Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 58(11): 5772-5789, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34406600

ABSTRACT

Increasing evidence suggests that transmembrane protein 16A (TMEM16A) in nociceptive neurons is an important molecular component contributing to peripheral pain transduction. The present study aimed to evaluate the role and mechanism of TMEM16A in chronic nociceptive responses elicited by spared nerve injury (SNI). In this study, SNI was used to induce neuropathic pain. Drugs were administered intrathecally. The expression and cellular localization of TMEM16A, the ERK pathway, and NK-1 in the dorsal root ganglion (DRG) were detected by western blot and immunofluorescence. Behavioral tests were used to evaluate the role of TMEM16A and p-ERK in SNI-induced persistent pain and hypersensitivity. The role of TMEM16A in the hyperexcitability of primary nociceptor neurons was assessed by electrophysiological recording. The results show that TMEM16A, p-ERK, and NK-1 are predominantly expressed in small neurons associated with nociceptive sensation. TMEM16A is colocalized with p-ERK/NK-1 in DRG. TMEM16A, the MEK/ERK pathway, and NK-1 are activated in DRG after SNI. ERK inhibitor or TMEM16A antagonist prevents SNI-induced allodynia. ERK and NK-1 are downstream of TMEM16A activation. Electrophysiological recording showed that CaCC current increases and intrathecal application of T16Ainh-A01, a selective TMEM16A inhibitor, reverses the hyperexcitability of DRG neurons harvested from rats after SNI. We conclude that TMEM16A activation in DRG leads to a positive interaction of the ERK pathway with activation of NK-1 production and is involved in the development of neuropathic pain after SNI. Also, the blockade of TMEM16A or inhibition of the downstream ERK pathway or NK-1 upregulation may prevent the development of neuropathic pain.


Subject(s)
Anoctamins/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Ganglia, Spinal/pathology , Hyperalgesia/physiopathology , Neuralgia/physiopathology , Peroneal Nerve/injuries , Receptors, Neurokinin-1/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Tibial Nerve/injuries , Animals , Anoctamins/antagonists & inhibitors , Butadienes/pharmacology , Chronic Pain/etiology , Chronic Pain/physiopathology , Hyperalgesia/etiology , Ligation , Male , Neuralgia/etiology , Nitriles/pharmacology , Nociception/physiology , Pyrimidines/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacology
2.
Nature ; 594(7861): 88-93, 2021 06.
Article in English | MEDLINE | ID: mdl-33827113

ABSTRACT

COVID-19 is a disease with unique characteristics that include lung thrombosis1, frequent diarrhoea2, abnormal activation of the inflammatory response3 and rapid deterioration of lung function consistent with alveolar oedema4. The pathological substrate for these findings remains unknown. Here we show that the lungs of patients with COVID-19 contain infected pneumocytes with abnormal morphology and frequent multinucleation. The generation of these syncytia results from activation of the SARS-CoV-2 spike protein at the cell plasma membrane level. On the basis of these observations, we performed two high-content microscopy-based screenings with more than 3,000 approved drugs to search for inhibitors of spike-driven syncytia. We converged on the identification of 83 drugs that inhibited spike-mediated cell fusion, several of which belonged to defined pharmacological classes. We focused our attention on effective drugs that also protected against virus replication and associated cytopathicity. One of the most effective molecules was the antihelminthic drug niclosamide, which markedly blunted calcium oscillations and membrane conductance in spike-expressing cells by suppressing the activity of TMEM16F (also known as anoctamin 6), a calcium-activated ion channel and scramblase that is responsible for exposure of phosphatidylserine on the cell surface. These findings suggest a potential mechanism for COVID-19 disease pathogenesis and support the repurposing of niclosamide for therapy.


Subject(s)
Anoctamins/antagonists & inhibitors , COVID-19/pathology , Cell Fusion , Drug Evaluation, Preclinical , Giant Cells/drug effects , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Aged , Aged, 80 and over , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Animals , Anoctamins/metabolism , COVID-19/metabolism , COVID-19/virology , Calcium Signaling/drug effects , Cell Line , Chloride Channels/metabolism , Chlorocebus aethiops , Female , Giant Cells/metabolism , Giant Cells/virology , Humans , Lung/drug effects , Lung/pathology , Lung/virology , Male , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Virus Replication/drug effects
3.
Cancer Sci ; 112(3): 1026-1037, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33404124

