Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 318, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031603

ABSTRACT

Lung emphysema and chronic bronchitis are the two most common causes of chronic obstructive pulmonary disease. Excess macrophage elastase MMP-12, which is predominantly secreted from alveolar macrophages, is known to mediate the development of lung injury and emphysema. Here, we discovered the endolysosomal cation channel mucolipin 3 (TRPML3) as a regulator of MMP-12 reuptake from broncho-alveolar fluid, driving in two independently generated Trpml3-/- mouse models enlarged lung injury, which is further exacerbated after elastase or tobacco smoke treatment. Mechanistically, using a Trpml3IRES-Cre/eR26-τGFP reporter mouse model, transcriptomics, and endolysosomal patch-clamp experiments, we show that in the lung TRPML3 is almost exclusively expressed in alveolar macrophages, where its loss leads to defects in early endosomal trafficking and endocytosis of MMP-12. Our findings suggest that TRPML3 represents a key regulator of MMP-12 clearance by alveolar macrophages and may serve as therapeutic target for emphysema and chronic obstructive pulmonary disease.


Subject(s)
Macrophages, Alveolar/enzymology , Matrix Metalloproteinase 12/metabolism , Pancreatic Elastase/metabolism , Pulmonary Emphysema/enzymology , Transient Receptor Potential Channels/deficiency , Animals , Disease Models, Animal , Endosomes/metabolism , Female , Humans , Lung/enzymology , Matrix Metalloproteinase 12/genetics , Mice , Mice, Knockout , Pancreatic Elastase/genetics , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Transient Receptor Potential Channels/genetics
2.
Cell Calcium ; 79: 80-88, 2019 05.
Article in English | MEDLINE | ID: mdl-30889511

ABSTRACT

The triple-negative breast cancer (TNBC) that comprises approximately 10%-20% of breast cancers is an aggressive subtype lacking effective therapeutics. Among various signaling pathways, mTORC1 and purinergic signals have emerged as potentially fruitful targets for clinical therapy of TNBC. Unfortunately, drugs targeting these signaling pathways do not successfully inhibit the progression of TNBC, partially due to the fact that these signaling pathways are essential for the function of all types of cells. In this study, we report that TRPML1 is specifically upregulated in TNBCs and that its genetic downregulation and pharmacological inhibition suppress the growth of TNBC. Mechanistically, we demonstrate that TRPML1 regulates TNBC development, at least partially, through controlling mTORC1 activity and the release of lysosomal ATP. Because TRPML1 is specifically activated by cellular stresses found in tumor microenvironments, antagonists of TRPML1 could represent anticancer drugs with enhanced specificity and potency. Our findings are expected to have a major impact on drug targeting of TNBCs.


Subject(s)
Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction , Transient Receptor Potential Channels/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Calcium/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Transient Receptor Potential Channels/deficiency , Triple Negative Breast Neoplasms/pathology
3.
Sci Immunol ; 2(16)2017 10 27.
Article in English | MEDLINE | ID: mdl-29079589

ABSTRACT

Dendritic cells (DCs) patrol their environment by linking antigen acquisition by macropinocytosis to cell locomotion. DC activation upon bacterial sensing inhibits macropinocytosis and increases DC migration, thus promoting the arrival of DCs to lymph nodes for antigen presentation to T cells. The signaling events that trigger such changes are not fully understood. We show that lysosome signaling plays a critical role in this process. Upon bacterial sensing, lysosomal calcium is released by the ionic channel TRPML1 (transient receptor potential cation channel, mucolipin subfamily, member 1), which activates the actin-based motor protein myosin II at the cell rear, promoting fast and directional migration. Lysosomal calcium further induces the activation of the transcription factor EB (TFEB), which translocates to the nucleus to maintain TRPML1 expression. We found that the TRPML1-TFEB axis results from the down-regulation of macropinocytosis after bacterial sensing by DCs. Lysosomal signaling therefore emerges as a hitherto unexpected link between macropinocytosis, actomyosin cytoskeleton organization, and DC migration.


