Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.028
Filtrar
Más filtros

Intervalo de año de publicación
1.
Physiol Rev ; 100(2): 725-803, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31670612

RESUMEN

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.


Asunto(s)
Señalización del Calcio , Mecanotransducción Celular , Nocicepción , Células Receptoras Sensoriales/metabolismo , Canal Catiónico TRPA1/metabolismo , Sensación Térmica , Animales , Canalopatías/metabolismo , Canalopatías/fisiopatología , Células Quimiorreceptoras/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Mecanorreceptores/metabolismo , Nociceptores/metabolismo , Dolor/metabolismo , Dolor/fisiopatología , Termorreceptores/metabolismo
2.
J Neurophysiol ; 132(2): 544-569, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38985936

RESUMEN

Wide-range thermoreceptive neurons (WRT-EN) in monkey cortical area 7b that encoded innocuous and nocuous cutaneous thermal and threatening visuosensory stimulation with high fidelity were studied to identify their multisensory integrative response properties. Emphasis was given to characterizing the spatial and temporal effects of threatening visuosensory input on the thermal stimulus-response properties of these multisensory nociceptive neurons. Threatening visuosensory stimulation was most efficacious in modulating thermal evoked responses when presented as a downward ("looming"), spatially congruent, approaching and closely proximal target in relation to the somatosensory receptive field. Both temporal alignment and misalignment of spatially aligned threatening visual and thermal stimulation significantly increased mean discharge frequencies above those evoked by thermal stimulation alone, particularly at near noxious (43°C) and mildly noxious (45°C) temperatures. The enhanced multisensory discharge frequencies were equivalent to the discharge frequency evoked by overtly noxious thermal stimulation alone at 47°C (monkey pain tolerance threshold). A significant increase in behavioral mean escape frequency with shorter escape latency was evoked by multisensory stimulation at near noxious temperature (43°C), which was equivalent to that evoked by noxious stimulation alone (47°C). The remarkable concordance of elevating both neural discharge and escape frequency from a nonnociceptive and prepain level by near noxious thermal stimulation to a nociceptive and pain level by multisensory visual and near noxious thermal stimulation and integration is an elegantly designed defensive neural mechanism that in effect lowers both nociceptive response and pain thresholds to preemptively engage nocifensive behavior and, consequently, avert impending and actual injurious noxious thermal stimulation.NEW & NOTEWORTHY Multisensory nociceptive neurons in cortical area 7b are engaged in integration of threatening visuosensory and a wide range of innocuous and nocuous somatosensory (thermoreceptive) inputs. The enhancement of neuronal activity and escape behavior in monkey by multisensory integration is consistent and supportive of human psychophysical studies. The spatial features of visuosensory stimulation in peripersonal space in relation to somatic stimulation in personal space are critical to multisensory integration, nociception, nocifensive behavior, and pain.


Asunto(s)
Macaca mulatta , Nociceptores , Animales , Nociceptores/fisiología , Masculino , Nocicepción/fisiología , Calor , Percepción Visual/fisiología , Umbral del Dolor/fisiología , Estimulación Luminosa , Reacción de Fuga/fisiología , Termorreceptores/fisiología
3.
Nature ; 555(7694): 98-102, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29466329

RESUMEN

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.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Drosophila melanogaster/fisiología , Neuronas/fisiología , Sueño/fisiología , Temperatura , Sensación Térmica/fisiología , Animales , Frío , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/citología , Femenino , Calor , Locomoción/fisiología , Masculino , Red Nerviosa/citología , Red Nerviosa/fisiología , Inhibición Neural , Termorreceptores/metabolismo , Factores de Tiempo
4.
Proc Natl Acad Sci U S A ; 117(11): 6178-6188, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123108

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Caenorhabditis elegans/fisiología , Interneuronas/fisiología , Navegación Espacial/fisiología , Termorreceptores/fisiología , Animales , Técnicas de Observación Conductual , Locomoción/fisiología , Vías Nerviosas/fisiología , Temperatura , Sensación Térmica/fisiología
5.
Sensors (Basel) ; 23(13)2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37447667

RESUMEN

Pyroelectric infrared sensors (PIR) are widely used as infrared (IR) detectors due to their basic implementation, low cost, low power, and performance. Combined with a Fresnel lens, they can be used as a binary detector in applications of presence and motion control. Furthermore, due to their features, they can be used in autonomous intelligent devices or included in robotics applications or sensor networks. In this work, two neural processing architectures are presented: (1) an analog processing approach to achieve the behavior of a presynaptic neuron from a PIR sensor. An analog circuit similar to the leaky integrate and fire model is implemented to be able to generate spiking rates proportional to the IR stimuli received at a PIR sensor. (2) An embedded postsynaptic neuron where a spiking neural network matrix together with an algorithm based on digital processing techniques is introduced. This structure allows connecting a set of sensors to the post-synaptic circuit emulating an optic nerve. As a case study, the entire neural processing approach presented in this paper is applied to optical flow detection considering a four-PIR array as input. The results validate both the spiking approach for an analog sensor presented and the ability to retrieve the analog information sent as spike trains in a simulated optic nerve.


Asunto(s)
Neuronas , Termorreceptores , Neuronas/fisiología , Movimiento/fisiología , Redes Neurales de la Computación , Algoritmos
6.
Int J Mol Sci ; 24(16)2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37629206

RESUMEN

Cyclosporine A (CsA) is used for the treatment of dry eye (DE) with good clinical results, improving tear secretion and decreasing subjective symptoms. These effects are attributed to the improved tear film dynamics, but there are no data on the effect of CsA on the abnormal sensory nerve activity characteristic in DE. Our purpose was to evaluate the CsA effect on the enhanced activity of corneal cold thermoreceptors in a tear-deficient DE animal model using in vitro extracellular recording of cold thermoreceptors nerve terminal impulses (NTIs) before and in the presence of CsA. NTI shape was also analyzed. Blinking frequency and tearing rate were also measured in awake animals before and after topical CsA. CsA increased the tearing and blinking of treated animals. CsA significantly decreased the peak response to cold of cold thermoreceptors. Neither their spontaneous NTIs discharge rate nor their cooling threshold were modified. CsA also seemed to reverse some of the changes in NTI shape induced by tear deficiency. These data suggest that, at least in part, the beneficial clinical effects of CsA in DE can be attributed to a direct effect on sensory nerve endings, although the precise mechanisms underlying this effect need further studies to be fully clarified.


Asunto(s)
Ciclosporina , Enfermedades del Aparato Lagrimal , Animales , Ciclosporina/farmacología , Terminaciones Nerviosas , Termorreceptores , Células Receptoras Sensoriales , Potenciales de Acción
7.
Mol Microbiol ; 113(3): 588-592, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31971637

RESUMEN

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.


Asunto(s)
Bacterias/metabolismo , Interacciones Microbiota-Huesped/fisiología , Termorreceptores/fisiología , Bacterias/patogenicidad , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Calor , Listeria monocytogenes/metabolismo , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , ARN/genética , ARN/metabolismo , Termorreceptores/metabolismo , Sensación Térmica/genética , Sensación Térmica/fisiología , Transactivadores/metabolismo , Virulencia/genética , Factores de Virulencia/genética
8.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R91-R99, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34075801

RESUMEN

Human thermoregulatory control is often evaluated through the relationship between thermoeffector output and core or mean body temperature. In addition to providing a general indication of whether a variable of interest alters thermoregulatory control, this relationship is often used to determine how this alteration may occur. This latter interpretation relies upon two parameters of the thermoeffector output-body temperature relationship: the onset threshold and thermosensitivity. Traditionally, changes in the onset threshold and thermosensitivity are interpreted as "central" or "peripheral" modulation of thermoregulatory control, respectively. This mini-review revisits the origins of the thermoeffector output-body temperature relationship and its use to interpret "central" or "peripheral" modulation of thermoregulatory control. Against this background, we discuss the strengths and weaknesses of this approach and highlight that "central" thermoregulatory control reflects the neural control of body temperature whereas "peripheral" thermoregulatory control reflects properties specific to the thermoeffector organs. We highlight studies that employed more direct approaches to investigate the neural control of body temperature and peripheral properties of thermoeffector organs. We conclude by encouraging future investigations interested in studying thermoregulatory control to more directly investigate the component of the thermoeffector loop under investigation.heat; human; skin blood flow; sweat; thermoregulatory.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Vasos Sanguíneos/inervación , Encéfalo/fisiología , Temperatura Cutánea , Piel/irrigación sanguínea , Sudoración , Termorreceptores/fisiología , Vasodilatación , Humanos , Modelos Biológicos , Flujo Sanguíneo Regional , Sensación Térmica
9.
Nature ; 519(7543): 353-7, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25739502

