Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 546
Filter
1.
Compr Physiol ; 14(2): 5389-5406, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-39109978

ABSTRACT

Uncontrolled angiogenesis underlies various pathological conditions such as cancer, age-related macular degeneration (AMD), and proliferative diabetic retinopathy (PDR). Hence, targeting pathological angiogenesis has become a promising strategy for the treatment of cancer and neovascular ocular diseases. However, current pharmacological treatments that target VEGF signaling have met with limited success either due to acquiring resistance against anti-VEGF therapies with serious side effects including nephrotoxicity and cardiovascular-related adverse effects in cancer patients or retinal vasculitis and intraocular inflammation after intravitreal injection in patients with AMD or PDR. Therefore, there is an urgent need to develop novel strategies which can control multiple aspects of the pathological microenvironment and regulate the process of abnormal angiogenesis. To this end, vascular normalization has been proposed as an alternative for antiangiogenesis approach; however, these strategies still focus on targeting VEGF or FGF or PDGF which has shown adverse effects. In addition to these growth factors, calcium has been recently implicated as an important modulator of tumor angiogenesis. This article provides an overview on the role of major calcium channels in endothelium, TRP channels, with a special focus on TRPV4 and its downstream signaling pathways in the regulation of pathological angiogenesis and vascular normalization. We also highlight recent findings on the modulation of TRPV4 activity and endothelial phenotypic transformation by tumor microenvironment through Rho/YAP/VEGFR2 mechanotranscriptional pathways. Finally, we provide perspective on endothelial TRPV4 as a novel VEGF alternative therapeutic target for vascular normalization and improved therapy. © 2024 American Physiological Society. Compr Physiol 14:5389-5406, 2024.


Subject(s)
Neovascularization, Pathologic , Humans , Animals , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Signal Transduction
2.
Front Neural Circuits ; 18: 1435757, 2024.
Article in English | MEDLINE | ID: mdl-39045140

ABSTRACT

Thermoregulation is a fundamental mechanism for maintaining homeostasis in living organisms because temperature affects essentially all biochemical and physiological processes. Effector responses to internal and external temperature cues are critical for achieving effective thermoregulation by controlling heat production and dissipation. Thermoregulation can be classified as physiological, which is observed primarily in higher organisms (homeotherms), and behavioral, which manifests as crucial physiological functions that are conserved across many species. Neuronal pathways for physiological thermoregulation are well-characterized, but those associated with behavioral regulation remain unclear. Thermoreceptors, including Transient Receptor Potential (TRP) channels, play pivotal roles in thermoregulation. Mammals have 11 thermosensitive TRP channels, the functions for which have been elucidated through behavioral studies using knockout mice. Behavioral thermoregulation is also observed in ectotherms such as the fruit fly, Drosophila melanogaster. Studies of Drosophila thermoregulation helped elucidate significant roles for thermoreceptors as well as regulatory actions of membrane lipids in modulating the activity of both thermosensitive TRP channels and thermoregulation. This review provides an overview of thermosensitive TRP channel functions in behavioral thermoregulation based on results of studies involving mice or Drosophila melanogaster.


Subject(s)
Body Temperature Regulation , Transient Receptor Potential Channels , Animals , Body Temperature Regulation/physiology , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Behavior, Animal/physiology , Thermosensing/physiology , Drosophila melanogaster/physiology , Mice , Humans
3.
Subcell Biochem ; 104: 207-244, 2024.
Article in English | MEDLINE | ID: mdl-38963489

ABSTRACT

The transient receptor potential ion channel TRPA1 is a Ca2+-permeable nonselective cation channel widely expressed in sensory neurons, but also in many nonneuronal tissues typically possessing barrier functions, such as the skin, joint synoviocytes, cornea, and the respiratory and intestinal tracts. Here, the primary role of TRPA1 is to detect potential danger stimuli that may threaten the tissue homeostasis and the health of the organism. The ability to directly recognize signals of different modalities, including chemical irritants, extreme temperatures, or osmotic changes resides in the characteristic properties of the ion channel protein complex. Recent advances in cryo-electron microscopy have provided an important framework for understanding the molecular basis of TRPA1 function and have suggested novel directions in the search for its pharmacological regulation. This chapter summarizes the current knowledge of human TRPA1 from a structural and functional perspective and discusses the complex allosteric mechanisms of activation and modulation that play important roles under physiological or pathophysiological conditions. In this context, major challenges for future research on TRPA1 are outlined.