Subject(s)
Cell Movement , Dendritic Cells/immunology , Lysosomes/metabolism , Signal Transduction , Animals , Antigen Presentation , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Calcium/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Dendritic Cells/physiology , Down-Regulation , Lysosomes/immunology , Mice , Myosin Type II/genetics , Myosin Type II/metabolism , Pinocytosis , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
4.
eNeuro ; 4(1)2017.
Article in English | MEDLINE | ID: mdl-28303259

ABSTRACT

The nonselective cation channel transient receptor potential ankyrin 1 (TRPA1) is known to be a key contributor to both somatosensation and pain. Recent studies have implicated TRPA1 in additional physiologic functions and have also suggested that TRPA1 is expressed in nonneuronal tissues. Thus, it has become necessary to resolve the importance of TRPA1 expressed in primary sensory neurons, particularly since previous research has largely used global knock-out animals and chemical TRPA1 antagonists. We therefore sought to isolate the physiological relevance of TRPA1 specifically within sensory neurons. To accomplish this, we used Advillin-Cre mice, in which the promoter for Advillin is used to drive expression of Cre recombinase specifically within sensory neurons. These Advillin-Cre mice were crossed with Trpa1fl/fl mice to generate sensory neuron-specific Trpa1 knock-out mice. Here, we show that tissue-specific deletion of TRPA1 from sensory neurons produced strong deficits in behavioral sensitivity to mechanical stimulation, while sensitivity to cold and heat stimuli remained intact. The mechanical sensory deficit was incomplete compared to the mechanosensory impairment of TRPA1 global knock-out mice, in line with the incomplete (∼80%) elimination of TRPA1 from sensory neurons in the tissue-specific Advillin-Cre knock-out mice. Equivalent findings were observed in tissue-specific knock-out animals originating from two independently-generated Advillin-Cre lines. As such, our results show that sensory neuron TRPA1 is required for mechanical, but not cold, responsiveness in noninjured skin.


Subject(s)
Nociception/physiology , Sensory Receptor Cells/metabolism , Sensory Thresholds/physiology , Skin/innervation , Touch/physiology , Transient Receptor Potential Channels/deficiency , Animals , Calcium/metabolism , Cells, Cultured , Cold Temperature , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hot Temperature , Lumbar Vertebrae , Male , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nociception/drug effects , Promoter Regions, Genetic , RNA, Messenger/metabolism , Sensory Receptor Cells/drug effects , Sensory Thresholds/drug effects , Skin/metabolism , TRPA1 Cation Channel , Touch/drug effects , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/genetics
5.
J Bone Miner Res ; 32(2): 385-396, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27589205

ABSTRACT

Lysosomal Ca2+ emerges as a critical component of receptor-evoked Ca2+ signaling and plays a crucial role in many lysosomal and physiological functions. Lysosomal Ca2+ release is mediated by the transient receptor potential (TRP) family member TRPML1, mutations that cause the lysosomal storage disease mucolipidosis type 4. Lysosomes play a key role in osteoclast function. However, nothing is known about the role of lysosomal Ca2+ signaling in osteoclastogenesis and bone metabolism. In this study, we addressed this knowledge gap by studying the role of lysosomal Ca2+ signaling in osteoclastogenesis, osteoclast and osteoblast functions, and bone homeostasis in vivo. We manipulated lysosomal Ca2+ signaling by acute knockdown of TRPML1, deletion of TRPML1 in mice, pharmacological inhibition of lysosomal Ca2+ influx, and depletion of lysosomal Ca2+ storage using the TRPML agonist ML-SA1. We found that knockdown and deletion of TRPML1, although it did not have an apparent effect on osteoblast differentiation and bone formation, markedly attenuated osteoclast function, RANKL-induced cytosolic Ca2+ oscillations, inhibited activation of NFATc1 and osteoclastogenesis-controlling genes, suppressed the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs), and markedly reduced the differentiation of bone marrow-derived macrophages into osteoclasts. Moreover, deletion of TRPML1 resulted in enlarged lysosomes, inhibition of lysosomal secretion, and attenuated the resorptive activity of mature osteoclasts. Notably, depletion of lysosomal Ca2+ with ML-SA1 similarly abrogated RANKL-induced Ca2+ oscillations and MNC formation. Deletion of TRPML1 in mice reduced the TRAP-positive bone surfaces and impaired bone remodeling, resulting in prominent osteopetrosis. These findings demonstrate the essential role of lysosomal Ca2+ signaling in osteoclast differentiation and mature osteoclast function, which play key roles in bone homeostasis. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Remodeling , Calcium Signaling , Lysosomes/metabolism , Osteoclasts/metabolism , Osteogenesis , Animals , Bone Remodeling/drug effects , Bone Resorption/pathology , Calcium Signaling/drug effects , Cell Size , Gene Deletion , Lysosomes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase/metabolism , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/metabolism
6.
Nutrients ; 8(10)2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27735854