RESUMEN

In Drosophila, just as in vertebrates, changes in external temperature are encoded by bidirectional opponent thermoreceptor cells: some cells are excited by warming and inhibited by cooling, whereas others are excited by cooling and inhibited by warming. The central circuits that process these signals are not understood. In Drosophila, a specific brain region receives input from thermoreceptor cells. Here we show that distinct genetically identified projection neurons (PNs) in this brain region are excited by cooling, warming, or both. The PNs excited by cooling receive mainly feed-forward excitation from cool thermoreceptors. In contrast, the PNs excited by warming ('warm-PNs') receive both excitation from warm thermoreceptors and crossover inhibition from cool thermoreceptors through inhibitory interneurons. Notably, this crossover inhibition elicits warming-evoked excitation, because warming suppresses tonic activity in cool thermoreceptors. This in turn disinhibits warm-PNs and sums with feed-forward excitation evoked by warming. Crossover inhibition could cancel non-thermal activity (noise) that is positively correlated among warm and cool thermoreceptor cells, while reinforcing thermal activity which is anti-correlated. Our results show how central circuits can combine signals from bidirectional opponent neurons to construct sensitive and robust neural codes.


Asunto(s)
Encéfalo/citología , Encéfalo/fisiología , Drosophila melanogaster/fisiología , Temperatura , Termorreceptores/fisiología , Sensación Térmica/fisiología , Animales , Drosophila melanogaster/citología , Femenino , Interneuronas/fisiología
10.
Nature ; 519(7543): 358-61, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25739506

RESUMEN

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.


Asunto(s)
Encéfalo/fisiología , Drosophila melanogaster/fisiología , Vías Nerviosas , Temperatura , Sensación Térmica/fisiología , Animales , Encéfalo/anatomía & histología , Encéfalo/citología , Mapeo Encefálico , Calcio/análisis , Calcio/metabolismo , Drosophila melanogaster/citología , Cuerpos Pedunculados/inervación , Neuronas/metabolismo , Sinapsis/metabolismo , Termorreceptores/metabolismo , Factores de Tiempo
11.
Indoor Air ; 31(6): 2266-2280, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34048603

RESUMEN

Research into human thermal perception indoors has focused on "neutrality" under steady-state conditions. Recent interest in thermal alliesthesia has highlighted the hedonic dimension of our thermal world that has been largely overlooked by science. Here, we show the activity of sensory neurons can predict thermal pleasure under dynamic exposures. A numerical model of cutaneous thermoreceptors was applied to skin temperature measurements from 12 human subjects. A random forest model trained on simulated thermoreceptor impulses could classify pleasure responses (F1 score of 67%) with low false positives/negatives (4%). Accuracy increased (83%) when excluding the few extreme (dis)pleasure responses. Validation on an independent dataset confirmed model reliability. This is the first empirical demonstration of the relationship between thermoreceptors and pleasure arising from thermal stimuli. Insights into the neurophysiology of thermal perception can enhance the experience of built environments through designs that promote sensory excitation instead of neutrality.


Asunto(s)
Contaminación del Aire Interior , Termorreceptores , Humanos , Placer , Reproducibilidad de los Resultados , Temperatura Cutánea
12.
Adv Exp Med Biol ; 1349: 355-370, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35138622

RESUMEN

Ambient temperature detection and core body temperature maintenance are critical for the environment adaptability of mammals, requiring an elaborate neural network that converts the temperature information sensed by thermoreceptors into physiological and behavioral thermoregulatory responses. The molecular basis of thermosensation lies in the activation of various thermosensitive ion channels with distinct temperature thresholds expressed on the cell membrane of sensory neurons. These channels are able to convert thermal stimuli into electrical activities by gating ions into and out of the cell. In this chapter, we briefly introduce the physiological functions of the main thermosensitive ion channels involved in the core body temperature homeostasis orchestrated by the neural circuits in the peripheral and central nerve systems.