ABSTRACT

The function of ANO9 in gastrointestinal cancer remains unclear. We investigated the biological behaviors and clinical prognostic values of ANO9 in gastric cancer (GC). Knockdown experiments were performed on human GC cell lines using ANO9 siRNA. Eighty-four primary tissue samples from patients with advanced GC were examined immunohistochemically (IHC). Knockdown of ANO9 reduced the progression of cancer cells in MKN7 and MKN74 cells. A microarray analysis revealed that ANO9 regulated PD-L2 via interferon (IFN)-related genes. We confirmed using flow cytometry that the depletion of ANO9 reduced the binding ability to PD-1 by downregulating the expression of PD-L2 in MKN7 and MKN74 cells. IHC revealed a correlation between the expression of ANO9 and PD-L2 and also that the strong expression of ANO9 was an independent poor prognostic factor in patients with advanced GC. The present results indicate that ANO9 regulates PD-L2 and binding ability to PD-1 via IFN-related genes in GC. Therefore, ANO9 has potential as a biomarker and target of immune checkpoint blockage (ICB) for GC.


Subject(s)
Anoctamins/metabolism , Biomarkers, Tumor/metabolism , Phospholipid Transfer Proteins/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Stomach Neoplasms/genetics , Aged , Anoctamins/antagonists & inhibitors , Anoctamins/genetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Follow-Up Studies , Gastrectomy , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interferons/metabolism , Male , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/genetics , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Stomach Neoplasms/therapy , Survival Rate
4.
J Biol Chem ; 295(35): 12537-12544, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32709749

ABSTRACT

TMEM16 Ca2+-activated phospholipid scramblases (CaPLSases) mediate rapid transmembrane phospholipid flip-flop and as such play essential roles in various physiological and pathological processes such as blood coagulation, skeletal development, viral infection, cell-cell fusion, and ataxia. Pharmacological tools specifically targeting TMEM16 CaPLSases are urgently needed to understand these novel membrane transporters and their contributions to health and disease. Tannic acid (TA) and epigallocatechin gallate (EGCG) were recently reported as promising TMEM16F CaPLSase inhibitors. However, our present study shows that TA and EGCG do not inhibit the phospholipid-scrambling or ion conduction activities of the dual-functional TMEM16F. Instead, we found that TA and EGCG mainly acted as fluorescence quenchers that rapidly suppress the fluorophores conjugated to annexin V, a phosphatidylserine-binding probe commonly used to report on TMEM16 CaPLSase activity. These data demonstrate the false positive effects of TA and EGCG on inhibiting TMEM16F phospholipid scrambling and discourage the use of these polyphenols as CaPLSase inhibitors. Appropriate controls as well as a combination of both fluorescence imaging and electrophysiological validation are necessary in future endeavors to develop TMEM16 CaPLSase inhibitors.


Subject(s)
Anoctamins/chemistry , Phospholipid Transfer Proteins/chemistry , Phospholipids/chemistry , Animals , Anoctamins/antagonists & inhibitors , Anoctamins/metabolism , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Mice , Phospholipid Transfer Proteins/antagonists & inhibitors , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Polyphenols/chemistry , Polyphenols/pharmacology , Tannins/chemistry , Tannins/pharmacology
5.
PLoS One ; 15(4): e0231812, 2020.
Article in English | MEDLINE | ID: mdl-32302365

ABSTRACT

TMEM16A, a Ca2+-sensitive Cl- channel, plays key roles in many physiological functions related to Cl- transport across lipid membranes. Activation of this channel is mediated via binding intracellular Ca2+ to the channel with a relatively high apparent affinity, roughly in the sub-µM to low µM concentration range. Recently available high-resolution structures of TMEM16 molecules reveal that the high-affinity Ca2+ activation sites are formed by several acidic amino acids, using their negatively charged sidechain carboxylates to coordinate the bound Ca2+. In this study, we examine the interaction of TMEM16A with a divalent cation, Co2+, which by itself cannot activate current in TMEM16A. This divalent cation, however, has two effects when applied intracellularly. It inhibits the Ca2+-induced TMEM16A current by competing with Ca2+ for the aforementioned high-affinity activation sites. In addition, Co2+ also potentiates the Ca2+-induced current with a low affinity. This potentiation effect requires high concentration (mM) of Co2+, similar to our previous findings that high concentrations (mM) of intracellular Ca2+ ([Ca2+]i) can induce more TMEM16A current after the Ca2+-activation sites are saturated by tens of µM [Ca2+]i. The degrees of potentiation by Co2+ and Ca2+ also roughly correlate with each other. Interestingly, mutating a pore residue of TMEM16A, Y589, alters the degree of potentiation in that the smaller the sidechain of the replaced residue, the larger the potentiation induced by divalent cations. We suggest that the Co2+ potentiation and the Ca2+ potentiation share a similar mechanism by increasing Cl- flux through the channel pore, perhaps due to an increase of positive pore potential after the binding of divalent cations to phospholipids in the pore. A smaller sidechain of a pore residue may allow the pore to accommodate more phospholipids, thus enhancing the current potentiation caused by high concentrations of divalent cations.


Subject(s)
Anoctamins/agonists , Anoctamins/antagonists & inhibitors , Cobalt/pharmacology , Ion Channel Gating/drug effects , Anoctamins/metabolism , Calcium , Chloride Channel Agonists/pharmacology , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Ions , Kinetics , Mutant Proteins/metabolism
6.
JCI Insight ; 4(15)2019 08 08.
Article in English | MEDLINE | ID: mdl-31391337

ABSTRACT

Inflammatory airway diseases, such as asthma, cystic fibrosis (CF), and chronic obstructive pulmonary disease (COPD), are characterized by mucus hypersecretion and airway plugging. In both CF and asthma, enhanced expression of the Ca2+-activated Cl- channel TMEM16A is detected in mucus-producing club/goblet cells and airway smooth muscle. TMEM16A contributes to mucus hypersecretion and bronchoconstriction, which are both inhibited by blockers of TMEM16A, such as niflumic acid. Here we demonstrate that the FDA-approved drug niclosamide, a potent inhibitor of TMEM16A identified by high-throughput screening, is an inhibitor of both TMEM16A and TMEM16F. In asthmatic mice, niclosamide reduced mucus production and secretion, as well as bronchoconstriction, and showed additional antiinflammatory effects. Using transgenic asthmatic mice, we found evidence that TMEM16A and TMEM16F are required for normal mucus production/secretion, which may be due to their effects on intracellular Ca2+ signaling. TMEM16A and TMEM16F support exocytic release of mucus and inflammatory mediators, both of which are blocked by niclosamide. Thus, inhibition of mucus and cytokine release, bronchorelaxation, and reported antibacterial effects make niclosamide a potentially suitable drug for the treatment of inflammatory airway diseases, such as CF, asthma, and COPD.


Subject(s)
Anoctamins/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Asthma/drug therapy , Bronchi/drug effects , Mucus/metabolism , Niclosamide/pharmacology , Animals , Anoctamins/genetics , Anoctamins/metabolism , Anti-Inflammatory Agents/therapeutic use , Asthma/immunology , Asthma/pathology , Bronchi/metabolism , Cell Line, Tumor , Cystic Fibrosis/drug therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/pathology , Disease Models, Animal , Drug Repositioning , Goblet Cells/drug effects , Goblet Cells/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Mice, Transgenic , Niclosamide/therapeutic use , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction
7.
Biochem Biophys Res Commun ; 517(4): 603-610, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31409484

ABSTRACT

Spinal cord injury (SCI) leads to sensorimotor deficits and autonomic changes. Macrophages and microglia could be polarized into the classically activated pro-inflammatory M1 phenotype or the alternatively activated anti-inflammatory M2 phenotype. Transmembrane protein with unknown function 16F (TMEM16F) exhibits functional diversity and may contribute to microglial function. However, the effects of TMEM16F on the modulation of macrophage/microglial polarization are still not fully understood. In the study, TMEM16F up-regulation was detected after SCI in mice, and TMEM16F protein was found in macrophages/microglia in injured spinal cord sections. Depletion of TMEM16F improved motor function in male mice with SCI. M1-type macrophages/microglia accumulated in lower numbers in the injured spinal cord of TMEM16F-knockout (KO) mice. M2 polarization inhibited by SCI was improved in mice with TMEM16F deficiency. TMEM16F deletion also attenuated microglial/macrophage pro-inflammatory response. Furthermore, significant down-regulation of A disintegrin and metalloprotease 17 (ADAM17) was observed in TMEM16F-KO mice. Importantly, TMEM16F-promoted M1 polarization and -inhibited M1 polarization were largely associated with the suppression of ADAM17. Overall, our findings provided new insights into the regulatory mechanisms of macrophage/microglial polarization, thereby possibly facilitating the development of new therapeutic strategies for SCI through the regulation of TMEM16F/ADAM17 signaling.