ABSTRACT

TRPA1 is a ligand-activated cation channel found in the intestine and other tissues. Components of food that stimulate TRPA1 receptors (phytonutrients) include allyl isothiocyanate, cinnamaldehyde and linalool, but these may also act at other receptors. Cells lining the intestinal mucosa are immunoreactive for TRPA1 and Trpa1 mRNA occurs in mucosal extracts, suggesting that the TRPA1 receptor is the target for these agonists. However, in situ hybridisation reveals Trpa1 expression in 5-HT containing enteroendocrine cells, not enterocytes. TRPA1 agonists evoke mucosal secretion, which may be indirect (through release of 5-HT) or direct by activation of enterocytes. We investigated effects of the phytonutrients on transmucosal ion currents in mouse duodenum and colon, and the specificity of the phytonutrients in cells transfected with Trpa1, and in Trpa1-deficient mice. The phytonutrients increased currents in the duodenum with the relative potencies: allyl isothiocyanate (AITC) > cinnamaldehyde > linalool (0.1 to 300 µM). The rank order was similar in the colon, but linalool was ineffective. Responses to AITC were reduced by the TRPA1 antagonist HC-030031 (100 µM), and were greatly diminished in Trpa1-/- duodenum and colon. Responses were not reduced by tetrodotoxin, 5-HT receptor antagonists, or atropine, but inhibition of prostaglandin synthesis reduced responses. Thus, functional TRPA1 channels are expressed by enterocytes of the duodenum and colon. Activation of enterocyte TRPA1 by food components has the potential to facilitate nutrient absorption.


Subject(s)
Intestinal Mucosa/physiology , Phytochemicals/pharmacology , Transient Receptor Potential Channels/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Acyclic Monoterpenes , Animals , Calcium/metabolism , Colon/physiology , Duodenum/physiology , Electrophysiological Phenomena , Enterocytes/drug effects , Enterocytes/metabolism , Food , Gene Expression , HEK293 Cells , Humans , Intestinal Mucosa/chemistry , Isothiocyanates/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , TRPA1 Cation Channel , Transfection , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
7.
Cell Calcium ; 60(5): 356-362, 2016 11.
Article in English | MEDLINE | ID: mdl-27593159

ABSTRACT

Oxidative stress drives cell death in a number of diseases including ischemic stroke and neurodegenerative diseases. A better understanding of how cells recover from oxidative stress is likely to lead to better treatments for stroke and other diseases. The recent evidence obtained in several models ties the process of lysosomal exocytosis to the clearance of protein aggregates and toxic metals. The mechanisms that regulate lysosomal exocytosis, under normal or pathological conditions, are only beginning to emerge. Here we provide evidence for the biphasic effect of oxidative stress on lysosomal exocytosis. Lysosomal exocytosis was measured using the extracellular levels of the lysosomal enzyme beta-hexosaminidase (ß-hex). Low levels or oxidative stress stimulated lysosomal exocytosis, but inhibited it at high levels. Deletion of the lysosomal ion channel TRPML1 eliminated the stimulatory effect of low levels of oxidative stress. The inhibitory effects of oxidative stress appear to target the component of lysosomal exocytosis that is driven by extracellular Ca2+. We propose that while moderate oxidative stress promotes cellular repair by stimulating lysosomal exocytosis, at high levels oxidative stress has a dual pathological effect: it directly causes cell damage and impairs damage repair by inhibiting lysosomal exocytosis. Harnessing these adaptive mechanisms may point to pharmacological interventions for diseases involving oxidative proteotoxicity or metal toxicity.


Subject(s)
Exocytosis , Lysosomes/metabolism , Oxidative Stress , Calcium/metabolism , Exocytosis/drug effects , HeLa Cells , Humans , Lysosomes/drug effects , Metals, Heavy/pharmacology , Oxidative Stress/drug effects , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/metabolism , Tumor Cells, Cultured
8.
Br J Pharmacol ; 173(15): 2419-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27189253