Subject(s)
TRPA1 Cation Channel , Humans , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/physiology , Cryoelectron Microscopy/methods , Animals , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/physiology , Structure-Activity Relationship , Allosteric Regulation
4.
J Therm Biol ; 122: 103868, 2024 May.
Article in English | MEDLINE | ID: mdl-38852485

ABSTRACT

Transient Receptor Potential (TRP) ion channels are important for sensing environmental temperature. In rodents, TRPV4 senses warmth (25-34 °C), TRPV1 senses heat (>42 °C), TRPA1 putatively senses cold (<17 °C), and TRPM8 senses cool-cold (18-26 °C). We investigated if knockout (KO) mice lacking these TRP channels exhibited changes in thermal preference. Thermal preference was tested using a dual hot-cold plate with one thermoelectric surface set at 30 °C and the adjacent surface at a temperature of 15-45 °C in 5 °C increments. Blinded observers counted the number of times mice crossed through an opening between plates and the percentage of time spent on the 30 °C plate. In a separate experiment, observers blinded as to genotype also assessed the temperature at the location on a thermal gradient (1.83 m, 4-50 °C) occupied by the mouse at 5- or 10-min intervals over 2 h. Male and female wildtype mice preferred 30 °C and significantly avoided colder (15-20 °C) and hotter (40-45 °C) temperatures. Male TRPV1KOs and TRPA1KOs, and TRPV4KOs of both sexes, were similar, while female WTs, TRPV1KOs, TRPA1KOs and TRPM8KOs did not show significant thermal preferences across the temperature range. Male and female TRPM8KOs did not significantly avoid the coldest temperatures. Male mice (except for TRPM8KOs) exhibited significantly fewer plate crossings at hot and cold temperatures and more crossings at thermoneutral temperatures, while females exhibited a similar but non-significant trend. Occupancy temperatures along the thermal gradient exhibited a broad distribution that shrank somewhat over time. Mean occupancy temperatures (recorded at 90-120 min) were significantly higher for females (30-34 °C) compared to males (26-27 °C) of all genotypes, except for TRPA1KOs which exhibited no sex difference. The results indicate (1) sex differences with females (except TRPA1KOs) preferring warmer temperatures, (2) reduced thermosensitivity in female TRPV1KOs, and (3) reduced sensitivity to cold and innocuous warmth in male and female TRPM8KOs consistent with previous studies.


Subject(s)
Mice, Knockout , TRPA1 Cation Channel , TRPV Cation Channels , Thermosensing , Animals , Female , Male , Mice , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Mice, Inbred C57BL , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Hot Temperature , Cold Temperature
5.
Open Biol ; 13(10): 230215, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37848053

ABSTRACT

Antarctic notothenioid fishes (cryonotothenioids) live in waters that range between -1.86°C and an extreme maximum +4°C. Evidence suggests these fish sense temperature peripherally, but the molecular mechanism of temperature sensation in unknown. Previous work identified transient receptor potential (TRP) channels TRPA1b, TRPM4 and TRPV1a as the top candidates for temperature sensors. Here, cryonotothenioid TRPA1b and TRPV1a are characterized using Xenopus oocyte electrophysiology. TRPA1b and TRPV1a showed heat-evoked currents with Q10s of 11.1 ± 2.2 and 20.5 ± 2.4, respectively. Unexpectedly, heat activation occurred at a threshold of 22.9 ± 1.3°C for TRPA1b and 32.1 ± 0.6°C for TRPV1a. These fish have not experienced such temperatures for at least 15 Myr. Either (1) another molecular mechanism underlies temperature sensation, (2) these fishes do not sense temperatures below these thresholds despite having lethal limits as low as 5°C, or (3) native cellular conditions modify the TRP channels to function at relevant temperatures. The effects of osmolytes, pH, oxidation, phosphorylation, lipids and accessory proteins were tested. No conditions shifted the activity range of TRPV1a. Oxidation in combination with reduced cholesterol significantly dropped activation threshold of TRPA1b to 11.3 ± 2.3°C, it is hypothesized the effect may be due to lipid raft disruption.