Subject(s)
Anoctamins/antagonists & inhibitors , Behavior, Animal , Cell Polarity , Microglia/metabolism , Microglia/pathology , Motor Activity , Pain/metabolism , Phospholipid Transfer Proteins/antagonists & inhibitors , ADAM17 Protein/metabolism , Animals , Anoctamins/metabolism , Cell Line , Gene Deletion , Macrophage Activation , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phospholipid Transfer Proteins/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/prevention & control
8.
Platelets ; 30(8): 962-966, 2019.
Article in English | MEDLINE | ID: mdl-31008669

ABSTRACT

TMEM16F is a surface membrane protein critical for platelet procoagulant activity, which exhibits both phospholipid scramblase and ion channel activities following sustained elevation of cytosolic Ca2+. The extent to which the ionic permeability of TMEM16F is important for platelet scramblase responses remains controversial. To date, only one study has reported the electrophysiological properties of TMEM16F in cells of platelet/megakaryocyte lineage, which observed cation-selectivity within excised patch recordings from murine marrow-derived megakaryocytes. This contrasts with reports using whole-cell recordings that describe this channel as displaying either selectivity for anions or being relatively non-selective amongst the major physiological monovalent ions. We have studied TMEM16F expression and channel activity in primary rat and mouse megakaryocytes and the human erythroleukemic (HEL) cell line that exhibits megakaryocytic surface markers. Immunocytochemical analysis was consistent with surface TMEM16F expression in cells from all three species. Whole-cell recordings in the absence of K+-selective currents revealed an outwardly rectifying conductance activated by a high intracellular Ca2+ concentration in all three species. These currents appeared after 5-6 minutes and were blocked by CaCCinh-A01, properties typical of TMEM16F. Ion substitution experiments showed that the underlying conductance was predominantly Cl--permeable in rat megakaryocytes and HEL cells, yet non-selective between monovalent anions and cations in mouse megakaryocytes. In conclusion, the present study further highlights the difference in ionic selectivity of TMEM16F in platelet lineage cells of the mouse compared to other mammalian species. This provides additional support for the ionic "leak" hypothesis that the scramblase activity of TMEM16F does not rely upon its ability to conduct ions of a specific type.


Subject(s)
Anoctamins/antagonists & inhibitors , Calcium/metabolism , Megakaryocytes/metabolism , Phospholipid Transfer Proteins/antagonists & inhibitors , Animals , Biological Transport , Humans , Mice , Rats
9.
Br J Pharmacol ; 174(18): 2984-2999, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28616863

ABSTRACT

BACKGROUND AND PURPOSE: Ca2+ -activated Cl- channels (CaCCs) are gated open by a rise in intracellular Ca2+ concentration ([Ca2+ ]i ), typically provoked by activation of Gq -protein coupled receptors (Gq PCR). Gq PCR activation initiates depletion of plasmalemmal phosphatidylinositol 4,5-bisphosphate (PIP2 ). Here, we determined whether PIP2 acts as a signalling lipid for CaCCs coded by the TMEM16A and TMEM16B genes. EXPERIMENTAL APPROACH: Patch-clamp electrophysiology, in conjunction with genetically encoded systems to control cellular PIP2 content, was used to define the mechanism of action of PIP2 on TMEM16A and TMEM16B channels. KEY RESULTS: A water-soluble PIP2 analogue (diC8-PIP2 ) activated TMEM16A channels by up to fivefold and inhibited TMEM16B by ~0.2-fold. The effects of diC8-PIP2 on TMEM16A currents were especially pronounced at low [Ca2+ ]i . In contrast, diC8-PIP2 modulation of TMEM16B channels did not vary over a broad [Ca2+ ]i range but was only detectable at highly depolarized membrane potentials. Modulation of TMEM16A and TMEM16B currents was due to changes in channel gating, while single channel conductance was unaltered. Co-expression of TMEM16A or TMEM16B with a Danio rerio voltage-sensitive phosphatase (DrVSP), which degrades PIP2 , led to reduction and enhancement of TMEM16A and TMEM16B currents respectively. These effects were abolished by an inactivating mutation in DrVSP and antagonized by simultaneous co-expression of a phosphatidylinositol-4-phosphate 5-kinase that catalyses PIP2 formation. CONCLUSIONS AND IMPLICATIONS: PIP2 acts as a modifier of TMEM16A and TMEM16B channel gating. Drugs interacting with PIP2 signalling may affect TMEM16A and TMEM16B channel gating and have potential uses in basic science and implications for therapy.


Subject(s)
Anoctamin-1/metabolism , Anoctamins/antagonists & inhibitors , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Animals , Anoctamins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Mice , Phosphatidylinositol 4,5-Diphosphate/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...