Asunto(s)
Regulación de la Temperatura Corporal , Termorreceptores , Animales , Iones , Mamíferos , Neuronas Aferentes
13.
J Neurosci ; 39(41): 8177-8192, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31471469

RESUMEN

The cornea is extensively innervated by trigeminal ganglion cold thermoreceptor neurons expressing TRPM8 (transient receptor potential cation channel subfamily M member 8). These neurons respond to cooling, hyperosmolarity and wetness of the corneal surface. Surgical injury of corneal nerve fibers alters tear production and often causes dry eye sensation. The contribution of TRPM8-expressing corneal cold-sensitive neurons (CCSNs) to these symptoms is unclear. Using extracellular recording of CCSNs nerve terminals combined with in vivo confocal tracking of reinnervation, Ca2+ imaging and patch-clamp recordings of fluorescent retrogradely labeled corneal neurons in culture, we analyzed the functional modifications of CCSNs induced by peripheral axonal damage in male mice. After injury, the percentage of CCSNs, the cold- and menthol-evoked intracellular [Ca2+] rises and the TRPM8 current density in CCSNs were larger than in sham animals, with no differences in the brake K+ current IKD Active and passive membrane properties of CCSNs from both groups were alike and corresponded mainly to those of canonical low- and high-threshold cold thermoreceptor neurons. Ongoing firing activity and menthol sensitivity were higher in CCSN terminals of injured mice, an observation accounted for by mathematical modeling. These functional changes developed in parallel with a partial reinnervation of the cornea by TRPM8(+) fibers and with an increase in basal tearing in injured animals compared with sham mice. Our results unveil key TRPM8-dependent functional changes in CCSNs in response to injury, suggesting that increased tearing rate and ocular dryness sensation derived from deep surgical ablation of corneal nerves are due to enhanced functional expression of TRPM8 channels in these injured trigeminal primary sensory neurons.SIGNIFICANCE STATEMENT We unveil a key role of TRPM8 channels in the sensory and autonomic disturbances associated with surgical damage of eye surface nerves. We studied the damage-induced functional alterations of corneal cold-sensitive neurons using confocal tracking of reinnervation, extracellular corneal nerve terminal recordings, tearing measurements in vivo, Ca2+ imaging and patch-clamp recordings of cultured corneal neurons, and mathematical modeling. Corneal nerve ablation upregulates TRPM8 mainly in canonical cold thermoreceptors, enhancing their cold and menthol sensitivity, inducing a rise in the ongoing firing activity of TRPM8(+) nerve endings and an increase in basal tearing. Our results suggest that unpleasant dryness sensations, together with augmented tearing rate after corneal nerve injury, are largely due to upregulation of TRPM8 in cold thermoreceptor neurons.


Asunto(s)
Axones/fisiología , Frío , Córnea/inervación , Córnea/fisiología , Células Receptoras Sensoriales/fisiología , Canales Catiónicos TRPM/fisiología , Sensación Térmica/fisiología , Animales , Lesiones de la Cornea/fisiopatología , Fenómenos Electrofisiológicos , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Neurológicos , Modelos Teóricos , Fibras Nerviosas , Técnicas de Placa-Clamp , Lágrimas , Termorreceptores/fisiología
14.
J Neurosci ; 39(6): 949-969, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30545944

