Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
Circ Res ; 134(5): 572-591, 2024 03.
Article in English | MEDLINE | ID: mdl-38422173

ABSTRACT

The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.


Subject(s)
Anemia, Hemolytic, Congenital , Hypertension, Pulmonary , Animals , Female , Humans , Blood Pressure , Biophysics , Hydrops Fetalis , Mammals
2.
bioRxiv ; 2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37131687

ABSTRACT

The enteric nervous system (ENS) is a complex network of diverse molecularly defined classes of neurons embedded in the gastrointestinal wall and responsible for controlling the major functions of the gut. As in the central nervous system, the vast array of ENS neurons is interconnected by chemical synapses. Despite several studies reporting the expression of ionotropic glutamate receptors in the ENS, their roles in the gut remain elusive. Here, by using an array of immunohistochemistry, molecular profiling and functional assays, we uncover a new role for d-serine (d-Ser) and non-conventional GluN1-GluN3 N-methyl d-aspartate receptors (NMDARs) in regulating ENS functions. We demonstrate that d-Ser is produced by serine racemase (SR) expressed in enteric neurons. By using both in situ patch clamp recording and calcium imaging, we show that d-Ser alone acts as an excitatory neurotransmitter in the ENS independently of the conventional GluN1-GluN2 NMDARs. Instead, d-Ser directly gates the non-conventional GluN1-GluN3 NMDARs in enteric neurons from both mouse and guinea-pig. Pharmacological inhibition or potentiation of GluN1-GluN3 NMDARs had opposite effects on mouse colonic motor activities, while genetically driven loss of SR impairs gut transit and fluid content of pellet output. Our results demonstrate the existence of native GluN1-GluN3 NMDARs in enteric neurons and open new perspectives on the exploration of excitatory d-Ser receptors in gut function and diseases.

4.
Neuron ; 110(17): 2713-2727, 2022 09 07.
Article in English | MEDLINE | ID: mdl-35907398

ABSTRACT

Many ion channels have been described as mechanosensitive according to various criteria. Most broadly defined, an ion channel is called mechanosensitive if its activity is controlled by application of a physical force. The last decade has witnessed a revolution in mechanosensory physiology at the molecular, cellular, and system levels, both in health and in diseases. Since the discovery of the PIEZO proteins as prototypical mechanosensitive channel, many proteins have been proposed to transduce mechanosensory information in mammals. However, few of these newly identified candidates have all the attributes of bona fide, pore-forming mechanosensitive ion channels. In this perspective, we will cover and discuss new data that have advanced our understanding of mechanosensation at the molecular level.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Animals , Ion Channels/metabolism , Mammals/metabolism , Mechanotransduction, Cellular/physiology
5.
FASEB J ; 35(12): e22025, 2021 12.
Article in English | MEDLINE | ID: mdl-34758144

ABSTRACT

Mepyramine, a first-generation antihistamine targeting the histamine H(1) receptor, was extensively prescribed to patients suffering from allergic reactions and urticaria. Serious adverse effects, especially in case of overdose, were frequently reported, including drowsiness, impaired thinking, convulsion, and coma. Many of these side effects were associated with the blockade of histaminergic or cholinergic receptors. Here we show that mepyramine directly inhibits a variety of voltage-gated sodium channels, including the Tetrodotoxin-sensitive isoforms and the main isoforms (Nav1.7, Nav1.8, and Nav1.9) of nociceptors. Estimated IC50 were within the range of drug concentrations detected in poisoned patients. Mepyramine inhibited sodium channels through fast- or slow-inactivated state preference depending on the isoform. Moreover, mepyramine inhibited the firing responses of C- and Aß-type nerve fibers in ex vivo skin-nerve preparations. Locally applied mepyramine had analgesic effects on the scorpion toxin-induced excruciating pain and produced pain relief in acute, inflammatory, and chronic pain models. Collectively, these data provide evidence that mepyramine has the potential to be developed as a topical analgesic agent.


Subject(s)
Arthritis, Experimental/complications , Ganglia, Spinal/drug effects , NAV1.8 Voltage-Gated Sodium Channel/physiology , Nociceptors/drug effects , Pain/drug therapy , Pyrilamine/pharmacology , Sodium Channel Blockers/pharmacology , Action Potentials , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Histamine H1 Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.8 Voltage-Gated Sodium Channel/chemistry , Nociceptors/metabolism , Nociceptors/pathology , Pain/etiology , Pain/metabolism , Pain/pathology
6.
Cell Rep ; 37(5): 109914, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731626

ABSTRACT

A variety of mechanosensory neurons are involved in touch, proprioception, and pain. Many molecular components of the mechanotransduction machinery subserving these sensory modalities remain to be discovered. Here, we combine recordings of mechanosensitive (MS) currents in mechanosensory neurons with single-cell RNA sequencing. Transcriptional profiles are mapped onto previously identified sensory neuron types to identify cell-type correlates between datasets. Correlation of current signatures with single-cell transcriptomes provides a one-to-one correspondence between mechanoelectric properties and transcriptomically defined neuronal populations. Moreover, a gene-expression differential comparison provides a set of candidate genes for mechanotransduction complexes. Piezo2 is expectedly found to be enriched in rapidly adapting MS current-expressing neurons, whereas Tmem120a and Tmem150c, thought to mediate slow-type MS currents, are uniformly expressed in all mechanosensory neuron subtypes. Further knockdown experiments disqualify them as mediating MS currents in sensory neurons. This dataset constitutes an open resource to explore further the cell-type-specific determinants of mechanosensory properties.


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Profiling , Mechanotransduction, Cellular/genetics , Neurons/metabolism , Transcriptome , Animals , Ganglia, Spinal/cytology , Gene Expression Regulation , HEK293 Cells , Humans , Ion Channels/genetics , Ion Channels/metabolism , Male , Membrane Potentials , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Patch-Clamp Techniques , RNA-Seq , Single-Cell Analysis
8.
J Gen Physiol ; 153(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34694360

ABSTRACT

Cholangiocytes actively contribute to the final composition of secreted bile. These cells are exposed to abnormal mechanical stimuli during obstructive cholestasis, which has a deep impact on their function. However, the effects of mechanical insults on cholangiocyte function are not understood. Combining gene silencing and pharmacological assays with live calcium imaging, we probed molecular candidates essential for coupling mechanical force to ATP secretion in mouse cholangiocytes. We show that Piezo1 and Pannexin1 are necessary for eliciting the downstream effects of mechanical stress. By mediating a rise in intracellular Ca2+, Piezo1 acts as a mechanosensor responsible for translating cell swelling into activation of Panx1, which triggers ATP release and subsequent signal amplification through P2X4R. Co-immunoprecipitation and pull-down assays indicated physical interaction between Piezo1 and Panx1, which leads to stable plasma membrane complexes. Piezo1-Panx1-P2X4R ATP release pathway could be reconstituted in HEK Piezo1 KO cells. Thus, our data suggest that Piezo1 and Panx1 can form a functional signaling complex that controls force-induced ATP secretion in cholangiocytes. These findings may foster the development of novel therapeutic strategies for biliary diseases.


Subject(s)
Adenosine Triphosphate , Connexins , Epithelial Cells , Ion Channels , Nerve Tissue Proteins , Animals , Calcium/metabolism , Cell Membrane/metabolism , Connexins/genetics , Epithelial Cells/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Mice , Nerve Tissue Proteins/genetics , Signal Transduction
9.
Br J Pharmacol ; 178(13): 2632-2650, 2021 07.
Article in English | MEDLINE | ID: mdl-33742442

ABSTRACT

BACKGROUND AND PURPOSE: Protoxin II (ProTx II) is a high affinity gating modifier that is thought to selectively block the Nav 1.7 voltage-dependent Na+ channel, a major therapeutic target for the control of pain. We aimed at producing ProTx II analogues entitled with novel functionalities for cell distribution studies and biochemical characterization of its Nav channel targets. EXPERIMENTAL APPROACH: We took advantage of the high affinity properties of the peptide, combined to its slow off rate, to design a number of new tagged analogues useful for imaging and biochemistry purposes. We used high-throughput automated patch-clamp to identify the analogues best matching the native properties of ProTx II and validated them on various Nav -expressing cells in pull-down and cell distribution studies. KEY RESULTS: Two of the produced ProTx II analogues, Biot-ProTx II and ATTO488-ProTx II, best emulate the pharmacological properties of unlabelled ProTx II, whereas other analogues remain high affinity blockers of Nav 1.7. The biotinylated version of ProTx II efficiently works for the pull-down of several Nav isoforms tested in a concentration-dependent manner, whereas the fluorescent ATTO488-ProTx II specifically labels the Nav 1.7 channel over other Nav isoforms tested in various experimental conditions. CONCLUSIONS AND IMPLICATIONS: The properties of these ProTx II analogues as tools for Nav channel purification and cell distribution studies pave the way for a better understanding of ProTx II channel receptors in pain and their pathophysiological implications in sensory neuronal processing. The new fluorescent ProTx II should also be useful in the design of new drug screening strategies.


Subject(s)
Spider Venoms , Humans , NAV1.7 Voltage-Gated Sodium Channel , Pain , Peptides
10.
J Pain ; 22(4): 440-453, 2021 04.
Article in English | MEDLINE | ID: mdl-33227509

ABSTRACT

Oral amitriptyline hydrochloride (amitriptyline) is ineffective against some forms of chronic pain and is often associated with dose-limiting adverse events. We evaluated the potential effectiveness of high-dose topical amitriptyline in a preliminary case series of chemotherapy-induced peripheral neuropathy patients and investigated whether local or systemic adverse events associated with the use of amitriptyline were present in these patients. We also investigated the mechanism of action of topically administered amitriptyline in mice. Our case series suggested that topical 10% amitriptyline treatment was associated with pain relief in chemotherapy-induced peripheral neuropathy patients, without the side effects associated with systemic absorption. Topical amitriptyline significantly increased mechanical withdrawal thresholds when applied to the hind paw of mice, and inhibited the firing responses of C-, Aß- and Aδ-type peripheral nerve fibers in ex vivo skin-saphenous nerve preparations. Whole-cell patch-clamp recordings on cultured sensory neurons revealed that amitriptyline was a potent inhibitor of the main voltage-gated sodium channels (Nav1.7, Nav1.8, and Nav1.9) found in nociceptors. Calcium imaging showed that amitriptyline activated the transient receptor potential cation channel, TRPA1. Our case series indicated that high-dose 10% topical amitriptyline could alleviate neuropathic pain without adverse local or systemic effects. This analgesic action appeared to be mediated through local inhibition of voltage-gated sodium channels. PERSPECTIVE: Our preliminary case series suggested that topical amitriptyline could provide effective pain relief for chemotherapy-induced peripheral neuropathy patients without any systemic or local adverse events. Investigation of the mechanism of this analgesic action in mice revealed that this activity was mediated through local inhibition of nociceptor Nav channels.


Subject(s)
Amitriptyline/pharmacology , Analgesics, Non-Narcotic/pharmacology , Antineoplastic Agents/adverse effects , Nociceptive Pain/drug therapy , Nociceptors/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , TRPA1 Cation Channel/drug effects , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/drug effects , Administration, Topical , Adolescent , Adult , Aged , Aged, 80 and over , Amitriptyline/administration & dosage , Amitriptyline/adverse effects , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/adverse effects , Animals , Behavior, Animal/drug effects , Child , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Voltage-Gated Sodium Channel Blockers/administration & dosage , Voltage-Gated Sodium Channel Blockers/adverse effects , Young Adult
11.
Cell ; 183(1): 284-284.e1, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33007264

ABSTRACT

Ophthalmic, maxillary, and mandibular branches of the trigeminal nerve provide sensory innervation to orofacial tissues. Trigeminal sensory neurons respond to a diverse array of sensory stimuli to generate distinct sensations, including thermosensation, mechanosensation, itching, and pain. These sensory neurons also detect the distinct sharpness or pungency of many foods and beverages. This SnapShot highlights the transduction ion channels critical to orofacial sensation.


Subject(s)
Sensation/physiology , Trigeminal Nerve/anatomy & histology , Trigeminal Nerve/physiology , Cranial Nerves/anatomy & histology , Cranial Nerves/physiology , Humans , Neurons, Afferent/physiology , Pain/physiopathology
13.
Nat Commun ; 10(1): 4253, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534133

ABSTRACT

Medication-overuse headaches (MOH) occur with both over-the-counter and pain-relief medicines, including paracetamol, opioids and combination analgesics. The mechanisms that lead to MOH are still uncertain. Here, we show that abnormal activation of Nav1.9 channels by Nitric Oxide (NO) is responsible for MOH induced by triptan migraine medicine. Deletion of the Scn11a gene in MOH mice abrogates NO-mediated symptoms, including cephalic and extracephalic allodynia, photophobia and phonophobia. NO strongly activates Nav1.9 in dural afferent neurons from MOH but not normal mice. Abnormal activation of Nav1.9 triggers CGRP secretion, causing artery dilatation and degranulation of mast cells. In turn, released mast cell mediators potentiates Nav1.9 in meningeal nociceptors, exacerbating inflammation and pain signal. Analysis of signaling networks indicates that PKA is downregulated in trigeminal neurons from MOH mice, relieving its inhibitory action on NO-Nav1.9 coupling. Thus, anomalous activation of Nav1.9 channels by NO, as a result of chronic medication, promotes MOH.


Subject(s)
Headache Disorders, Secondary/pathology , Migraine Disorders/pathology , NAV1.9 Voltage-Gated Sodium Channel/metabolism , Neurons, Afferent/metabolism , Nitric Oxide/metabolism , Tryptamines/adverse effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Cell Degranulation/physiology , Cells, Cultured , Female , Headache Disorders, Secondary/chemically induced , Hyperalgesia/physiopathology , Male , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.9 Voltage-Gated Sodium Channel/genetics , Neurons, Afferent/drug effects , Nociceptors/physiology , Pain/physiopathology , Prescription Drug Overuse/adverse effects
15.
Nat Protoc ; 14(7): 2258, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30349046

ABSTRACT

In the HTML version of this article published online, the abstract contains a typo in the first sentence: "key to understanding intestinal motility anGutn of therapeutic strategies" should read "key to understanding intestinal motility and crucial to the design of theraputic strategies." The PDF version of the article is correct.

16.
Nat Commun ; 9(1): 3804, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228263

ABSTRACT

Defects in cerebrospinal fluid (CSF) flow may contribute to idiopathic scoliosis. However, the mechanisms underlying detection of CSF flow in the central canal of the spinal cord are unknown. Here we demonstrate that CSF flows bidirectionally along the antero-posterior axis in the central canal of zebrafish embryos. In the cfap298tm304 mutant, reduction of cilia motility slows transport posteriorly down the central canal and abolishes spontaneous activity of CSF-contacting neurons (CSF-cNs). Loss of the sensory Pkd2l1 channel nearly abolishes CSF-cN calcium activity and single channel opening. Recording from isolated CSF-cNs in vitro, we show that CSF-cNs are mechanosensory and require Pkd2l1 to respond to pressure. Additionally, adult pkd2l1 mutant zebrafish develop an exaggerated spine curvature, reminiscent of kyphosis in humans. These results indicate that CSF-cNs are mechanosensory cells whose Pkd2l1-driven spontaneous activity reflects CSF flow in vivo. Furthermore, Pkd2l1 in CSF-cNs contributes to maintenance of natural curvature of the spine.


Subject(s)
Cerebrospinal Fluid/metabolism , Mechanotransduction, Cellular , Neurons/metabolism , Spinal Cord/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cilia/metabolism
17.
EMBO J ; 37(8)2018 04 13.
Article in English | MEDLINE | ID: mdl-29459435

ABSTRACT

Cholesterol is a major lipid component of the mammalian plasma membrane. While much is known about its metabolism, its transport, and its role in atherosclerotic vascular disease, less is known about its role in neuronal pathophysiology. This study reveals an unexpected function of cholesterol in controlling pain transmission. We show that inflammation lowers cholesterol content in skin tissue and sensory DRG culture. Pharmacological depletion of cellular cholesterol entails sensitization of nociceptive neurons and promotes mechanical and thermal hyperalgesia through the activation of voltage-gated Nav1.9 channels. Inflammatory mediators enhance the production of reactive oxygen species and induce partitioning of Nav1.9 channels from cholesterol-rich lipid rafts to cholesterol-poor non-raft regions of the membrane. Low-cholesterol environment enhances voltage-dependent activation of Nav1.9 channels leading to enhanced neuronal excitability, whereas cholesterol replenishment reversed these effects. Consistently, we show that transcutaneous delivery of cholesterol alleviates hypersensitivity in animal models of acute and chronic inflammatory pain. In conclusion, our data establish that membrane cholesterol is a modulator of pain transmission and shed a new light on the relationship between cholesterol homeostasis, inflammation, and pain.


Subject(s)
Cell Membrane/physiology , Cholesterol/physiology , Inflammation/physiopathology , NAV1.9 Voltage-Gated Sodium Channel/physiology , Pain/physiopathology , Animals , Ganglia, Spinal/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/physiology
18.
J Invest Dermatol ; 138(4): 946-956, 2018 04.
Article in English | MEDLINE | ID: mdl-29138055

ABSTRACT

In the skin, Merkel cells connect with keratinocytes and Aß nerve fibers to form a touch receptor that functions as a slow adapting mechanoreceptor (slow adapting type 1). In human and mouse Merkel cells, we observed an increased concentration of intracellular Ca2+ ions in response to cold temperature and transient receptor potential melastatine 8 (TRPM8) ion channel agonists. A reduction in the response to cooling and TRPM8 agonists occurred after the addition of TRPM8 antagonists, as well as in TRPM8 knockout mice. Cold temperature and TRPM8 agonists also induced a current that was inhibited by a TRPM8 antagonist. Our results indicate that Merkel cells sense cooling through TRPM8 channels. We hypothesized that cooling modulates the slow adapting type 1 receptor response. Cooling mouse skin to 22°C reduced the slow adapting type 1 receptor discharge frequency. Interestingly, we observed no such reduction in TRPM8 knockout mice. Similarly, in human skin, a temperature of 22°C applied to the slow adapting type 1 receptive field reduced the spiking discharge. Altogether, our results indicate that Merkel cells are polymodal sensory cells that respond to mild cold stimuli through the activation of TRPM8 channels. Thermal activation of Merkel cells, and possibly other TRPM8-expressing non-neuronal cells, such as keratinocytes, potentially adapts the discharge of slow adapting type 1 receptors during cooling.


Subject(s)
Gene Expression Regulation , Merkel Cells/metabolism , RNA, Messenger/genetics , TRPM Cation Channels/genetics , Animals , Cells, Cultured , Cold Temperature , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mechanoreceptors/metabolism , Merkel Cells/cytology , Mice , Mice, Knockout , Models, Animal , TRPM Cation Channels/biosynthesis
20.
Nat Commun ; 6: 7223, 2015 May 26.
Article in English | MEDLINE | ID: mdl-26008989

ABSTRACT

Piezo1 and Piezo2 encode mechanically activated cation channels that function as mechanotransducers involved in vascular system development and touch sensing, respectively. Structural features of Piezos remain unknown. Mouse Piezo1 is bioinformatically predicted to have 30-40 transmembrane (TM) domains. Here, we find that nine of the putative inter-transmembrane regions are accessible from the extracellular side. We use chimeras between mPiezo1 and dPiezo to show that ion-permeation properties are conferred by C-terminal region. We further identify a glutamate residue within a conserved region adjacent to the last two putative TM domains of the protein, that when mutated, affects unitary conductance and ion selectivity, and modulates pore block. We propose that this amino acid is either in the pore or closely associates with the pore. Our results describe important structural motifs of this channel family and lay the groundwork for a mechanistic understanding of how Piezos are mechanically gated and conduct ions.


Subject(s)
Drosophila Proteins/chemistry , Ion Channels/chemistry , Amino Acid Sequence , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mice , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...