , Middle Aged , Temperature , Body Temperature Regulation/physiology , Thermosensing/physiology , Cold Temperature , Skin Temperature
20.
J Therm Biol ; 114: 103567, 2023 May.
Article in English | MEDLINE | ID: mdl-37209632

ABSTRACT

In this study, we explored the association between physiological and perceptual heat strain while wearing stab-resistant body armor (SRBA). Human trials were performed on ten participants in warm and hot environments. Physiological responses (core temperature, skin temperature, and heart rate), and perceptual responses (thermal sensation vote, thermal comfort vote, restriction of perceived exertion (RPE), wetness of skin, and wetness of clothing) were recorded throughout the trials, and subsequently, the physiological strain index (PSI), and perceptual strain index (PeSI) were calculated. The results indicated that the PeSI showed a significant moderate association with the PSI, and was capable of predicting PSI for low (PSI = 3) and high (PSI = 7) levels of physiological strain with the areas under the curves of 0.80 and 0.64, respectively. Moreover, Bland-Altman analysis indicated that the majority of the PSI ranged within the 95% confidence interval, and the mean difference between PSI and PeSI was 0.14 ± 2.02 with the lower 95% limit and upper 95% limit being -3.82 to 4.10, respectively. Therefore, the subjective responses could be used as an indicator for predicting physiological strain while wearing SRBA. This study could provide fundamental knowledge for the usage of SRBA, and the development of physiological heat strain assessment.


Subject(s)
Body Temperature , Hot Temperature , Humans , Body Temperature/physiology , Skin Temperature , Heart Rate/physiology , Thermosensing/physiology , Body Temperature Regulation , Protective Clothing
SELECTION OF CITATIONS
SEARCH DETAIL