ABSTRACT

BACKGROUND AND PURPOSE: Transient receptor potential ankyrin-1 (TRPA1) activation is known to mediate neurogenic vasodilatation. We investigated the mechanisms involved in TRPA1-mediated peripheral vasodilatation in vivo using the TRPA1 agonist cinnamaldehyde. EXPERIMENTAL APPROACH: Changes in vascular ear blood flow were measured in anaesthetized mice using laser Doppler flowmetry. KEY RESULTS: Topical application of cinnamaldehyde to the mouse ear caused a significant increase in blood flow in the skin of anaesthetized wild-type (WT) mice but not in TRPA1 knockout (KO) mice. Cinnamaldehyde-induced vasodilatation was inhibited by the pharmacological blockade of the potent microvascular vasodilator neuropeptide CGRP and neuronal NOS-derived NO pathways. Cinnamaldehyde-mediated vasodilatation was significantly reduced by treatment with reactive oxygen nitrogen species (RONS) scavenger such as catalase and the SOD mimetic TEMPOL, supporting a role of RONS in the downstream vasodilator TRPA1-mediated response. Co-treatment with a non-selective NOS inhibitor L-NAME and antioxidant apocynin further inhibited the TRPA1-mediated vasodilatation. Cinnamaldehyde treatment induced the generation of peroxynitrite that was blocked by the peroxynitrite scavenger FeTPPS and shown to be dependent on TRPA1, as reflected by an increase in protein tyrosine nitration in the skin of WT, but not in TRPA1 KO mice. CONCLUSION AND IMPLICATIONS: This study provides in vivo evidence that TRPA1-induced vasodilatation mediated by cinnamaldehyde requires neuronal NOS-derived NO, in addition to the traditional neuropeptide component. A novel role of peroxynitrite is revealed, which is generated downstream of TRPA1 activation by cinnamaldehyde. This mechanistic pathway underlying TRPA1-mediated vasodilatation may be important in understanding the role of TRPA1 in pathophysiological situations.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Neurogenesis , Nitrogen Oxides/metabolism , Reactive Nitrogen Species/metabolism , Transient Receptor Potential Channels/metabolism , Vasodilation , Acrolein/analogs & derivatives , Acrolein/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/drug effects , TRPA1 Cation Channel , Transient Receptor Potential Channels/deficiency , Vasodilation/drug effects
9.
PLoS One ; 11(3): e0151602, 2016.
Article in English | MEDLINE | ID: mdl-26978657

ABSTRACT

Keratinocytes are the first cells that come into direct contact with external tactile stimuli; however, their role in touch transduction in vivo is not clear. The ion channel Transient Receptor Potential Ankyrin 1 (TRPA1) is essential for some mechanically-gated currents in sensory neurons, amplifies mechanical responses after inflammation, and has been reported to be expressed in human and mouse skin. Other reports have not detected Trpa1 mRNA transcripts in human or mouse epidermis. Therefore, we set out to determine whether selective deletion of Trpa1 from keratinocytes would impact mechanosensation. We generated K14Cre-Trpa1fl/fl mice lacking TRPA1 in K14-expressing cells, including keratinocytes. Surprisingly, Trpa1 transcripts were very poorly detected in epidermis of these mice or in controls, and detection was minimal enough to preclude observation of Trpa1 mRNA knockdown in the K14Cre-Trpa1fl/fl mice. Unexpectedly, these K14Cre-Trpa1fl/fl mice nonetheless exhibited a pronounced deficit in mechanosensitivity at the behavioral and primary afferent levels, and decreased mechanically-evoked ATP release from skin. Overall, while these data suggest that the intended targeted deletion of Trpa1 from keratin 14-expressing cells of the epidermis induces functional deficits in mechanotransduction and ATP release, these deficits are in fact likely due to factors other than reduction of Trpa1 expression in adult mouse keratinocytes because they express very little, if any, Trpa1.


Subject(s)
Adenosine Triphosphate/metabolism , Keratinocytes/metabolism , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Transient Receptor Potential Channels/deficiency , Afferent Pathways/physiology , Animals , Animals, Congenic , Arthritis, Experimental/physiopathology , Epidermal Cells , Epidermis/metabolism , Freund's Adjuvant/toxicity , Gene Expression Profiling , Gene Knockdown Techniques , Genes, Reporter , Integrases , Mice , Mice, Inbred C57BL , Nociception/physiology , Organ Specificity , Pain Threshold/physiology , Physical Stimulation/adverse effects , RNA, Messenger/biosynthesis , Sensory Receptor Cells/physiology , Skin/cytology , Skin/embryology , TRPA1 Cation Channel , Transient Receptor Potential Channels/biosynthesis , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/physiology
10.
Sci Rep ; 6: 23261, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26983498

ABSTRACT

Dysesthesia is an unpleasant abnormal sensation, which is often accompanied by peripheral neuropathy or vascular impairment. Here, we examined the roles of transient receptor potential ankyrin 1 (TRPA1) in dysesthesia-like behaviours elicited by transient hindlimb ischemia (15-60 min) by tightly compressing the hindlimb, and reperfusion by releasing the ligature. The paw-withdrawal responses to tactile stimulation were reduced during ischemia and lasted for a while after reperfusion. Hindlimb ischemia/reperfusion elicited spontaneous licking of the ischemic hindpaw that peaked within 10 min. The licking was inhibited by reactive oxygen species (ROS) scavengers, a TRPA1 antagonist, or TRPA1 deficiency, but not by TRPV1 deficiency. In human TRPA1-expressing cells as well as cultured mouse dorsal root ganglion neurons, the H2O2-evoked TRPA1 response was significantly increased by pretreatment with hypoxia (80 mmHg) for 30 min. This hypoxia-induced TRPA1 sensitisation to H2O2 was inhibited by overexpressing a catalytically-inactive mutant of prolyl hydroxylase (PHD) 2 or in a TRPA1 proline mutant resistant to PHDs. Consistent with these results, a PHD inhibitor increased H2O2-evoked nocifensive behaviours through TRPA1 activation. Our results suggest that transient hindlimb ischemia/reperfusion-evoked spontaneous licking, i.e. painful dysesthesia, is caused by ROS-evoked activation of TRPA1 sensitised by hypoxia through inhibiting PHD-mediated hydroxylation of a proline residue in TRPA1.


Subject(s)
Cell Hypoxia , Paresthesia/pathology , Transient Receptor Potential Channels/metabolism , Animals , Behavior, Animal , Calcium/metabolism , Cells, Cultured , Disease Models, Animal , Fluorometry , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , HEK293 Cells , Hindlimb/physiology , Humans , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Mutagenesis, Site-Directed , Paresthesia/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , TRPA1 Cation Channel , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
11.
Arthritis Res Ther ; 18: 7, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26754745

ABSTRACT

BACKGROUND: The effect of cold temperature on arthritis symptoms is unclear. The aim of this study was to investigate how environmental cold affects pain and blood flow in mono-arthritic mice, and examine a role for transient receptor potential ankyrin 1 (TRPA1), a ligand-gated cation channel that can act as a cold sensor. METHODS: Mono-arthritis was induced by unilateral intra-articular injection of complete Freund's adjuvant (CFA) in CD1 mice, and in mice either lacking TRPA1 (TRPA1 KO) or respective wildtypes (WT). Two weeks later, nociception and joint blood flow were measured following exposure to 10 °C (1 h) or room temperature (RT). Primary mechanical hyperalgesia in the knee was measured by pressure application apparatus; secondary mechanical hyperalgesia by automated von Frey system; thermal hyperalgesia by Hargreaves technique, and weight bearing by the incapacitance test. Joint blood flow was recorded by full-field laser perfusion imager (FLPI) and using clearance of (99m)Technetium. Blood flow was assessed after pretreatment with antagonists of either TRPA1 (HC-030031), substance P neurokinin 1 (NK1) receptors (SR140333) or calcitonin gene-related peptide (CGRP) (CGRP8-37). TRPA1, TAC-1 and CGRP mRNA levels were examined in dorsal root ganglia, synovial membrane and patellar cartilage samples. RESULTS: Cold exposure caused bilateral primary mechanical hyperalgesia 2 weeks after CFA injection, in a TRPA1-dependent manner. In animals maintained at RT, clearance techniques and FLPI showed that CFA-treated joints exhibited lower blood flow than saline-treated joints. In cold-exposed animals, this reduction in blood flow disappears, and increased blood flow in the CFA-treated joint is observed using FLPI. Cold-induced increased blood flow in CFA-treated joints was blocked by HC-030031 and not observed in TRPA1 KOs. Cold exposure increased TRPA1 mRNA levels in patellar cartilage, whilst reducing it in synovial membranes from CFA-treated joints. CONCLUSIONS: We provide evidence that environmental cold exposure enhances pain and increases blood flow in a mono-arthritis model. These changes are dependent on TRPA1. Thus, TRPA1 may act locally within the joint to influence blood flow via sensory nerves, in addition to its established nociceptive actions.


Subject(s)
Arthritis, Experimental/metabolism , Blood Flow Velocity/physiology , Cold Temperature/adverse effects , Freund's Adjuvant/toxicity , Joints/metabolism , Transient Receptor Potential Channels/biosynthesis , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Blood Flow Velocity/drug effects , Freund's Adjuvant/administration & dosage , Hindlimb/drug effects , Hindlimb/metabolism , Hindlimb/pathology , Injections, Intra-Articular , Joints/drug effects , Joints/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/drug effects , Pain Measurement/methods , Pain Threshold/drug effects , Pain Threshold/physiology , TRPA1 Cation Channel , Transient Receptor Potential Channels/deficiency
12.
Nature ; 529(7587): 523-7, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26760212

ABSTRACT

The myelin sheaths wrapped around axons by oligodendrocytes are crucial for brain function. In ischaemia myelin is damaged in a Ca(2+)-dependent manner, abolishing action potential propagation. This has been attributed to glutamate release activating Ca(2+)-permeable N-methyl-D-aspartate (NMDA) receptors. Surprisingly, we now show that NMDA does not raise the intracellular Ca(2+) concentration ([Ca(2+)]i) in mature oligodendrocytes and that, although ischaemia evokes a glutamate-triggered membrane current, this is generated by a rise of extracellular [K(+)] and decrease of membrane K(+) conductance. Nevertheless, ischaemia raises oligodendrocyte [Ca(2+)]i, [Mg(2+)]i and [H(+)]i, and buffering intracellular pH reduces the [Ca(2+)]i and [Mg(2+)]i increases, showing that these are evoked by the rise of [H(+)]i. The H(+)-gated [Ca(2+)]i elevation is mediated by channels with characteristics of TRPA1, being inhibited by ruthenium red, isopentenyl pyrophosphate, HC-030031, A967079 or TRPA1 knockout. TRPA1 block reduces myelin damage in ischaemia. These data suggest that TRPA1-containing ion channels could be a therapeutic target in white matter ischaemia.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Calcium/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Protons , Transient Receptor Potential Channels/metabolism , Animals , Calcium Signaling/drug effects , Electric Conductivity , Female , Hydrogen-Ion Concentration , Magnesium/metabolism , Male , Mice , Mice, Transgenic , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/drug effects , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Stroke/metabolism , Stroke/pathology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics , White Matter/metabolism , White Matter/pathology
13.
Arthritis Res Ther ; 18: 6, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26746673

ABSTRACT

BACKGROUND: The transient receptor potential ankyrin 1 (TRPA1) is a calcium-permeable cation channel that is expressed on capsaicin-sensitive sensory neurons, endothelial and inflammatory cells. It is activated by a variety of inflammatory mediators, such as methylglyoxal, formaldehyde and hydrogen sulphide. Since only few data are available about the role of TRPA1 in arthritis and related pain, we investigated its involvement in inflammation models of different mechanisms. METHODS: Chronic arthritis was induced by complete Freund's adjuvant (CFA), knee osteoarthritis by monosodium iodoacetate (MIA) in TRPA1 knockout (KO) mice and C57Bl/6 wildtype mice. For comparison, carrageenan- and CFA-evoked acute paw and knee inflammatory changes were investigated. Thermonociception was determined on a hot plate, cold tolerance in icy water, mechanonociception by aesthesiometry, paw volume by plethysmometry, knee diameter by micrometry, weight distribution with incapacitance tester, neutrophil myeloperoxidase activity and vascular leakage by in vivo optical imaging, and histopathological alterations by semiquantitative scoring. RESULTS: CFA-induced chronic mechanical hypersensitivity, tibiotarsal joint swelling and histopathological alterations, as well as myeloperoxidase activity in the early phase (day 2), and vascular leakage in the later stage (day 7), were significantly reduced in TRPA1 KO mice. Heat and cold sensitivities did not change in this model. Although in TRPA1 KO animals MIA-evoked knee swelling and histopathological destruction were not altered, hypersensitivity and impaired weight bearing on the osteoarthritic limb were significantly decreased. In contrast, carrageenan- and CFA-induced acute inflammation and pain behaviours were not modified by TRPA1 deletion. CONCLUSIONS: TRPA1 has an important role in chronic arthritis/osteoarthritis and related pain behaviours in the mouse. Therefore, it might be a promising target for novel analgesic/anti-inflammatory drugs.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Transient Receptor Potential Channels/deficiency , Animals , Chronic Disease , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , TRPA1 Cation Channel
14.
Am J Pathol ; 186(1): 199-209, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26608452

ABSTRACT

Mucolipidosis IV is a debilitating developmental lysosomal storage disorder characterized by severe neuromotor retardation and progressive loss of vision, leading to blindness by the second decade of life. Mucolipidosis IV is caused by loss-of-function mutations in the MCOLN1 gene, which encodes the transient receptor potential channel protein mucolipin-1. Ophthalmic pathology in patients includes corneal haze and progressive retinal and optic nerve atrophy. Herein, we report ocular pathology in Mcoln1(-/-) mouse, a good phenotypic model of the disease. Early, but non-progressive, thinning of the photoreceptor layer, reduced levels of rhodopsin, disrupted rod outer segments, and widespread accumulation of the typical storage inclusion bodies were the major histological findings in the Mcoln1(-/-) retina. Electroretinograms showed significantly decreased functional response (scotopic a- and b-wave amplitudes) in the Mcoln1(-/-) mice. At the ultrastructural level, we observed formation of axonal spheroids and decreased density of axons in the optic nerve of the aged (6-month-old) Mcoln1(-/-) mice, which indicates progressive axonal degeneration. Our data suggest that mucolipin-1 plays a role in postnatal development of photoreceptors and provides a set of outcome measures that can be used for ocular therapy development for mucolipidosis IV.


Subject(s)
Mucolipidoses/pathology , Optic Nerve/pathology , Retinal Dystrophies/pathology , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucolipidoses/complications , Tomography, Optical Coherence , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
16.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L812-20, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26472811

ABSTRACT

Cigarette smoke (CS) exposes chemosensory nerves in the airways to a multitude of chemicals, some acting through the irritant receptors TRPV1 and TRPA1 but potentially also through nicotinic acetylcholine receptors (nAChR). Our aim was to characterize the differences in sensory neuronal effects of CS, gas phase, and particulate matter as well as of typical constituents, such as nicotine and reactive carbonyls. Isolated mouse trachea and larynx were employed to measure release of calcitonin gene-related peptide (CGRP) as an index of sensory neuron activation evoked by CS, by filtered CS gas phase essentially free of nicotine, and by dilute total particulate matter (TPM) containing defined nicotine concentrations. With CS stimulation of the superfused trachea, TRPV1 null mutants showed about the same large responses as wild-type mice, whereas both TRPA1(-/-) and double knockouts exhibited 80% reduction; the retained 20% response was abolished by mecamylamine (10 µM), indicating a distinct contribution of nAChRs. These phenotypes were accentuated by using TPM to stimulate the immersed trachea; 50% of response was retained in TRPA1(-/-) and abolished by mecamylamine. In contrast, the gas phase acted like a sheer TRPA1 agonist, consistent with its composition, among other compounds, of volatile reactive carbonyls like formaldehyde and acrolein. In the trachea, the gas phase and CS were equally effective in releasing CGRP, whereas the larynx showed much larger CS than gas phase responses. Thus nicotinic receptors contribute to the sensory effects of cigarette smoke on the trachea, which are dominated by TRPA1. How this translates to human perception affords future research.


Subject(s)
Larynx/metabolism , Receptors, Nicotinic/metabolism , Smoking/adverse effects , Smoking/metabolism , TRPV Cation Channels/metabolism , Trachea/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sensory Receptor Cells/metabolism , Smoke/adverse effects , Smoke/analysis , TRPA1 Cation Channel , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
17.
Dis Model Mech ; 8(12): 1591-601, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26398942

ABSTRACT

Mucolipidosis type IV (MLIV) is a lysosomal storage disease caused by mutations in the MCOLN1 gene, which encodes the lysosomal transient receptor potential ion channel mucolipin-1 (TRPML1). MLIV causes impaired motor and cognitive development, progressive loss of vision and gastric achlorhydria. How loss of TRPML1 leads to severe psychomotor retardation is currently unknown, and there is no therapy for MLIV. White matter abnormalities and a hypoplastic corpus callosum are the major hallmarks of MLIV brain pathology. Here, we report that loss of TRPML1 in mice results in developmental aberrations of brain myelination as a result of deficient maturation and loss of oligodendrocytes. Defective myelination is evident in Mcoln1(-/-) mice at postnatal day 10, an active stage of postnatal myelination in the mouse brain. Expression of mature oligodendrocyte markers is reduced in Mcoln1(-/-) mice at postnatal day 10 and remains lower throughout the course of the disease. We observed reduced Perls' staining in Mcoln1(-/-) brain, indicating lower levels of ferric iron. Total iron content in unperfused brain is not significantly different between Mcoln1(-/-) and wild-type littermate mice, suggesting that the observed maturation delay or loss of oligodendrocytes might be caused by impaired iron handling, rather than by global iron deficiency. Overall, these data emphasize a developmental rather than a degenerative disease course in MLIV, and suggest that there should be a stronger focus on oligodendrocyte maturation and survival to better understand MLIV pathogenesis and aid treatment development.


Subject(s)
Brain/metabolism , Iron/metabolism , Mucolipidoses/metabolism , Mucolipidoses/pathology , Myelin Sheath/pathology , Animals , Axons/pathology , Brain/pathology , Cell Count , Corpus Callosum/pathology , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/metabolism , Oligodendroglia/pathology , Oxidative Stress , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
18.
Mol Pain ; 11: 24, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25934637

ABSTRACT

BACKGROUND: Hydrogen sulfide (H2S) is oxidized to polysulfide. Recent reports show that this sulfur compound modulates various biological functions. We have reported that H2S is involved in inflammatory pain in mice. On the other hand, little is known about the functional role of polysulfide in sensory neurons. Here we show that polysulfide selectively stimulates nociceptive TRPA1 and evokes acute pain, using TRPA1-gene deficient mice (TRPA1(-/-)), a heterologous expression system and a TRPA1-expressing cell line. RESULTS: In wild-type mouse sensory neurons, polysulfide elevated the intracellular Ca concentration ([Ca(2+)]i) in a dose-dependent manner. The half maximal effective concentration (EC50) of polysulfide was less than one-tenth that of H2S. The [Ca(2+)]i responses to polysulfide were observed in neurons responsive to TRPA1 agonist and were inhibited by blockers of TRPA1 but not of TRPV1. Polysulfide failed to evoke [Ca(2+)]i increases in neurons from TRPA1(-/-) mice. In RIN-14B cells, constitutively expressing rat TRPA1, polysulfide evoked [Ca(2+)]i increases with the same EC50 value as in sensory neurons. Heterologously expressed mouse TRPA1 was activated by polysulfide and that was suppressed by dithiothreitol. Analyses of the TRPA1 mutant channel revealed that cysteine residues located in the internal domain were related to the sensitivity to polysulfide. Intraplantar injection of polysulfide into the mouse hind paw induced acute pain and edema which were significantly less than in TRPA1(-/-) mice. CONCLUSIONS: The present data suggest that polysulfide functions as pronociceptive substance through the activation of TRPA1 in sensory neurons. Since the potency of polysulfide is higher than parental H2S and this sulfur compound is generated under pathophysiological conditions, it is suggested that polysulfide acts as endogenous ligand for TRPA1. Therefore, TRPA1 may be a promising therapeutic target for endogenous sulfur compound-related algesic action.


Subject(s)
Acute Pain/drug therapy , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sulfides/pharmacology , Transient Receptor Potential Channels/metabolism , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Mice, Inbred C57BL , Mice, Knockout , Neurons, Afferent/drug effects , TRPA1 Cation Channel , Transient Receptor Potential Channels/deficiency
19.
Metab Brain Dis ; 30(3): 681-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25156245

ABSTRACT

Mucolipidosis type IV (MLIV) is a very rare disorder of late endosome/lysosome transport, characterized by neurodevelopmental abnormalities and progressive visual impairment owing to corneal clouding and retinal dystrophy. Greater than 70 % of MLIV patients are of Ashkenazi Jewish ancestry. Here we report a novel MCOLN1double mutant allele [c.395_397delCTG;c.468_474dupTTGGACC] which introduces a premature stop codon [p.Ala132del; p.Asn159LeufsX27] leading to almost complete abrogation of the region coding mucolipin-1, a member of the transient receptor potential (TRP) cation channel family. The genomic lesion was identified in homozygous state, in a non-Jewish Italian MLIV patient, who also presented abnormal serum gastrin levels. Conventional and advanced MRI sequences, including diffusion tensor imaging and tractography, were used for the assessment of white matter involvement in the patient.


Subject(s)
Alleles , Mucolipidoses/genetics , Mutation/genetics , Transient Receptor Potential Channels/genetics , White People/genetics , Child, Preschool , Homozygote , Humans , Italy , Male , Mucolipidoses/diagnosis , Transient Receptor Potential Channels/deficiency
20.
Nat Commun ; 5: 5732, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25501034

ABSTRACT

The cold-induced vascular response, consisting of vasoconstriction followed by vasodilatation, is critical for protecting the cutaneous tissues against cold injury. Whilst this physiological reflex response is historic knowledge, the mechanisms involved are unclear. Here by using a murine model of local environmental cold exposure, we show that TRPA1 acts as a primary vascular cold sensor, as determined through TRPA1 pharmacological antagonism or gene deletion. The initial cold-induced vasoconstriction is mediated via TRPA1-dependent superoxide production that stimulates α2C-adrenoceptors and Rho-kinase-mediated MLC phosphorylation, downstream of TRPA1 activation. The subsequent restorative blood flow component is also dependent on TRPA1 activation being mediated by sensory nerve-derived dilator neuropeptides CGRP and substance P, and also nNOS-derived NO. The results allow a new understanding of the importance of TRPA1 in cold exposure and provide impetus for further research into developing therapeutic agents aimed at the local protection of the skin in disease and adverse climates.


Subject(s)
Hypothermia/metabolism , Receptors, Adrenergic, alpha/genetics , Skin/blood supply , Transient Receptor Potential Channels/genetics , Vasoconstriction/genetics , Acetanilides/pharmacology , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Cold Temperature/adverse effects , Gene Expression Regulation , Hindlimb , Hypothermia/etiology , Hypothermia/genetics , Hypothermia/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Purines/pharmacology , Receptors, Adrenergic, alpha/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , Substance P/genetics , Substance P/metabolism , Superoxides/metabolism , TRPA1 Cation Channel , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/deficiency , Vasodilation/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...