RESUMEN

TRPM8 is a polymodal, nonselective cation channel activated by cold temperature and cooling agents that plays a critical role in the detection of environmental cold. We found that TRPM8 is a pharmacological target of tacrolimus (FK506), a macrolide immunosuppressant with several clinical uses, including the treatment of organ rejection following transplants, treatment of atopic dermatitis, and dry eye disease. Tacrolimus is an inhibitor of the phosphatase calcineurin, an action shared with cyclosporine. Tacrolimus activates TRPM8 channels in different species, including humans, and sensitizes their response to cold temperature by inducing a leftward shift in the voltage-dependent activation curve. The effects of tacrolimus on purified TRPM8 in lipid bilayers demonstrates conclusively that it has a direct gating effect. Moreover, the lack of effect of cyclosporine rules out the canonical signaling pathway involving the phosphatase calcineurin. Menthol (TRPM8-Y745H)- and icilin (TRPM8-N799A)-insensitive mutants were also activated by tacrolimus, suggesting a different binding site. In cultured mouse DRG neurons, tacrolimus evokes an increase in intracellular calcium almost exclusively in cold-sensitive neurons, and these responses were drastically blunted in Trpm8 KO mice or after the application of TRPM8 antagonists. Cutaneous and corneal cold thermoreceptor endings are also activated by tacrolimus, and tacrolimus solutions trigger blinking and cold-evoked behaviors. Together, our results identify TRPM8 channels in sensory neurons as molecular targets of the immunosuppressant tacrolimus. The actions of tacrolimus on TRPM8 resemble those of menthol but likely involve interactions with other channel residues.SIGNIFICANCE STATEMENT TRPM8 is a polymodal TRP channel involved in cold temperature sensing, thermoregulation, and cold pain. TRPM8 is also involved in the pathophysiology of dry eye disease, and TRPM8 activation has antiallodynic and antipruritic effects, making it a prime therapeutic target in several cutaneous and neural diseases. We report the direct agonist effect of tacrolimus, a potent natural immunosuppressant with multiple clinical applications, on TRPM8 activity. This interaction represents a novel neuroimmune interface. The identification of a clinically approved drug with agonist activity on TRPM8 channels could be used experimentally to probe the function of TRPM8 in humans. Our findings may explain some of the sensory and anti-inflammatory effects described for this drug in the skin and the eye surface.


Asunto(s)
Inmunosupresores/farmacología , Canales Catiónicos TRPM/agonistas , Tacrolimus/farmacología , Animales , Conducta Animal/efectos de los fármacos , Células Cultivadas , Frío , Femenino , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Células HEK293 , Humanos , Membrana Dobles de Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Células Receptoras Sensoriales/efectos de los fármacos , Canales Catiónicos TRPM/genética , Termorreceptores/efectos de los fármacos
15.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R579-R589, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31967850

RESUMEN

In contrast to other species, humans are believed to lack hygroreceptors for sensing skin wetness. Yet, the molecular basis of human hygrosensation is currently unknown, and it remains unclear whether we possess a receptor-mediated sensing mechanism for skin wetness. The aim of this study was to assess the role of the cutaneous cold-sensitive transient receptor potential melastatin-8 (TRPM8) channel as a molecular mediator of human hygrosensation. To this end, we exploited both the thermal and chemical activation of TRPM8-expressing cutaneous Aδ cold thermoreceptors, and we assessed wetness sensing in healthy young men in response to 1) dry skin cooling in the TRPM8 range of thermosensitivity and 2) application of the TRPM8 agonist menthol. Our results indicate that 1) independently of contact with moisture, a cold-dry stimulus in the TRPM8 range of activation induced wetness perceptions across 12 different body regions and those wetness perceptions varied across the body following regional differences in cold sensitivity; and 2) independently of skin cooling, menthol-induced stimulation of TRPM8 triggered wetness perceptions that were greater than those induced by physical dry cooling and by contact with an aqueous cream containing actual moisture. For the first time, we show that the cutaneous cold-sensing TRPM8 channel plays the dual role of cold and wetness sensor in human skin and that this ion channel is a peripheral mediator of human skin wetness perception.


Asunto(s)
Frío , Temperatura Cutánea/fisiología , Canales Catiónicos TRPM/metabolismo , Termorreceptores/fisiología , Frío/efectos adversos , Ganglios Espinales/fisiología , Humanos , Células Receptoras Sensoriales/fisiología , Sensación Térmica/fisiología , Canales de Potencial de Receptor Transitorio/metabolismo
16.
J Neurogenet ; 34(3-4): 351-362, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32316810

RESUMEN

Caenorhabditis elegans has a simple nervous system of 302 neurons. It however senses environmental cues incredibly precisely and produces various behaviors by processing information in the neural circuit. In addition to classical genetic analysis, fluorescent proteins and calcium indicators enable in vivo monitoring of protein dynamics and neural activity on either fixed or free-moving worms. These analyses have provided the detailed molecular mechanisms of neuronal and systemic signaling that regulate worm responses. Here, we focus on responses of C. elegans against temperature and review key findings that regulate thermotaxis and cold tolerance. Thermotaxis of C. elegans has been studied extensively for almost 50 years, and cold tolerance is a relatively recent concept in C. elegans. Although both thermotaxis and cold tolerance require temperature sensation, the responsible neurons and molecular pathways are different, and C. elegans uses the proper mechanisms depending on its situation. We summarize the molecular mechanisms of the major thermosensory circuit as well as the modulatory strategy through neural and tissue communication that enables fine tuning of thermotaxis and cold tolerance.


Asunto(s)
Reacción de Prevención/fisiología , Caenorhabditis elegans/fisiología , Frío/efectos adversos , Taxia/fisiología , Sensación Térmica/fisiología , Adaptación Fisiológica/genética , Adaptación Fisiológica/fisiología , Animales , Caenorhabditis elegans/citología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Señalización del Calcio/fisiología , Dendritas/ultraestructura , Interneuronas/fisiología , Mamíferos/fisiología , Memoria/fisiología , Vías Nerviosas/fisiología , Oxígeno/farmacología , Órganos de los Sentidos/inervación , Órganos de los Sentidos/fisiología , Células Receptoras Sensoriales/clasificación , Células Receptoras Sensoriales/fisiología , Especificidad de la Especie , Termorreceptores/fisiología
17.
Int J Biometeorol ; 64(12): 2007-2017, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32820392

RESUMEN

A new neurophysiological human thermal model based on thermoreceptor responses, the NHTM model, has been developed to predict regulatory responses and physiological variables in asymmetric transient environments. The passive system is based on Wissler's model, which is more complex and refined. Wissler's model segments the human body into 21 cylindrical parts. Each part is divided into 21 layers, 15 for the tissues and 6 for clothes, and each layer is divided into 12 angular sectors. Thus, we have 3780 nodes for the tissues and 1512 for clothes. The passive system simulates heat exchange within the body and between the body and the surroundings. The active system is composed of the thermoregulatory mechanisms, i.e., skin blood flow, shivering thermogenesis, and sweating. The skin blood flow model and the shivering model are based on thermoreceptor responses. The sweating model is that of Fiala et al. and is based on error signals. The NHTM model was compared with Wissler's model, and the results showed that a calculation based on neurophysiology can improve the performance of the thermoregulation model. The NHTM model was more accurate in the prediction of mean skin temperature, with a mean absolute error of 0.27 °C versus 0.80 °C for the original Wissler model. The prediction accuracy of the NHTM model for local skin temperatures and core temperature could be improved via an optimization method to prove the ability of the new thermoregulation model to fit with the physiological characteristics of different populations.


Asunto(s)
Neurofisiología , Termorreceptores , Regulación de la Temperatura Corporal , Humanos , Temperatura Cutánea , Sudoración
18.
Int J Mol Sci ; 21(12)2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32580281

RESUMEN

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.


Asunto(s)
Frío , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Canales Catiónicos TRPM/metabolismo , Termorreceptores/metabolismo , Sensación Térmica , Nervio Trigémino/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Células Receptoras Sensoriales/metabolismo , Nervio Trigémino/citología
19.
J Physiol ; 597(13): 3315-3332, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31093981

RESUMEN

KEY POINTS: Humans lack skin receptors for wetness (i.e. hygroreceptors), yet we present a remarkable wetness sensitivity. Afferent inputs from skin cold-sensitive thermoreceptors are key for sensing wetness; yet, it is unknown whether males and females differ in their wetness sensitivity across their body and whether high intensity exercise modulates this sensitivity. We mapped sensitivity to cold, neutral and warm wetness across five body regions and show that females are more sensitive to skin wetness than males, and that this difference is greater for cold than warm wetness sensitivity. We also show that a single bout of maximal exercise reduced the sensitivity to skin wetness (i.e. hygro-hypoesthesia) of both sexes as a result of concurrent decreases in thermal sensitivity. These novel findings clarify the physiological mechanisms underpinning this fundamental human sensory experience. In addition, they indicate sex differences in thermoregulatory responses and will inform the design of more effective sport and protective clothing, as well as thermoregulatory models. ABSTRACT: Humans lack skin hygroreceptors and we rely on integrating cold and tactile inputs from A-type skin nerve fibres to sense wetness. Yet, it is unknown whether sex and exercise independently modulate skin wetness sensitivity across the body. We mapped local sensitivity to cold, neutral and warm wetness of the forehead, neck, underarm, lower back and dorsal foot in 10 males (27.8 ± 2.7 years; 1.92 ± 0.1 m2 body surface area) and 10 females (25.4 ± 3.9 years; 1.68 ± 0.1 m2 body surface area), at rest and post maximal incremental running. Participants underwent our quantitative sensory test where they reported the magnitude of thermal and wetness perceptions (visual analogue scale) resulting from the application of a cold (5°C below skin temperature) wet (0.8 mL of water), neutral wet and warm wet (5°C above skin temperature) thermal probe (1.32 cm2 ) to five skin sites. We found that: (i) females were ∼14% to ∼17% more sensitive to cold-wetness than males, yet both sexes were as sensitive to neutral- and warm-wetness; (ii) regional differences were present for cold-wetness only, and these followed a craniocaudal increase that was more pronounced in males (i.e. the foot was ∼31% more sensitive than the forehead); and (iii) maximal exercise reduced cold-wetness sensitivity over specific regions in males (i.e. ∼40% decrease in foot sensitivity), and also induced a generalized reduction in warm-wetness sensitivity in both sexes (i.e. ∼4% to ∼6%). For the first time, we show that females are more sensitive to cold wetness than males and that maximal exercise induce hygro-hypoesthesia. These novel findings expand our knowledge on sex differences in thermoregulatory physiology.


Asunto(s)
Ejercicio Físico/fisiología , Descanso/fisiología , Carrera/fisiología , Piel/fisiopatología , Sensación Térmica/fisiología , Adulto , Frío , Femenino , Pie/fisiología , Calor , Humanos , Masculino , Estimulación Física/métodos , Caracteres Sexuales , Fenómenos Fisiológicos de la Piel , Temperatura Cutánea/fisiología , Termorreceptores/fisiología , Tacto/fisiología , Percepción del Tacto/fisiología
20.
J Neurophysiol ; 121(5): 1798-1808, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30864857

RESUMEN

Nonnoxious warmth reduces both perceived pain intensity and the amplitude of EEG markers of pain. However, the spatial properties of thermonociceptive interaction, and the level of sensory processing at which it occurs, remain unclear. We investigated whether interchannel warmth-pain interactions occur before or after intrachannel spatial summation of warmth. Warm stimuli were applied to the fingers of the right hand. Their number and location were manipulated in different conditions. A concomitant noxious test pulse was delivered to the middle finger using a CO2 laser. We replicated the classical suppressive effect of warmth on both perceived pain intensity and EEG markers. Importantly, inhibition of pain was not affected by the location and the number of thermal stimuli, even though they increased the perceived intensity of warmth. Our results therefore suggest that the inhibitory effect of warmth on pain is not somatotopically organized. The results also rule out the possibility that warmth affects nociceptive processing after intrachannel warmth summation. NEW & NOTEWORTHY We used spatial summation of warmth as a model to investigate thermonociceptive interactions. Painful CO2 laser pulses were delivered during different thermal conditions. We found that warmth inhibited pain regardless of its location. Crucially, spatial summation of multiple warm stimuli did not further inhibit pain. These findings suggest that warmth-pain interaction occurs independently of or after spatial summation of warmth.


Asunto(s)
Calor , Nocicepción , Percepción del Dolor , Percepción del Tacto , Adulto , Femenino , Dedos/fisiología , Humanos , Masculino , Contracción Muscular , Nociceptores/fisiología , Termorreceptores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA