Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.981
Filter
Add more filters

Publication year range
1.
Cell ; 184(20): 5138-5150.e12, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34496225

ABSTRACT

Many transient receptor potential (TRP) channels respond to diverse stimuli and conditionally conduct small and large cations. Such functional plasticity is presumably enabled by a uniquely dynamic ion selectivity filter that is regulated by physiological agents. What is currently missing is a "photo series" of intermediate structural states that directly address this hypothesis and reveal specific mechanisms behind such dynamic channel regulation. Here, we exploit cryoelectron microscopy (cryo-EM) to visualize conformational transitions of the capsaicin receptor, TRPV1, as a model to understand how dynamic transitions of the selectivity filter in response to algogenic agents, including protons, vanilloid agonists, and peptide toxins, permit permeation by small and large organic cations. These structures also reveal mechanisms governing ligand binding substates, as well as allosteric coupling between key sites that are proximal to the selectivity filter and cytoplasmic gate. These insights suggest a general framework for understanding how TRP channels function as polymodal signal integrators.


Subject(s)
TRPV Cation Channels/chemistry , TRPV Cation Channels/metabolism , Allosteric Regulation , Cell Membrane Permeability/drug effects , Cryoelectron Microscopy , Diterpenes/pharmacology , HEK293 Cells , Humans , Ion Channel Gating , Lipids/chemistry , Meglumine/pharmacology , Models, Molecular , Protein Binding , Protein Conformation , Protons , TRPV Cation Channels/agonists
2.
Cell ; 184(2): 422-440.e17, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450207

ABSTRACT

Itch is an evolutionarily conserved sensation that facilitates expulsion of pathogens and noxious stimuli from the skin. However, in organ failure, cancer, and chronic inflammatory disorders such as atopic dermatitis (AD), itch becomes chronic, intractable, and debilitating. In addition to chronic itch, patients often experience intense acute itch exacerbations. Recent discoveries have unearthed the neuroimmune circuitry of itch, leading to the development of anti-itch treatments. However, mechanisms underlying acute itch exacerbations remain overlooked. Herein, we identify that a large proportion of patients with AD harbor allergen-specific immunoglobulin E (IgE) and exhibit a propensity for acute itch flares. In mice, while allergen-provoked acute itch is mediated by the mast cell-histamine axis in steady state, AD-associated inflammation renders this pathway dispensable. Instead, a previously unrecognized basophil-leukotriene (LT) axis emerges as critical for acute itch flares. By probing fundamental itch mechanisms, our study highlights a basophil-neuronal circuit that may underlie a variety of neuroimmune processes.


Subject(s)
Basophils/pathology , Neurons/pathology , Pruritus/pathology , Acute Disease , Allergens/immunology , Animals , Chronic Disease , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Histamine/metabolism , Humans , Immunoglobulin E/immunology , Inflammation/pathology , Leukotrienes/metabolism , Mast Cells/immunology , Mice, Inbred C57BL , Phenotype , Pruritus/immunology , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
3.
Cell ; 184(14): 3762-3773.e10, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34133943

ABSTRACT

Sneezing is a vital respiratory reflex frequently associated with allergic rhinitis and viral respiratory infections. However, its neural circuit remains largely unknown. A sneeze-evoking region was discovered in both cat and human brainstems, corresponding anatomically to the central recipient zone of nasal sensory neurons. Therefore, we hypothesized that a neuronal population postsynaptic to nasal sensory neurons mediates sneezing in this region. By screening major presynaptic neurotransmitters/neuropeptides released by nasal sensory neurons, we found that neuromedin B (NMB) peptide is essential for signaling sneezing. Ablation of NMB-sensitive postsynaptic neurons in the sneeze-evoking region or deficiency in NMB receptor abolished the sneezing reflex. Remarkably, NMB-sensitive neurons further project to the caudal ventral respiratory group (cVRG). Chemical activation of NMB-sensitive neurons elicits action potentials in cVRG neurons and leads to sneezing behavior. Our study delineates a peptidergic pathway mediating sneezing, providing molecular insights into the sneezing reflex arc.


Subject(s)
Brain Stem/physiopathology , Neuropeptides/metabolism , Nose/physiopathology , Reflex/physiology , Sneezing/physiology , Animals , Disease Models, Animal , Hypersensitivity/physiopathology , Male , Mice, Inbred C57BL , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Neurons/metabolism , RNA, Small Interfering/metabolism , Sensory Receptor Cells/physiology , TRPV Cation Channels/metabolism , Video Recording
4.
Cell ; 178(4): 919-932.e14, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353219

ABSTRACT

Cutaneous TRPV1+ neurons directly sense noxious stimuli, inflammatory cytokines, and pathogen-associated molecules and are required for innate immunity against some skin pathogens. Important unanswered questions are whether TRPV1+ neuron activation in isolation is sufficient to initiate innate immune responses and what is the biological function for TRPV1+ neuron-initiated immune responses. We used TRPV1-Ai32 optogenetic mice and cutaneous light stimulation to activate cutaneous neurons in the absence of tissue damage or pathogen-associated products. We found that TRPV1+ neuron activation was sufficient to elicit a local type 17 immune response that augmented host defense to C. albicans and S. aureus. Moreover, local neuron activation elicited type 17 responses and augmented host defense at adjacent, unstimulated skin through a nerve reflex arc. These data show the sufficiency of TRPV1+ neuron activation for host defense and demonstrate the existence of functional anticipatory innate immunity at sites adjacent to infection that depends on antidromic neuron activation.


Subject(s)
Immunity, Innate/immunology , Interleukin-23/metabolism , Interleukin-6/metabolism , Sensory Receptor Cells/immunology , Skin/immunology , TRPV Cation Channels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Candida albicans/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics/methods , Skin/microbiology , Staphylococcus aureus/immunology , TRPV Cation Channels/genetics
5.
Cell ; 173(5): 1083-1097.e22, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29754819

ABSTRACT

The nervous system, the immune system, and microbial pathogens interact closely at barrier tissues. Here, we find that a bacterial pathogen, Streptococcus pyogenes, hijacks pain and neuronal regulation of the immune response to promote bacterial survival. Necrotizing fasciitis is a life-threatening soft tissue infection in which "pain is out of proportion" to early physical manifestations. We find that S. pyogenes, the leading cause of necrotizing fasciitis, secretes streptolysin S (SLS) to directly activate nociceptor neurons and produce pain during infection. Nociceptors, in turn, release the neuropeptide calcitonin gene-related peptide (CGRP) into infected tissues, which inhibits the recruitment of neutrophils and opsonophagocytic killing of S. pyogenes. Botulinum neurotoxin A and CGRP antagonism block neuron-mediated suppression of host defense, thereby preventing and treating S. pyogenes necrotizing infection. We conclude that targeting the peripheral nervous system and blocking neuro-immune communication is a promising strategy to treat highly invasive bacterial infections. VIDEO ABSTRACT.


Subject(s)
Neurons/metabolism , Neutrophils/metabolism , Streptococcal Infections/pathology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Botulinum Toxins, Type A/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Diterpenes/pharmacology , Fasciitis, Necrotizing/etiology , Fasciitis, Necrotizing/pathology , Fasciitis, Necrotizing/veterinary , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neutrophils/immunology , Pain/etiology , Signal Transduction , Skin/metabolism , Skin/pathology , Streptococcal Infections/complications , Streptococcal Infections/veterinary , Streptococcus pyogenes/metabolism , Streptolysins/immunology , Streptolysins/metabolism , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
6.
Cell ; 174(2): 481-496.e19, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30007419

ABSTRACT

Dopamine (DA) is a central monoamine neurotransmitter involved in many physiological and pathological processes. A longstanding yet largely unmet goal is to measure DA changes reliably and specifically with high spatiotemporal precision, particularly in animals executing complex behaviors. Here, we report the development of genetically encoded GPCR-activation-based-DA (GRABDA) sensors that enable these measurements. In response to extracellular DA, GRABDA sensors exhibit large fluorescence increases (ΔF/F0 ∼90%) with subcellular resolution, subsecond kinetics, nanomolar to submicromolar affinities, and excellent molecular specificity. GRABDA sensors can resolve a single-electrical-stimulus-evoked DA release in mouse brain slices and detect endogenous DA release in living flies, fish, and mice. In freely behaving mice, GRABDA sensors readily report optogenetically elicited nigrostriatal DA release and depict dynamic mesoaccumbens DA signaling during Pavlovian conditioning or during sexual behaviors. Thus, GRABDA sensors enable spatiotemporally precise measurements of DA dynamics in a variety of model organisms while exhibiting complex behaviors.


Subject(s)
Dopamine/analysis , Drosophila/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Behavior, Animal , Dopamine/metabolism , Female , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neurons/cytology , Neurons/metabolism , Optogenetics/methods , Receptors, G-Protein-Coupled/genetics , TRPV Cation Channels/genetics , Zebrafish Proteins/genetics
7.
Nat Immunol ; 20(11): 1435-1443, 2019 11.
Article in English | MEDLINE | ID: mdl-31591569

ABSTRACT

Allergic skin diseases, such as atopic dermatitis, are clinically characterized by severe itching and type 2 immunity-associated hypersensitivity to widely distributed allergens, including those derived from house dust mites (HDMs). Here we found that HDMs with cysteine protease activity directly activated peptidergic nociceptors, which are neuropeptide-producing nociceptive sensory neurons that express the ion channel TRPV1 and Tac1, the gene encoding the precursor for the neuropeptide substance P. Intravital imaging and genetic approaches indicated that HDM-activated nociceptors drive the development of allergic skin inflammation by inducing the degranulation of mast cells contiguous to such nociceptors, through the release of substance P and the activation of the cationic molecule receptor MRGPRB2 on mast cells. These data indicate that, after exposure to HDM allergens, activation of TRPV1+Tac1+ nociceptor-MRGPRB2+ mast cell sensory clusters represents a key early event in the development of allergic skin reactions.


Subject(s)
Allergens/immunology , Dermatitis, Atopic/immunology , Mast Cells/immunology , Nociceptors/immunology , Pyroglyphidae/immunology , Animals , Cell Communication/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Female , Humans , Male , Mast Cells/metabolism , Mice, Knockout , Nociceptors/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin/cytology , Skin/immunology , TRPV Cation Channels/metabolism , Tachykinins/genetics , Tachykinins/metabolism
8.
Mol Cell ; 83(4): 556-573.e7, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36696898

ABSTRACT

The protection of DNA replication forks under stress is essential for genome maintenance and cancer suppression. One mechanism of fork protection involves an elevation in intracellular Ca2+ ([Ca2+]i), which in turn activates CaMKK2 and AMPK to prevent uncontrolled fork processing by Exo1. How replication stress triggers [Ca2+]i elevation is unclear. Here, we report a role of cytosolic self-DNA (cytosDNA) and the ion channel TRPV2 in [Ca2+]i induction and fork protection. Replication stress leads to the generation of ssDNA and dsDNA species that, upon translocation into cytoplasm, trigger the activation of the sensor protein cGAS and the production of cGAMP. The subsequent binding of cGAMP to STING causes its dissociation from TRPV2, leading to TRPV2 derepression and Ca2+ release from the ER, which in turn activates the downstream signaling cascade to prevent fork degradation. This Ca2+-dependent genome protection pathway is also activated in response to replication stress caused by oncogene activation.


Subject(s)
DNA , Nucleotidyltransferases , DNA/genetics , DNA/metabolism , DNA Replication , DNA, Single-Stranded , Membrane Proteins , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , TRPV Cation Channels
9.
Cell ; 160(4): 759-770, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679765

ABSTRACT

Sensitization of the capsaicin receptor TRPV1 is central to the initiation of pathological forms of pain, and multiple signaling cascades are known to enhance TRPV1 activity under inflammatory conditions. How might detrimental escalation of TRPV1 activity be counteracted? Using a genetic-proteomic approach, we identify the GABAB1 receptor subunit as bona fide inhibitor of TRPV1 sensitization in the context of diverse inflammatory settings. We find that the endogenous GABAB agonist, GABA, is released from nociceptive nerve terminals, suggesting an autocrine feedback mechanism limiting TRPV1 sensitization. The effect of GABAB on TRPV1 is independent of canonical G protein signaling and rather relies on close juxtaposition of the GABAB1 receptor subunit and TRPV1. Activating the GABAB1 receptor subunit does not attenuate normal functioning of the capsaicin receptor but exclusively reverts its sensitized state. Thus, harnessing this mechanism for anti-pain therapy may prevent adverse effects associated with currently available TRPV1 blockers.


Subject(s)
Autocrine Communication , Neurons/metabolism , Pain/metabolism , Receptors, GABA-B/metabolism , TRPV Cation Channels/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cells, Cultured , Feedback , Female , Male , Mice, Inbred C57BL , Mice, Transgenic
10.
Cell ; 160(6): 1222-32, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25748653

ABSTRACT

The nervous system evolved to coordinate flexible goal-directed behaviors by integrating interoceptive and sensory information. Hypothalamic Agrp neurons are known to be crucial for feeding behavior. Here, however, we show that these neurons also orchestrate other complex behaviors in adult mice. Activation of Agrp neurons in the absence of food triggers foraging and repetitive behaviors, which are reverted by food consumption. These stereotypic behaviors that are triggered by Agrp neurons are coupled with decreased anxiety. NPY5 receptor signaling is necessary to mediate the repetitive behaviors after Agrp neuron activation while having minor effects on feeding. Thus, we have unmasked a functional role for Agrp neurons in controlling repetitive behaviors mediated, at least in part, by neuropeptidergic signaling. The findings reveal a new set of behaviors coupled to the energy homeostasis circuit and suggest potential therapeutic avenues for diseases with stereotypic behaviors.


Subject(s)
Hypothalamus/physiology , Neurons/physiology , Stereotyped Behavior , Agouti-Related Protein/metabolism , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Capsaicin/administration & dosage , Feeding Behavior/drug effects , Female , GABA Antagonists/administration & dosage , Hypothalamus/cytology , Male , Neurons/classification , Stereotyped Behavior/drug effects , TRPV Cation Channels/metabolism
11.
Immunity ; 53(2): 371-383.e5, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32673566

ABSTRACT

Cutaneous wound healing is associated with the unpleasant sensation of itching. Here we investigated the mechanisms underlying this type of itch, focusing on the contribution of soluble factors released during healing. We found high amounts of interleukin 31 (IL-31) in skin wound tissue during the peak of itch responses. Il31-/- mice lacked wound-induced itch responses. IL-31 was released by dermal conventional type 2 dendritic cells (cDC2s) recruited to wounds and increased itch sensory neuron sensitivity. Transfer of cDC2s isolated from late-stage wounds into healthy skin was sufficient to induce itching in a manner dependent on IL-31 expression. Addition of the cytokine TGF-ß1, which promotes wound healing, to dermal DCs in vitro was sufficient to induce Il31 expression, and Tgfbr1f/f CD11c-Cre mice exhibited reduced scratching and decreased Il31 expression in wounds in vivo. Thus, cDC2s promote itching during skin would healing via a TGF-ß-IL-31 axis with implications for treatment of wound itching.


Subject(s)
Interleukins/metabolism , Langerhans Cells/physiology , Pruritus/pathology , Sensory Receptor Cells/physiology , Transforming Growth Factor beta1/metabolism , Animals , Female , Humans , Interleukins/genetics , Langerhans Cells/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Interleukin/metabolism , Skin/cytology , Skin/growth & development , Skin/injuries , TRPV Cation Channels/metabolism , Wound Healing/physiology
12.
Cell ; 157(5): 1004-6, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855937

ABSTRACT

Riera et al. identify a neuroendocrine circuit that controls longevity and the age-dependent onset of metabolic decline via the pain-transducing channel TRPV1. Thus, pharmacological inhibition of TRPV1 may provide a new approach to treat not only metabolic disorders but also a broader range of age-related pathologies.


Subject(s)
Longevity , Signal Transduction , TRPV Cation Channels/metabolism , Animals , Female , Male
13.
Cell ; 157(5): 1023-36, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855942

ABSTRACT

The sensation of pain is associated with increased mortality, but it is unknown whether pain perception can directly affect aging. We find that mice lacking TRPV1 pain receptors are long-lived, displaying a youthful metabolic profile at old age. Loss of TRPV1 inactivates a calcium-signaling cascade that ends in the nuclear exclusion of the CREB-regulated transcriptional coactivator CRTC1 within pain sensory neurons originating from the spinal cord. In long-lived TRPV1 knockout mice, CRTC1 nuclear exclusion decreases production of the neuropeptide CGRP from sensory endings innervating the pancreatic islets, subsequently promoting insulin secretion and metabolic health. In contrast, CGRP homeostasis is disrupted with age in wild-type mice, resulting in metabolic decline. We show that pharmacologic inactivation of CGRP receptors in old wild-type animals can restore metabolic health. These data suggest that ablation of select pain sensory receptors or the inhibition of CGRP are associated with increased metabolic health and control longevity.


Subject(s)
Longevity , Signal Transduction , TRPV Cation Channels/metabolism , Animals , CREB-Binding Protein/metabolism , Caenorhabditis elegans , Cells, Cultured , Diet , Female , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurons/metabolism , Nociceptors/metabolism , TRPV Cation Channels/genetics , Transcription Factors/metabolism
14.
Nature ; 621(7977): 206-214, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648856

ABSTRACT

Transient receptor potential (TRP) channels are a large, eukaryotic ion channel superfamily that control diverse physiological functions, and therefore are attractive drug targets1-5. More than 210 structures from more than 20 different TRP channels have been determined, and all are tetramers4. Despite this wealth of structures, many aspects concerning TRPV channels remain poorly understood, including the pore-dilation phenomenon, whereby prolonged activation leads to increased conductance, permeability to large ions and loss of rectification6,7. Here, we used high-speed atomic force microscopy (HS-AFM) to analyse membrane-embedded TRPV3 at the single-molecule level and discovered a pentameric state. HS-AFM dynamic imaging revealed transience and reversibility of the pentamer in dynamic equilibrium with the canonical tetramer through membrane diffusive protomer exchange. The pentamer population increased upon diphenylboronic anhydride (DPBA) addition, an agonist that has been shown to induce TRPV3 pore dilation. On the basis of these findings, we designed a protein production and data analysis pipeline that resulted in a cryogenic-electron microscopy structure of the TRPV3 pentamer, showing an enlarged pore compared to the tetramer. The slow kinetics to enter and exit the pentameric state, the increased pentamer formation upon DPBA addition and the enlarged pore indicate that the pentamer represents the structural correlate of pore dilation. We thus show membrane diffusive protomer exchange as an additional mechanism for structural changes and conformational variability. Overall, we provide structural evidence for a non-canonical pentameric TRP-channel assembly, laying the foundation for new directions in TRP channel research.


Subject(s)
Protein Multimerization , TRPV Cation Channels , Anhydrides/chemistry , Anhydrides/pharmacology , Data Analysis , Diffusion , Protein Subunits/chemistry , Protein Subunits/drug effects , Protein Subunits/metabolism , TRPV Cation Channels/chemistry , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism , TRPV Cation Channels/ultrastructure , Microscopy, Atomic Force , Molecular Targeted Therapy , Cryoelectron Microscopy , Protein Structure, Quaternary/drug effects , Protein Multimerization/drug effects
15.
EMBO J ; 43(11): 2264-2290, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38671253

ABSTRACT

Transient receptor potential (TRP) ion channels are involved in the surveillance or regulation of the acid-base balance. Here, we demonstrate that weak carbonic acids, including acetic acid, lactic acid, and CO2 activate and sensitize TRPV2 through a mechanism requiring permeation through the cell membrane. TRPV2 channels in cell-free inside-out patches maintain weak acid-sensitivity, but protons applied on either side of the membrane do not induce channel activation or sensitization. The involvement of proton modulation sites for weak acid-sensitivity was supported by the identification of titratable extracellular (Glu495, Glu561) and intracellular (His521) residues on a cryo-EM structure of rat TRPV2 (rTRPV2) treated with acetic acid. Molecular dynamics simulations as well as patch clamp experiments on mutant rTRPV2 constructs confirmed that these residues are critical for weak acid-sensitivity. We also demonstrate that the pore residue Glu609 dictates an inhibition of weak acid-induced currents by extracellular calcium. Finally, TRPV2-expression in HEK293 cells is associated with an increased weak acid-induced cytotoxicity. Together, our data provide new insights into weak acids as endogenous modulators of TRPV2.


Subject(s)
TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/chemistry , Humans , HEK293 Cells , Animals , Rats , Molecular Dynamics Simulation , Cryoelectron Microscopy , Calcium/metabolism , Patch-Clamp Techniques , Acids/metabolism
16.
Immunity ; 51(3): 426-428, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533055

ABSTRACT

The skin is densely innervated with nociceptive neurons specialized in detecting noxious and painful stimuli. In a recent issue of Cell, Cohen et al. report that activation of cutaneous nociceptive neurons leads to a nerve-reflex action that is sufficient to provide a danger signal that triggers regional immunity to fight a microbial challenge.


Subject(s)
Nociceptors , Pain , Humans , Neurons , Reflex , Skin , TRPV Cation Channels
17.
Nature ; 611(7935): 365-373, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323783

ABSTRACT

Cells respond to physical stimuli, such as stiffness1, fluid shear stress2 and hydraulic pressure3,4. Extracellular fluid viscosity is a key physical cue that varies under physiological and pathological conditions, such as cancer5. However, its influence on cancer biology and the mechanism by which cells sense and respond to changes in viscosity are unknown. Here we demonstrate that elevated viscosity counterintuitively increases the motility of various cell types on two-dimensional surfaces and in confinement, and increases cell dissemination from three-dimensional tumour spheroids. Increased mechanical loading imposed by elevated viscosity induces an actin-related protein 2/3 (ARP2/3)-complex-dependent dense actin network, which enhances Na+/H+ exchanger 1 (NHE1) polarization through its actin-binding partner ezrin. NHE1 promotes cell swelling and increased membrane tension, which, in turn, activates transient receptor potential cation vanilloid 4 (TRPV4) and mediates calcium influx, leading to increased RHOA-dependent cell contractility. The coordinated action of actin remodelling/dynamics, NHE1-mediated swelling and RHOA-based contractility facilitates enhanced motility at elevated viscosities. Breast cancer cells pre-exposed to elevated viscosity acquire TRPV4-dependent mechanical memory through transcriptional control of the Hippo pathway, leading to increased migration in zebrafish, extravasation in chick embryos and lung colonization in mice. Cumulatively, extracellular viscosity is a physical cue that regulates both short- and long-term cellular processes with pathophysiological relevance to cancer biology.


Subject(s)
Cell Movement , Extracellular Fluid , Neoplasm Metastasis , Neoplasms , Viscosity , Animals , Chick Embryo , Mice , Actins/metabolism , Extracellular Fluid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Sodium-Hydrogen Exchangers/metabolism , TRPV Cation Channels , Zebrafish/metabolism , Neoplasm Metastasis/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Hippo Signaling Pathway , Spheroids, Cellular/pathology , Actin-Related Protein 2-3 Complex , rhoA GTP-Binding Protein , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Lung/pathology
18.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958798

ABSTRACT

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Subject(s)
Colon/physiology , Gastrointestinal Motility , Macrophages/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction , TRPV Cation Channels/metabolism , Animals , CX3C Chemokine Receptor 1/metabolism , Colon/physiopathology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Dinoprostone/analysis , Dinoprostone/metabolism , Female , Gastric Mucosa/cytology , Gene Expression , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscle Contraction , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
19.
Cell ; 151(1): 96-110, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021218

ABSTRACT

PGC1α is a key transcriptional coregulator of oxidative metabolism and thermogenesis. Through a high-throughput chemical screen, we found that molecules antagonizing the TRPVs (transient receptor potential vanilloid), a family of ion channels, induced PGC1α expression in adipocytes. In particular, TRPV4 negatively regulated the expression of PGC1α, UCP1, and cellular respiration. Additionally, it potently controlled the expression of multiple proinflammatory genes involved in the development of insulin resistance. Mice with a null mutation for TRPV4 or wild-type mice treated with a TRPV4 antagonist showed elevated thermogenesis in adipose tissues and were protected from diet-induced obesity, adipose inflammation, and insulin resistance. This role of TRPV4 as a cell-autonomous mediator for both the thermogenic and proinflammatory programs in adipocytes could offer a target for treating obesity and related metabolic diseases.


Subject(s)
Energy Metabolism , TRPV Cation Channels/metabolism , Thermogenesis , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Female , Gene Knockdown Techniques , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Trans-Activators/metabolism , Transcription Factors , Uncoupling Protein 1
20.
Proc Natl Acad Sci U S A ; 121(15): e2322135121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38568964

ABSTRACT

Endothelial cells (ECs) line the wall of blood vessels and regulate arterial contractility to tune regional organ blood flow and systemic pressure. Chloride (Cl-) is the most abundant anion in ECs and the Cl- sensitive With-No-Lysine (WNK) kinase is expressed in this cell type. Whether intracellular Cl- signaling and WNK kinase regulate EC function to alter arterial contractility is unclear. Here, we tested the hypothesis that intracellular Cl- signaling in ECs regulates arterial contractility and examined the signaling mechanisms involved, including the participation of WNK kinase. Our data obtained using two-photon microscopy and cell-specific inducible knockout mice indicated that acetylcholine, a prototypical vasodilator, stimulated a rapid reduction in intracellular Cl- concentration ([Cl-]i) due to the activation of TMEM16A, a Cl- channel, in ECs of resistance-size arteries. TMEM16A channel-mediated Cl- signaling activated WNK kinase, which phosphorylated its substrate proteins SPAK and OSR1 in ECs. OSR1 potentiated transient receptor potential vanilloid 4 (TRPV4) currents in a kinase-dependent manner and required a conserved binding motif located in the channel C terminus. Intracellular Ca2+ signaling was measured in four dimensions in ECs using a high-speed lightsheet microscope. WNK kinase-dependent activation of TRPV4 channels increased local intracellular Ca2+ signaling in ECs and produced vasodilation. In summary, we show that TMEM16A channel activation reduces [Cl-]i, which activates WNK kinase in ECs. WNK kinase phosphorylates OSR1 which then stimulates TRPV4 channels to produce vasodilation. Thus, TMEM16A channels regulate intracellular Cl- signaling and WNK kinase activity in ECs to control arterial contractility.


Subject(s)
Chlorides , Protein Serine-Threonine Kinases , Mice , Animals , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Chlorides/metabolism , Endothelial Cells/metabolism , TRPV Cation Channels/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL