Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(25): e2322475121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857404

ABSTRACT

Low temperatures and cooling agents like menthol induce cold sensation by activating the peripheral cold receptors TRPM8 and TRPA1, cation channels belonging to the TRP channel family, while the reduction of potassium currents provides an additional and/or synergistic mechanism of cold sensation. Despite extensive studies over the past decades to identify the molecular receptors that mediate thermosensation, cold sensation is still not fully understood and many cold-sensitive peripheral neurons do not express the well-established cold sensor TRPM8. We found that the voltage-gated potassium channel KCNQ1 (Kv7.1), which is defective in cardiac LQT1 syndrome, is, in addition to its known function in the heart, a highly relevant and sex-specific sensor of moderately cold temperatures. We found that KCNQ1 is expressed in skin and dorsal root ganglion neurons, is sensitive to menthol and cooling agents, and is highly sensitive to moderately cold temperatures, in a temperature range at which TRPM8 is not thermosensitive. C-fiber recordings from KCNQ1-/- mice displayed altered action potential firing properties. Strikingly, only male KCNQ1-/- mice showed substantial deficits in cold avoidance at moderately cold temperatures, with a strength of the phenotype similar to that observed in TRPM8-/- animals. While sex-dependent differences in thermal sensitivity have been well documented in humans and mice, KCNQ1 is the first gene reported to play a role in sex-specific temperature sensation. Moreover, we propose that KCNQ1, together with TRPM8, is a key instrumentalist that orchestrates the range and intensity of cold sensation.


Subject(s)
Cold Temperature , KCNQ1 Potassium Channel , Animals , Male , Female , Mice , KCNQ1 Potassium Channel/metabolism , KCNQ1 Potassium Channel/genetics , Mice, Knockout , Ganglia, Spinal/metabolism , Thermosensing/physiology , TRPM Cation Channels/metabolism , TRPM Cation Channels/genetics , Mice, Inbred C57BL , Action Potentials/physiology , Sex Characteristics , Menthol/pharmacology
3.
BMC Cancer ; 24(1): 346, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38500100

ABSTRACT

BACKGROUND: The androgen/androgen receptor (AR)-signaling axis plays a central role in prostate cancer (PCa). Upon androgen-binding the AR dimerizes with another AR, and translocates into the nucleus where the AR-dimer activates/inactivates androgen-dependent genes. Consequently, treatments for PCa are commonly based on androgen deprivation therapy (ADT). The clinical benefits of ADT are only transitory and most tumors develop mechanisms allowing the AR to bypass its need for physiological levels of circulating androgens. Clinical failure of ADT is often characterized by the synthesis of a constitutively active AR splice variant, termed AR-V7. AR-V7 mRNA expression is considered as a resistance mechanism following ADT. AR-V7 no longer needs androgenic stimuli for nuclear entry and/or dimerization. METHODS: Our goal was to mechanistically decipher the interaction between full-length AR (AR-FL) and AR-V7 in AR-null HEK-293 cells using the NanoLuc Binary Technology under androgen stimulation and deprivation conditions. RESULTS: Our data point toward a hypothesis that AR-FL/AR-FL homodimers form in the cytoplasm, whereas AR-V7/AR-V7 homodimers localize in the nucleus. However, after androgen stimulation, all the AR-FL/AR-FL, AR-FL/AR-V7 and AR-V7/AR-V7 dimers were localized in the nucleus. CONCLUSIONS: We showed that AR-FL and AR-V7 form heterodimers that localize to the nucleus, whereas AR-V7/AR-V7 dimers were found to localize in the absence of androgens in the nucleus.


Subject(s)
Luciferases , Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Androgens , Prostatic Neoplasms/pathology , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , HEK293 Cells , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms/genetics
4.
Ocul Surf ; 30: 204-212, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37774917

ABSTRACT

PURPOSE: To study the tissue architecture, isthmus (connection between two lobes) of the lacrimal gland using preclinical 7T MRI in combination with histology and electron microscopy. METHODS: Ten lacrimal glands from Caucasian body donors (mean age 78.7 years) were studied using 7T-MRI (N = 5; scanned at 75-µm intervals), histology, and electron microscopy (N = 5) and 3D cinematic rendering (CR) techniques. RESULTS: 3D CR images showed uniform-sized lobules (widest lobule diameter, 1.68 ± 0.19 mm in orbital lobe, 1.68 ± 0.17 mm in palpebral lobe) in both lobes, separated by septae (size, 0.29 ± 0.09 mm). The internal framework of the gland resembled a honeycoomb pattern. In CR and histology, the isthmus contained glandular acini, large blood vessels, nerves, and no more than two ducts having a tortuous course towards the conjunctival surface. On assigning a color display to the rendered lacrimal gland, all glands showed a blood vessel originating from the main lacrimal artery just 5 mm beyond the hilum and making it course to the palpebral lobe via isthmus. The distance between the conjunctiva and the central substance of the orbital and palpebral lobe was 9.4 ± 0.2 mm and 2.8 ± 0.7 mm, respectively. Electron microscopy of the palpebral lobe revealed compact subepithelial layer in the overlying conjunctiva, followed by loosely scattered collagen bundles that contained the gland lobules. CONCLUSION: 3D-CR can be used to study the lacrimal gland microstructure, help fabricate a 3D scaffold for lacrimal gland bioprinting, and serve as guide for transconjunctival lacrimal gland targeted therapies i.e., 2.9 & 9 mm long needle to reach the orbital and palpebral lobe center, respectively in normal-size glands.


Subject(s)
Lacrimal Apparatus Diseases , Lacrimal Apparatus , Humans , Aged , Lacrimal Apparatus/diagnostic imaging , Microscopy, Electron , Magnetic Resonance Imaging , Bioengineering
5.
Front Immunol ; 13: 933191, 2022.
Article in English | MEDLINE | ID: mdl-36505422

ABSTRACT

Vitamin D (VD) deficiency is a highly prevalent worldwide phenomenon and is extensively discussed as a risk factor for the development of systemic lupus erythematosus (SLE) and other immune-mediated diseases. In addition, it is now appreciated that VD possesses multiple immunomodulatory effects. This study aims to explore the impact of dietary VD intake on lupus manifestation and pathology in lupus-prone NZB/W F1 mice and identify the underlying immunological mechanisms modulated by VD. Here, we show that low VD intake accelerates lupus progression, reflected in reduced overall survival and an earlier onset of proteinuria, as well higher concentrations of anti-double-stranded DNA autoantibodies. This unfavorable effect gained statistical significance with additional low maternal VD intake during the prenatal period. Among examined immunological effects, we found that low VD intake consistently hampered the adoption of a regulatory phenotype in lymphocytes, significantly reducing both IL-10-expressing and regulatory CD4+ T cells. This goes along with a mildly decreased frequency of IL-10-expressing B cells. We did not observe consistent effects on the phenotype and function of innate immune cells, including cytokine production, costimulatory molecule expression, and phagocytic capacity. Hence, our study reveals that low VD intake promotes lupus pathology, likely via the deviation of adaptive immunity, and suggests that the correction of VD deficiency might not only exert beneficial functions by preventing osteoporosis but also serve as an important module in prophylaxis and as an add-on in the treatment of lupus and possibly other immune-mediated diseases. Further research is required to determine the most appropriate dosage, as too-high VD serum levels may also induce adverse effects, possibly also on lupus pathology.


Subject(s)
Vitamin D Deficiency , Vitamin D , Animals , Mice , Female , Pregnancy , Interleukin-10 , Mice, Inbred NZB , Vitamins , Diet
6.
Gastroenterology ; 161(4): 1270-1287.e19, 2021 10.
Article in English | MEDLINE | ID: mdl-34224738

ABSTRACT

BACKGROUND & AIMS: The molecular checkpoints driving T cell activation and cytokine responses in ulcerative colitis (UC) are incompletely understood. Here, we studied the Tec kinase ITK in UC. METHODS: We analyzed patients with inflammatory bowel disease (n = 223) and evaluated ITK activity as well as the functional effects of cyclosporine-A (CsA). In addition, 3 independent murine colitis models were used to investigate the functional role of ITK. Finally, the activity of ITK was blocked via pharmacological inhibitors and genetically engineered mice. Readout parameters were mini-endoscopy, histopathology, mucosal T cell apoptosis, and cytokine production. RESULTS: We found an expansion of pITK-expressing mucosal CD4+ T cells in UC rather than Crohn's disease that correlated with disease severity. CsA suppressed activation of ITK in cultured CD4+ T cells and calcineurin-containing microclusters adjacent to the T cell receptor signaling complex. Functionally, the capacity of CsA to suppress activity of experimental colitis was critically dependent on ITK. Genetic inactivation of Itk via gene targeting or induction of allele-sensitive Itk mutants prevented experimental colitis in 3 colitis models, and treatment with pharmacological ITK blockers suppressed established colitis. In addition, ITK controlled apoptosis and activation of mucosal Th2 and Th17 lymphocytes via NFATc2 signaling pathways. CONCLUSIONS: ITK activation was detected in UC and could be down-regulated in cultured T cells by CsA administration. Selective targeting of ITK emerges as an attractive approach for treatment of chronic intestinal inflammation and potentially UC by driving resolution of mucosal inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/prevention & control , Colon/drug effects , Intestinal Mucosa/drug effects , Intraepithelial Lymphocytes/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cells, Cultured , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/enzymology , Colon/immunology , Colon/pathology , Cyclosporine/pharmacology , Cytokines/metabolism , Disease Models, Animal , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intraepithelial Lymphocytes/enzymology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/pathology , Mice, Knockout , Molecular Targeted Therapy , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction
7.
Bioinformatics ; 37(24): 4901-4902, 2021 12 11.
Article in English | MEDLINE | ID: mdl-34152405

ABSTRACT

SUMMARY: Creating 3D animations from microscopy data is computationally expensive and requires high-end hardware. We therefore developed 3Dscript.server, a 3D animation software that runs as a service on dedicated, shared workstations. Using 3Dscript as the underlying rendering engine, it offers unique features not found in existing software: rendering is performed completely server-side. The target animation is specified on the client without the rendering engine, eliminating any hardware requirements client-side. Still, defining an animation is intuitive due to 3Dscript's natural language-based animation description. We implemented a new OMERO web app to utilize 3Dscript.server directly from the OMERO web interface; a Fiji client to use 3Dscript.server from Fiji for integration into image processing pipelines; and batch scripts to run 3Dscript.server on compute clusters for large-scale visualization projects. AVAILABILITY AND IMPLEMENTATION: Source code and documentation is available at https://github.com/bene51/omero_3Dscript, https://github.com/bene51/3Dscript.server and https://github.com/bene51/3Dscript.cluster. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Computers , Microscopy , Humans , Software , Language , Image Processing, Computer-Assisted
8.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33771873

ABSTRACT

Teeth are composed of many tissues, covered by an inflexible and obdurate enamel. Unlike most other tissues, teeth become extremely cold sensitive when inflamed. The mechanisms of this cold sensation are not understood. Here, we clarify the molecular and cellular components of the dental cold sensing system and show that sensory transduction of cold stimuli in teeth requires odontoblasts. TRPC5 is a cold sensor in healthy teeth and, with TRPA1, is sufficient for cold sensing. The odontoblast appears as the direct site of TRPC5 cold transduction and provides a mechanism for prolonged cold sensing via TRPC5's relative sensitivity to intracellular calcium and lack of desensitization. Our data provide concrete functional evidence that equipping odontoblasts with the cold-sensor TRPC5 expands traditional odontoblast functions and renders it a previously unknown integral cellular component of the dental cold sensing system.

9.
Phytomedicine ; 34: 44-49, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28899508

ABSTRACT

BACKGROUND: Transient Receptor Potential Vanilloid 1 (TRPV1) confers noxious heat and inflammatory pain signals in the peripheral nervous system. Clinical trial of resiniferatoxin from Euphorbia species is successfully aimed at TRPV1 in cancer pain management and heading toward new selective painkiller status that further validates this target for drug discovery efforts. Evodia species, used in traditional medicine for hundreds of years, are a recognised source of different TRPV1 agonists, but no antagonist has yet been reported. HYPOTHESIS/PURPOSE: In a search for painkiller leads, we noted for the first time a TRPV1 antagonist activity in the fresh fruits of Tetradium daniellii (Benn.) T.G. Hartley (syn. Evodia hupehensis Dode). METHODS: Through a combination of extraction and purification methods with functional TRPV1-specific Ca2+ uptake assays (bioactivity-guided fractionation/isolation/purification); we isolated a new painkiller candidate that is a distant structural homologue of capsiate exovanilloids and endovanilloids such as anandamide, but a putative competitive inhibitor of the TRPV1. Four additional inactive compounds (N-isobutyl-4,5-epoxy-2E-decadienamide, geranylpsoralen, 8-(7',8'-epoxygeranyloxy)psoralen, and xanthotoxol) were also co-purified with pellitorine. Their structures were established by extensive 1D- and 2D-NMR spectroscopic analysis. RESULTS: 1H- and 13C NMR determination of the chemical structure revealed it to be pellitorine, (2E,4E)-N-(2-methylpropyl)deca-2,4-dienamide, which can compete structurally with algesics released in inflammation. In contrast to previous isolates from Evodia species, pellitorine blocked capsaicin-evoked Ca2+ uptake with an IC50 of 154 µg/ml (0.69 mM/l). N-Isobutyl-4,5-epoxy-2E-decadienamide and geranylpsoralen, 8-(7',8'-epoxygeranyloxy)psoralen, and xanthotoxol did not affect the TRPV1. CONCLUSION: This is the first evidence that pellitorine, an aliphatic alkylamide analogue of capsaicin, can serve as an antagonist of the TRPV1 and may inhibit exovanilloid-induced pain.


Subject(s)
Analgesics/pharmacology , Fatty Acids, Unsaturated/pharmacology , Plant Extracts/pharmacology , Polyunsaturated Alkamides/pharmacology , Rutaceae/chemistry , TRPV Cation Channels/antagonists & inhibitors , Animals , Cell Line , Evodia/chemistry , Fatty Acids, Unsaturated/chemistry , Fruit/chemistry , Humans , Mice, Inbred BALB C , Polyunsaturated Alkamides/chemistry
10.
Front Mol Neurosci ; 10: 209, 2017.
Article in English | MEDLINE | ID: mdl-28713241

ABSTRACT

Previous research identified TRPM8 and TRPA1 cold transducers with separate functions, one being functional in the non-noxious range and the second one being a nociceptive transducer. TRPM8-deficient mice present overt deficits in the detection of environmental cool, but not a lack of cold avoidance and TRPA1-deficient mice show clear deficits in some cold nocifensive assays. The extent of TRPA1's contribution to cold sensing in vivo is still unclear, because mice lacking both TRPM8 and TRPA1 (DKO) were described with unchanged cold avoidance from TRPM8-/- based on a two-temperature-choice assay and by c-fos measurement. The present study was designed to differentiate how much TRPM8 alone and combined TRPA1 and TRPM8 contribute to cold sensing. We analyzed behavior in the thermal ring track assay adjusted between 30 and 5°C and found a large reduction in cold avoidance of the double knockout mice as compared to the TRPM8-deficient mice. We also revisited skin-nerve recordings from saphenous-nerve skin preparations with regard to nociceptors and thermoreceptors. We compared the frequency and characteristics of the cold responses of TRPM8-expressing and TRPM8-negative C-fiber nociceptors in C57BL/6J mice with nociceptors of TRPM8-deficient and DKO mice and found that TRPM8 enables nociceptors to encode cold temperatures with higher firing rates and larger responses with sustained, static component. In TRPM8-/-, C-fiber cold nociceptors were markedly reduced and appeared further reduced in DKO. Nevertheless, the remaining cold responses in both knockout strains were similar in their characteristics and they were indifferent from the TRPM8-negative cold responses found in C57BL/6J mice. TRPM8 had a comparably essential role for encoding cold in thermoreceptors and lack of TRPM8 reduced response magnitude, peak and mean firing rates and the incidence of thermoreceptors. The encoding deficits were similar in the DKO strain. Our data illustrate that lack of TRPA1 in TRPM8-deficient mice results in a disproportionately large reduction in cold avoidance behavior and also affects the incidence of cold encoding fiber types. Presumably TRPA1 compensates for lack of TRPM8 to a certain extent and both channels cooperate to cover the entire cold temperature range, making cold-temperature encoding by TRPA1-although less powerful-synergistic to TRPM8.

11.
FEBS Lett ; 590(16): 2768-75, 2016 08.
Article in English | MEDLINE | ID: mdl-27339229

ABSTRACT

There is enormous interest toward vanilloid agonists of the pain receptor TRPV1 in analgesic therapy, but the mechanisms of their sensory neuron-blocking effects at high or repeated doses are still a matter of debate. Our results have demonstrated that capsaicin and resiniferatoxin form nanomolar complexes with calmodulin, and competitively inhibit TRPV1-calmodulin interaction. These interactions involve the protein recognition interface of calmodulin, which is responsible for all of the cell-regulatory calmodulin-protein interactions. These results draw attention to a previously unknown vanilloid target, which may contribute to the explanation of the paradoxical pain-modulating behavior of these important pharmacons.


Subject(s)
Calmodulin/metabolism , Pain/metabolism , Protein Interaction Maps/drug effects , TRPV Cation Channels/metabolism , Binding Sites , Calmodulin/chemistry , Calmodulin/genetics , Capsaicin/metabolism , Capsaicin/pharmacology , Diterpenes/metabolism , Diterpenes/pharmacology , Humans , Pain/drug therapy , Protein Binding , Protein Conformation , Protein Interaction Maps/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics
12.
Temperature (Austin) ; 3(1): 77-91, 2016.
Article in English | MEDLINE | ID: mdl-27227099

ABSTRACT

Currently available behavioral assays to quantify normal cold sensitivity, cold hypersensitivity and cold hyperalgesia in mice have betimes created conflicting results in the literature. Some only capture a limited spectrum of thermal experiences, others are prone to experimenter bias or are not sensitive enough to detect the contribution of ion channels to cold sensing because in mice smaller alterations in cold nociception do not manifest as frank behavioral changes. To overcome current limitations we have designed a novel device that is automated, provides a high degree of freedom, i.e. thermal choice, and eliminates experimenter bias. The device represents a thermal gradient assay designed as a circular running track. It allows discerning exploratory behavior from thermal selection behavior and provides increased accuracy by providing measured values in duplicate and by removing edge artifacts. Our custom-designed automated offline analysis by a blob detection algorithm is devoid of movement artifacts, removes light reflection artifacts and provides an internal quality control parameter which we validated. The assay delivers discrete information on a large range of parameters extracted from the occupancy of thermally defined zones such as preference temperature and skew of the distribution. We demonstrate that the assay allows increasingly accurate phenotyping of thermal sensitivity in transgenic mice by disclosing yet unrecognized details on the phenotypes of TRPM8-, TRPA1- and TRPM8/A1-deficient mice.

13.
Toxins (Basel) ; 8(3)2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26999206

ABSTRACT

Loss-of-function mutations of Na(V)1.7 lead to congenital insensitivity to pain, a rare condition resulting in individuals who are otherwise normal except for the inability to sense pain, making pharmacological inhibition of Na(V)1.7 a promising therapeutic strategy for the treatment of pain. We characterized a novel mouse model of Na(V)1.7-mediated pain based on intraplantar injection of the scorpion toxin OD1, which is suitable for rapid in vivo profiling of Na(V)1.7 inhibitors. Intraplantar injection of OD1 caused spontaneous pain behaviors, which were reversed by co-injection with Na(V)1.7 inhibitors and significantly reduced in Na(V)1.7(-/-) mice. To validate the use of the model for profiling Na(V)1.7 inhibitors, we determined the Na(V) selectivity and tested the efficacy of the reported Na(V)1.7 inhibitors GpTx-1, PF-04856264 and CNV1014802 (raxatrigine). GpTx-1 selectively inhibited Na(V)1.7 and was effective when co-administered with OD1, but lacked efficacy when delivered systemically. PF-04856264 state-dependently and selectively inhibited Na(V)1.7 and significantly reduced OD1-induced spontaneous pain when delivered locally and systemically. CNV1014802 state-dependently, but non-selectively, inhibited Na(V) channels and was only effective in the OD1 model when delivered systemically. Our novel model of Na(V)1.7-mediated pain based on intraplantar injection of OD1 is thus suitable for the rapid in vivo characterization of the analgesic efficacy of Na(V)1.7 inhibitors.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/physiology , Pain/drug therapy , Peptides/therapeutic use , Phenyl Ethers/therapeutic use , Proline/analogs & derivatives , Scorpion Venoms/therapeutic use , Sodium Channel Blockers/therapeutic use , Spider Venoms/therapeutic use , Analgesics , Animals , Behavior, Animal/drug effects , CHO Cells , Cricetulus , Disease Models, Animal , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , NAV1.7 Voltage-Gated Sodium Channel/genetics , Nerve Fibers/drug effects , Nerve Fibers/physiology , Pain/chemically induced , Proline/therapeutic use , Saphenous Vein/innervation , Sulfonamides/therapeutic use
14.
Sci Rep ; 4: 6776, 2014 Oct 27.
Article in English | MEDLINE | ID: mdl-25345415

ABSTRACT

The B6.Cg-Tg(Thy1-YFP)16Jrs/J transgenic mouse strain, widely used to study neuronal development and regeneration, expresses the yellow fluorescent protein (YFP) in the peripheral nerves and the central nervous system under the control of regulatory sequences of the Thy1 gene. The Thy1 (CD90) cell surface glycoprotein is present on many cell types besides neurons, and is known to be involved in cell adhesion, migration and signal transduction. We hypothesized that Thy1-activating conditions could probably activate the truncated Thy1 regulatory sequences used in the Thy1-YFP construct, resulting in YFP transgene expression outside the nervous system. We demonstrated that the stroma of subcutaneous tumours induced by the injection of 4T1 or MC26 carcinoma cells into BALB/c(Thy1-YFP) mice, carrying the same construct, indeed expressed the YFP transgene. In the tumour mass, the yellow-green fluorescent stromal cells were clearly distinguishable from 4T1 carcinoma cells stably transfected with red fluorescent protein. Local inflammation induced by subcutaneous injection of complete Freund's adjuvant, as well as the experimental wound-healing milieu, also triggered YFP fluorescence in both the BALB/c(Thy1-YFP) and B6.Cg-Tg(Thy1-YFP)16Jrs/J mice, pointing to eventual overlapping pathways of wound-healing, inflammation and tumour growth.


Subject(s)
Diagnostic Imaging/methods , Inflammation/diagnosis , Neoplasms, Experimental/diagnosis , Wound Healing , Animals , Inflammation/genetics , Inflammation/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Molecular Imaging/methods , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Transport , Thy-1 Antigens/genetics , Thy-1 Antigens/metabolism , Transcriptional Activation , Wound Healing/genetics
15.
Mol Pain ; 9: 30, 2013 Jun 22.
Article in English | MEDLINE | ID: mdl-23800232

ABSTRACT

This review aims to create an overview of the currently available results of site-directed mutagenesis studies on transient receptor potential vanilloid type 1 (TRPV1) receptor. Systematization of the vast number of data on the functionally important amino acid mutations of TRPV1 may provide a clearer picture of this field, and may promote a better understanding of the relationship between the structure and function of TRPV1. The review summarizes information on 112 unique mutated sites along the TRPV1, exchanged to multiple different residues in many cases. These mutations influence the effect or binding of different agonists, antagonists, and channel blockers, alter the responsiveness to heat, acid, and voltage dependence, affect the channel pore characteristics, and influence the regulation of the receptor function by phosphorylation, glycosylation, calmodulin, PIP2, ATP, and lipid binding. The main goal of this paper is to publish the above mentioned data in a form that facilitates in silico molecular modelling of the receptor by promoting easier establishment of boundary conditions. The better understanding of the structure-function relationship of TRPV1 may promote discovery of new, promising, more effective and safe drugs for treatment of neurogenic inflammation and pain-related diseases and may offer new opportunities for therapeutic interventions.


Subject(s)
Mutation , TRPV Cation Channels/metabolism , Amino Acid Sequence , Animals , Binding Sites , Humans , Models, Biological , Mutagenesis, Site-Directed , Rats , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics
16.
Biol Trace Elem Res ; 151(3): 451-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23264033

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is a non-selective cation channel involved in pain sensation and in a wide range of non-pain-related physiological and pathological conditions. The aim of the present study was to explore the effects of selected heavy metal cations on the function of TRPV1. The cations ranked in the following sequence of pore-blocking activity: Co(2+) [half-maximal inhibitory concentration (IC(50)) = 13 µM] > Cd(2+) (I (50) = 38 µM) > Ni(2+) (IC(50) = 62 µM) > Cu(2+) (IC(50) = 200 µM). Zn(2+) proved to be a weak (IC(50) = 27 µM) and only partial inhibitor of the channel function, whereas Mg(2+), Mn(2+) and La(3+) did not exhibit any substantial effect. Co(2+), the most potent channel blocker, was able not only to compete with Ca(2+) but also to pass with it through the open channel of TRPV1. In response to heat activation or vanilloid treatment, Co(2+) accumulation was verified in TRPV1-transfected cell lines and in the TRPV1+ dorsal root ganglion neurons. The inhibitory effect was also demonstrated in vivo. Co(2+) applied together with vanilloid agonists attenuated the nocifensive eye wipe response in mice. Different rat TRPV1 pore point mutants (Y627W, N628W, D646N and E651W) were created that can validate the binding site of previously used channel blockers in agonist-evoked (45)Ca(2+) influx assays in cells expressing TRPV1. The IC(50) of Co(2+) on these point mutants were determined to be reasonably comparable to those on the wild type, which suggests that divalent cations passing through the TRPV1 channel use the same negatively charged amino acids as Ca(2+).


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Metals, Heavy/pharmacology , TRPV Cation Channels/antagonists & inhibitors , 3T3 Cells , Animals , COS Cells , Calcium/metabolism , Calcium Channel Blockers/chemistry , Cations, Divalent/chemistry , Cations, Divalent/pharmacology , Cell Line , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Metals, Heavy/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Rats , Structure-Activity Relationship , TRPV Cation Channels/metabolism
17.
Can J Neurol Sci ; 36(2): 234-41, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19378721

ABSTRACT

OBJECTIVES: Resiniferatoxin, the most potent agonist of inflammatory pain/vanilloid receptor/cation channel (TRPV1) can be used for neuron subtype specific ablation of pain generating cells at the level of the peripheral nervous system by Ca(2+)-excytotoxicity. Molecular neurosurgery is an emerging technology either to alleviate severe pain in cancer or treat/prevent different local neuropathies. Our aim was determining sensory modalities that may be lost after resiniferatoxin treatment. METHODS: Newborn or adult mice were treated with resiniferatoxin, then changes in chemical and heat sensitivity were correlated with alterations of the cell composition of sensory ganglions. RESULTS: Only mice treated at adult age became less sensitive to heat stimuli, while both treatment groups lost sensitivity to specific vanilloid agonists of TRPV1 and, interestingly, to allyl-isothiocyanate, a selective agonist of TRPA1. Our in vivo and post mortem analytical results confirmed that TRPV1 and TRPA1 function together and resiniferatoxin-mediated neurosurgery removes both sensor molecules. DISCUSSION: In adult mice resiniferatoxin causes: i) desensitization to heat and ii) sensitization to cold. Cold hyperalgesia, an imbalance in thermosensation, might be conferred by a prominent cold receptor that is expressed in surviving resiniferatoxin-resistant sensory neurons and compensates for pain signals lost with TRPA1 and TRPV1 double positive cells in the peripheral nervous system.


Subject(s)
Diterpenes/pharmacology , Pain/physiopathology , Sensory Receptor Cells/drug effects , TRPV Cation Channels/drug effects , Transient Receptor Potential Channels/drug effects , Animals , Blotting, Western , Cold Temperature , Hot Temperature , Immunohistochemistry , Mice , Pain Threshold/drug effects , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...