Subject(s)
Fishes , Transient Receptor Potential Channels , Animals , Temperature , Hot Temperature , Ion Channels , Antarctic Regions , Transient Receptor Potential Channels/physiology
6.
Prog Retin Eye Res ; 92: 101114, 2023 01.
Article in English | MEDLINE | ID: mdl-36163161

ABSTRACT

Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.


Subject(s)
Drosophila Proteins , Transient Receptor Potential Channels , Animals , Transient Receptor Potential Channels/physiology , Mechanotransduction, Cellular , Retina/metabolism , Drosophila/metabolism , Homeostasis , Drosophila Proteins/metabolism
7.
Biochim Biophys Acta Biomembr ; 1864(11): 184014, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35908608

ABSTRACT

Glucosylsphingosine (GS) is an endogenous sphingolipid that specifically accumulates in the skin of patients with atopic dermatitis (AD). Notably, it was recently found that GS can induce itch sensation by activating serotonin receptor 2A and TRPV4 ion channels. However, it is still uncertain whether other molecules are involved in GS-induced itch sensation. Therefore, by using the calcium imaging technique, we investigated whether serotonin receptor 2 - specifically 2A and 2B - can interact with TRPV1 and TRPA1, because these are representative ion channels in the transmission of itch. As a result, it was found that GS did not activate TRPV1 or TRPA1 per se. Moreover, cells expressing both serotonin receptor 2 and TRPV1 did not show any changes in calcium responses. However, enhanced calcium responses were observed in cells expressing serotonin receptor 2 and TRPA1, suggesting a possible interaction between these two molecules. Similar synergistic effects were also observed in cells expressing serotonin receptor 2 and TRPA1, but not TRPV1. Furthermore, a phospholipase C inhibitor (U73122) and a store-operated calcium entry blocker (SKF96365) significantly reduced GS-induced responses in cells expressing both serotonin receptor 2 and TRPA1, but not with pre-treatment with a Gßγ-complex blocker (gallein). Therefore, we propose a putative novel pathway for GS-induced itch sensation, such that serotonin receptor 2 could be coupled to TRPA1 but not TRPV1 in sensory neurons.


Subject(s)
Transient Receptor Potential Channels , Calcium/metabolism , Humans , Pruritus/metabolism , Psychosine/analogs & derivatives , Receptors, Serotonin/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels , Transient Receptor Potential Channels/physiology
8.
Int J Mol Sci ; 23(13)2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35806193

ABSTRACT

Neuropathic pain is common in diabetic peripheral neuropathy (DN), probably caused by pathogenic ion channel gene variants. Therefore, we performed molecular inversion probes-next generation sequencing of 5 transient receptor potential cation channels, 8 potassium channels and 2 calcium-activated chloride channel genes in 222 painful- and 304 painless-DN patients. Twelve painful-DN (5.4%) patients showed potentially pathogenic variants (five nonsense/frameshift, seven missense, one out-of-frame deletion) in ANO3 (n = 3), HCN1 (n = 1), KCNK18 (n = 2), TRPA1 (n = 3), TRPM8 (n = 3) and TRPV4 (n = 1) and fourteen painless-DN patients (4.6%-three nonsense/frameshift, nine missense, one out-of-frame deletion) in ANO1 (n = 1), KCNK18 (n = 3), KCNQ3 (n = 1), TRPA1 (n = 2), TRPM8 (n = 1), TRPV1 (n = 3) and TRPV4 (n = 3). Missense variants were present in both conditions, presumably with loss- or gain-of-functions. KCNK18 nonsense/frameshift variants were found in painless/painful-DN, making a causal role in pain less likely. Surprisingly, premature stop-codons with likely nonsense-mediated RNA-decay were more frequent in painful-DN. Although limited in number, painful-DN patients with ion channel gene variants reported higher maximal pain during the night and day. Moreover, painful-DN patients with TRP variants had abnormal thermal thresholds and more severe pain during the night and day. Our results suggest a role of ion channel gene variants in neuropathic pain, but functional validation is required.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Neuralgia , Transient Receptor Potential Channels , Anoctamins , Humans , Potassium Channels , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/physiology
9.
Sci Rep ; 11(1): 20627, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663887

ABSTRACT

Cnidarians are characterized by the possession of stinging organelles, called nematocysts, which they use for prey capture and defense. Nematocyst discharge is controlled by a mechanosensory apparatus with analogies to vertebrate hair cells. Members of the transient receptor potential (TRPN) ion channel family are supposed to be involved in the transduction of the mechanical stimulus. A small molecule screen was performed to identify compounds that affect nematocyst discharge in Hydra. We identified several [2.2]paracyclophanes that cause inhibition of nematocyst discharge in the low micro-molar range. Further structure-activity analyses within the compound class of [2.2]paracyclophanes showed common features that are required for the inhibitory activity of the [2.2]paracyclophane core motif. This study demonstrates that Hydra can serve as a model for small molecule screens targeting the mechanosensory apparatus in native tissues.


Subject(s)
Hydra/immunology , Nematocyst/drug effects , Nematocyst/physiology , Animals , Biomechanical Phenomena/drug effects , Biomechanical Phenomena/physiology , Cnidaria , Hydra/metabolism , Small Molecule Libraries/pharmacology , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/physiology
10.
Elife ; 102021 06 08.
Article in English | MEDLINE | ID: mdl-34101577

ABSTRACT

NompC is a mechanosensitive ion channel responsible for the sensation of touch and balance in Drosophila melanogaster. Based on a resolved cryo-EM structure, we performed all-atom molecular dynamics simulations and electrophysiological experiments to study the atomistic details of NompC gating. Our results showed that NompC could be opened by compression of the intracellular ankyrin repeat domain but not by a stretch, and a number of hydrogen bonds along the force convey pathway are important for the mechanosensitivity. Under intracellular compression, the bundled ankyrin repeat region acts like a spring with a spring constant of ~13 pN nm-1 by transferring forces at a rate of ~1.8 nm ps-1. The linker helix region acts as a bridge between the ankyrin repeats and the transient receptor potential (TRP) domain, which passes on the pushing force to the TRP domain to undergo a clockwise rotation, resulting in the opening of the channel. This could be the universal gating mechanism of similar tethered mechanosensitive TRP channels, which enable cells to feel compression and shrinkage.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/metabolism , Animals , Ankyrin Repeat , Cell Line , Drosophila Proteins/physiology , Drosophila Proteins/ultrastructure , Drosophila melanogaster , Hydrogen Bonding , Molecular Dynamics Simulation , Transient Receptor Potential Channels/physiology , Transient Receptor Potential Channels/ultrastructure
11.
Cells ; 10(5)2021 04 26.
Article in English | MEDLINE | ID: mdl-33925979

ABSTRACT

The transient receptor potential channels (TRPs) have been related to several different physiologies that range from a role in sensory physiology (including thermo- and osmosensation) to a role in some pathologies like cancer. The great diversity of functions performed by these channels is represented by nine sub-families that constitute the TRP channel superfamily. From the mid-2000s, several reports have shown the potential role of the TRP channels in cancers of multiple origin. The pancreatic cancer is one of the deadliest cancers worldwide. Its prevalence is predicted to rise further. Disappointingly, the treatments currently used are ineffective. There is an urgency to find new ways to counter this disease and one of the answers may lie in the ion channels belonging to the superfamily of TRP channels. In this review, we analyse the existing knowledge on the role of TRP channels in the development and progression of pancreatic ductal adenocarcinoma (PDAC). The functions of these channels in other cancers are also considered. This might be of interest for an extrapolation to the pancreatic cancer in an attempt to identify potential therapeutic interventions.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/metabolism , Transient Receptor Potential Channels/physiology , Animals , Calcium/metabolism , Calcium Signaling , Carcinoma, Pancreatic Ductal/pathology , Cations , Cell Movement , Cell Proliferation , Disease Progression , Humans , Ion Channels , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Phylogeny , Prevalence , Tumor Microenvironment
12.
Sci Rep ; 11(1): 8313, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859333

ABSTRACT

The cation channel TRPML1 is an important regulator of lysosomal function and autophagy. Loss of TRPML1 is associated with neurodegeneration and lysosomal storage disease, while temporary inhibition of this ion channel has been proposed to be beneficial in cancer therapy. Currently available TRPML1 channel inhibitors are not TRPML isoform selective and block at least two of the three human isoforms. We have now identified the first highly potent and isoform-selective TRPML1 antagonist, the steroid 17ß-estradiol methyl ether (EDME). Two analogs of EDME, PRU-10 and PRU-12, characterized by their reduced activity at the estrogen receptor, have been identified through systematic chemical modification of the lead structure. EDME and its analogs, besides being promising new small molecule tool compounds for the investigation of TRPML1, selectively affect key features of TRPML1 function: autophagy induction and transcription factor EB (TFEB) translocation. In addition, they act as inhibitors of triple-negative breast cancer cell migration and invasion.


Subject(s)
Autophagy/drug effects , Cell Movement/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Female , Humans , Neoplasm Invasiveness , Triple Negative Breast Neoplasms/pathology
13.
J Cell Mol Med ; 25(7): 3469-3483, 2021 04.
Article in English | MEDLINE | ID: mdl-33689230

ABSTRACT

The use of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is limited in drug discovery and cardiac disease mechanism studies due to cell immaturity. Micro-scaled grooves can promote the maturation of cardiomyocytes by aligning them in order, but the mechanism of cardiomyocytes alignment has not been studied. From the level of calcium activity, gene expression and cell morphology, we verified that the W20H5 grooves can effectively promote the maturation of cardiomyocytes. The transient receptor potential channels (TRP channels) also play an important role in the maturation and development of cardiomyocytes. These findings support the engineered hPSC-CMs as a powerful model to study cardiac disease mechanism and partly mimic the myocardial morphological development. The important role of the TRP channels in the maturation and development of myocardium is first revealed.


Subject(s)
Cell Differentiation , Connexin 43/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transient Receptor Potential Channels/physiology , Calcium/metabolism , Cell Movement , Cells, Cultured , Humans , Mechanoreceptors/physiology , Stress, Mechanical
14.
Neurosci Lett ; 748: 135719, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33587987

ABSTRACT

Transient Receptor Potential (TRP) channels expressed in specific subsets of airway sensory nerves function as transducers and integrators of a diverse range of sensory inputs including chemical, mechanical and thermal signals. These TRP sensors can detect inhaled irritants as well as endogenously released chemical substances. They play an important role in generating the afferent activity carried by these sensory nerves and regulating the centrally mediated pulmonary defense reflexes. Increasing evidence reported in recent investigations has revealed important involvements of several TRP channels (TRPA1, TRPV1, TRPV4 and TRPM8) in the manifestation of various symptoms and pathogenesis of certain acute and chronic airway diseases. This mini-review focuses primarily on these recent findings of the responses of these TRP sensors to the biological stresses emerging under the pathophysiological conditions of the lung and airways.


Subject(s)
Afferent Pathways/physiology , Lung/physiology , Sensory Receptor Cells/physiology , Transient Receptor Potential Channels/physiology , Animals , Humans , Lung/innervation , Peripheral Nervous System , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology
15.
Histol Histopathol ; 36(5): 515-526, 2021 May.
Article in English | MEDLINE | ID: mdl-33528023

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of death in the civilized world. Transient receptor potential channels (TRPs) are a heterogeneous family of cation channels that play an important role in gastrointestinal physiology. TRPs have been linked with carcinogenesis in the colon and their role as potential therapeutic targets and prognostic biomarkers is under investigation.


Subject(s)
Colorectal Neoplasms , Transient Receptor Potential Channels/physiology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Carcinogenesis , Colon/physiopathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Drug Delivery Systems , Gastrointestinal Tract/physiopathology , Humans , Prognosis
16.
Neuroendocrinology ; 111(1-2): 45-69, 2021.
Article in English | MEDLINE | ID: mdl-32028278

ABSTRACT

OBJECTIVE: We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS: Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS: In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS: These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Energy Metabolism/physiology , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Pro-Opiomelanocortin , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Transient Receptor Potential Channels/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Electrophysiological Phenomena , Energy Metabolism/drug effects , Female , Guinea Pigs , Homeostasis , Male , Mice , Mice, Transgenic , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/drug effects , Transient Receptor Potential Channels/drug effects
17.
Biomed Pharmacother ; 131: 110647, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32858500

ABSTRACT

Stroke is the leading cause of long-term disability, demanding an ever-increasing need to find treatment. Transient receptor potential (TRP) channels are nonselective Ca2+-permeable channels, among which TRPC, TRPM, and TRPV are widely expressed in the brain. Dysfunction of the blood brain barrier (BBB) is a core feature of stroke and is associated with severity of injury. As studies have shown, TRP channels influence various neuronal functions by regulating the BBB. Here, we briefly review the role of TRP channel in the BBB dysfunction after stroke, and explore the therapeutic potential of TRP-targeted therapy.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Ischemic Stroke/metabolism , Transient Receptor Potential Channels/physiology , Animals , Biological Transport/physiology , Blood-Brain Barrier/pathology , Brain/pathology , Brain Ischemia/pathology , Humans , Ischemic Stroke/pathology
18.
Elife ; 92020 08 17.
Article in English | MEDLINE | ID: mdl-32804077

ABSTRACT

The recent proliferation of published TRP channel structures provides a foundation for understanding the diverse functional properties of this important family of ion channel proteins. To facilitate mechanistic investigations, we constructed a structure-based alignment of the transmembrane domains of 120 TRP channel structures. Comparison of structures determined in the absence or presence of activating stimuli reveals similar constrictions in the central ion permeation pathway near the intracellular end of the S6 helices, pointing to a conserved cytoplasmic gate and suggesting that most available structures represent non-conducting states. Comparison of the ion selectivity filters toward the extracellular end of the pore supports existing hypotheses for mechanisms of ion selectivity. Also conserved to varying extents are hot spots for interactions with hydrophobic ligands, lipids and ions, as well as discrete alterations in helix conformations. This analysis therefore provides a framework for investigating the structural basis of TRP channel gating mechanisms and pharmacology, and, despite the large number of structures included, reveals the need for additional structural data and for more functional studies to establish the mechanistic basis of TRP channel function.


Subject(s)
Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/physiology , Animals , Humans , Ion Channel Gating , Models, Molecular , Protein Conformation , Protein Domains , Sequence Alignment , Structure-Activity Relationship
19.
Int J Mol Sci ; 21(11)2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32545371

ABSTRACT

Transient receptor potential (TRP) or transient receptor potential channels are a highly diverse family of mostly non-selective cation channels. In the mammalian genome, 28 members can be identified, most of them being expressed predominantly in the plasma membrane with the exception of the mucolipins or TRPMLs which are expressed in the endo-lysosomal system. In mammalian organisms, TRPMLs have been associated with a number of critical endo-lysosomal functions such as autophagy, endo-lysosomal fusion/fission and trafficking, lysosomal exocytosis, pH regulation, or lysosomal motility and positioning. The related non-selective two-pore cation channels (TPCs), likewise expressed in endosomes and lysosomes, have also been found to be associated with endo-lysosomal trafficking, autophagy, pH regulation, or lysosomal exocytosis, raising the question why these two channel families have evolved independently. We followed TRP/TRPML channels and TPCs through evolution and describe here in which species TRP/TRPMLs and/or TPCs are found, which functions they have in different species, and how this compares to the functions of mammalian orthologs.


Subject(s)
Calcium Channels/physiology , Transient Receptor Potential Channels/physiology , Animals , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Evolution, Molecular , Fungal Proteins/physiology , Humans , Plant Proteins/physiology , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
20.
Int J Mol Sci ; 21(11)2020 May 30.
Article in English | MEDLINE | ID: mdl-32486187

ABSTRACT

Transient receptor potential canonical (TRPC) proteins constitute a group of receptor-operated calcium-permeable nonselective cationic membrane channels of the TRP superfamily. They are largely expressed in the hippocampus and are able to modulate neuronal functions. Accordingly, they have been involved in different hippocampal functions such as learning processes and different types of memories, as well as hippocampal dysfunctions such as seizures. This review covers the mechanisms of activation of these channels, how these channels can modulate neuronal excitability, in particular the after-burst hyperpolarization, and in the persistent activity, how they control synaptic plasticity including pre- and postsynaptic processes and how they can interfere with cell survival and neurogenesis.


Subject(s)
Brain/physiology , Hippocampus/physiology , Seizures/physiopathology , Transient Receptor Potential Channels/physiology , Animals , Cell Movement , Cell Proliferation , Excitatory Postsynaptic Potentials , Humans , Long-Term Potentiation , Memory/physiology , Memory, Short-Term , Mice , Neurogenesis , Neuronal Plasticity , Neurons/physiology , Protein Isoforms , Receptors, Metabotropic Glutamate/physiology , Spatial Memory , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL