Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
2.
Br J Cancer ; 130(9): 1415-1419, 2024 May.
Article in English | MEDLINE | ID: mdl-38424164

ABSTRACT

BACKGROUND: Multi-faceted evidence from a range of cancers suggests strongly that de novo expression of voltage-gated sodium channels (VGSCs) plays a significant role in driving cancer cell invasiveness. Under hypoxic conditions, common to growing tumours, VGSCs develop a persistent current (INaP) which can be blocked selectively by ranolazine. METHODS: Several different carcinomas were examined. We used data from a range of experimental approaches relating to cellular invasiveness and metastasis. These were supplemented by survival data mined from cancer patients. RESULTS: In vitro, ranolazine inhibited invasiveness of cancer cells especially under hypoxia. In vivo, ranolazine suppressed the metastatic abilities of breast and prostate cancers and melanoma. These data were supported by a major retrospective epidemiological study on breast, colon and prostate cancer patients. This showed that risk of dying from cancer was reduced by ca.60% among those taking ranolazine, even if this started 4 years after the diagnosis. Ranolazine was also shown to reduce the adverse effects of chemotherapy on heart and brain. Furthermore, its anti-cancer effectiveness could be boosted by co-administration with other drugs. CONCLUSIONS: Ranolazine, alone or in combination with appropriate therapies, could be reformulated as a safe anti-metastatic drug offering many potential advantages over current systemic treatment modalities.


Subject(s)
Ranolazine , Ranolazine/pharmacology , Ranolazine/therapeutic use , Humans , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/drug effects , Male , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Female , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Neoplasm Invasiveness , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
3.
Nature ; 625(7995): 557-565, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38172636

ABSTRACT

Osteoarthritis (OA) is the most common joint disease. Currently there are no effective methods that simultaneously prevent joint degeneration and reduce pain1. Although limited evidence suggests the existence of voltage-gated sodium channels (VGSCs) in chondrocytes2, their expression and function in chondrocytes and in OA remain essentially unknown. Here we identify Nav1.7 as an OA-associated VGSC and demonstrate that human OA chondrocytes express functional Nav1.7 channels, with a density of 0.1 to 0.15 channels per µm2 and 350 to 525 channels per cell. Serial genetic ablation of Nav1.7 in multiple mouse models demonstrates that Nav1.7 expressed in dorsal root ganglia neurons is involved in pain, whereas Nav1.7 in chondrocytes regulates OA progression. Pharmacological blockade of Nav1.7 with selective or clinically used pan-Nav channel blockers significantly ameliorates the progression of structural joint damage, and reduces OA pain behaviour. Mechanistically, Nav1.7 blockers regulate intracellular Ca2+ signalling and the chondrocyte secretome, which in turn affects chondrocyte biology and OA progression. Identification of Nav1.7 as a novel chondrocyte-expressed, OA-associated channel uncovers a dual target for the development of disease-modifying and non-opioid pain relief treatment for OA.


Subject(s)
Chondrocytes , NAV1.7 Voltage-Gated Sodium Channel , Osteoarthritis , Voltage-Gated Sodium Channel Blockers , Animals , Humans , Mice , Calcium/metabolism , Calcium Signaling/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Progression , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , NAV1.7 Voltage-Gated Sodium Channel/deficiency , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neurons/metabolism , Osteoarthritis/complications , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Osteoarthritis/metabolism , Pain/complications , Pain/drug therapy , Pain/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
4.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614292

ABSTRACT

The voltage-gated sodium channels represent an important target for drug discovery since a large number of physiological processes are regulated by these channels. In several excitability disorders, including epilepsy, cardiac arrhythmias, chronic pain, and non-dystrophic myotonia, blockers of voltage-gated sodium channels are clinically used. Myotonia is a skeletal muscle condition characterized by the over-excitability of the sarcolemma, resulting in delayed relaxation after contraction and muscle stiffness. The therapeutic management of this disorder relies on mexiletine and other sodium channel blockers, which are not selective for the Nav1.4 skeletal muscle sodium channel isoform. Hence, the importance of deepening the knowledge of molecular requirements for developing more potent and use-dependent drugs acting on Nav1.4. Here, we review the available treatment options for non-dystrophic myotonia and the structure-activity relationship studies performed in our laboratory with a focus on new compounds with potential antimyotonic activity.


Subject(s)
Mexiletine , Myotonia , NAV1.4 Voltage-Gated Sodium Channel , Voltage-Gated Sodium Channel Blockers , Humans , Mexiletine/pharmacology , Mexiletine/therapeutic use , Muscle, Skeletal/drug effects , Myotonia/drug therapy , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Syndrome , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
5.
Am J Manag Care ; 28(9 Suppl): S159-S165, 2022 09.
Article in English | MEDLINE | ID: mdl-36198074

ABSTRACT

Cough serves a protective physiologic function as a vital defensive reflex preventing aspiration. However, exposure to viral infections or other triggers induces, in some individuals, a chronic cough (CC) that causes a significant symptomatic burden. Most cases of CC are due to conditions that respond to appropriate therapeutic trials (upper airway cough syndrome; asthma; reflux). Unfortunately, a significant subgroup of patients will have refractory CC, which does not respond to treatment of known underlying causes of CC. Currently, available therapeutic options for refractory CC are inadequate due to limited efficacy and frequently intolerable adverse effects. Current professional society guideline recommendations are discussed, and a promising pipeline of antitussive drugs in development is introduced, including purinergic 2X3 receptor antagonists, neurokinin-1 receptor antagonists, oral mixed ĸ-opioid receptor agonists/µ-opioid receptor antagonists, and voltage-gated sodium channel blockers.


Subject(s)
Antitussive Agents , Analgesics, Opioid/therapeutic use , Antitussive Agents/pharmacology , Antitussive Agents/therapeutic use , Chronic Disease , Cough/drug therapy , Cough/etiology , Humans , Narcotic Antagonists/therapeutic use , Neurokinin-1 Receptor Antagonists/therapeutic use , Receptors, Opioid/therapeutic use , Voltage-Gated Sodium Channel Blockers/therapeutic use
6.
Zool Res ; 43(5): 886-896, 2022 Sep 18.
Article in English | MEDLINE | ID: mdl-36052553

ABSTRACT

Various peptide toxins in animal venom inhibit voltage-gated sodium ion channel Nav1.7, including Nav-targeting spider toxin (NaSpTx) Family I. Toxins in NaSpTx Family I share a similar structure, i.e., N-terminal, loops 1-4, and C-terminal. Here, we used Mu-theraphotoxin-Ca2a (Ca2a), a peptide isolated from Cyriopagopus albostriatus, as a template to investigate the general properties of toxins in NaSpTx Family I. The toxins interacted with the cell membrane prior to binding to Nav1.7 via similar hydrophobic residues. Residues in loop 1, loop 4, and the C-terminal primarily interacted with the S3-S4 linker of domain II, especially basic amino acids binding to E818. We also identified the critical role of loop 2 in Ca2a regarding its affinity to Nav1.7. Our results provide further evidence that NaSpTx Family I toxins share similar structures and mechanisms of binding to Nav1.7.


Subject(s)
Spider Venoms , Animals , Peptides/chemistry , Sodium Channels , Spider Venoms/chemistry , Spider Venoms/genetics , Spider Venoms/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
7.
Bioorg Med Chem Lett ; 75: 128946, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35985458

ABSTRACT

The voltage-gated sodium (Nav) channel is one of most important targets for treatment of epilepsy, and rufinamide is an approved third-generation anti-seizure drug as Nav1.1 channel blocker. Herein, by triazenylation of rufinamide, we reported the triazenyl triazoles as new Nav1.1 channel blocker for treatment of epilepsy. Through the electrophysiological activity assay, compound 6a and 6e were found to modulate the inactivation voltage of Nav 1.1 channel with shift of -10.07 mv and -11.28 mV, respectively. In the pentylenetetrazole (PTZ) mouse model, 6a and 6e reduced the seizure level, prolonged seizure latency and improved the survival rate of epileptic mice at an intragastric administration of 50 mg/kg dosage. In addition, 6a also exhibited promising effectiveness in the maximal electroshock (MES) mouse model and possessed moderate pharmacokinetic profiles. These results demonstrated that 6a was a novel Nav1.1 channel blocker for treatment of epilepsy.


Subject(s)
Epilepsy , Pentylenetetrazole , Animals , Disease Models, Animal , Epilepsy/drug therapy , Mice , Sodium , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
8.
Biomed Pharmacother ; 147: 112671, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35104697

ABSTRACT

When peripheral neuropathy occurs due to chemotherapy treatment, it is referred to as chemotherapy-induced peripheral neuropathy (CIPN). Typically, symptoms are sensory rather than motor and include reduced feeling and heightened sensitivity to pressure, pain, temperature, and touch. The pathophysiology of CIPN is very complex, and it involves multiple mechanisms leading to its development which will be described specifically for each chemotherapeutic class. There are currently no approved or effective agents for CIPN prevention, and Duloxetine is the only medication that is an effective treatment against CIPN. There is an unavoidable necessity to develop preventative and treatment approaches for CIPN due to its detrimental impact on patients' lives. The purpose of this review is to examine CIPN, innovative pharmacological and nonpharmacological therapy and preventive strategies for this illness, and future perspectives for this condition and its therapies.


Subject(s)
Antineoplastic Agents/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/therapy , Analgesics/therapeutic use , Antioxidants/therapeutic use , Complementary Therapies , Humans , Neuroprotective Agents/therapeutic use , Patient Acuity , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/drug therapy , Risk Factors , Serotonin and Noradrenaline Reuptake Inhibitors/therapeutic use , Voltage-Gated Sodium Channel Blockers/therapeutic use
9.
Respir Physiol Neurobiol ; 299: 103856, 2022 05.
Article in English | MEDLINE | ID: mdl-35114369

ABSTRACT

Cough in chronic respiratory diseases is a common symptom associated with significant comorbidities including visceral pain. Available antitussive therapy still has limited efficacy. Recent advances in the understanding of voltage-gated sodium channels (NaVs) lead to the rational hypothesis that subtype NaV1.7 is involved in initiating cough and thus may present a promising therapeutic target for antitussive therapy. We evaluated the antitussive effect of NaV1.7 blocker PF-05089771 administered systemically and topically in awake guinea pigs using capsaicin cough challenge. Compared to vehicle, peroral or inhaled PF-05089771 administration caused about 50-60 % inhibition of cough at the doses that did not alter respiratory rate. We conclude that the NaV1.7 blocker PF-05089771 inhibits cough in a manner consistent with its electrophysiological effect on airway C-fibre nerve terminals.


Subject(s)
Antitussive Agents , Voltage-Gated Sodium Channels , Animals , Antitussive Agents/pharmacology , Antitussive Agents/therapeutic use , Cough/drug therapy , Guinea Pigs , Phenyl Ethers , Sulfonamides , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use , Voltage-Gated Sodium Channels/physiology
10.
Biochem Soc Trans ; 49(5): 1941-1961, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34643236

ABSTRACT

Voltage-dependent Na+ channel activation underlies action potential generation fundamental to cellular excitability. In skeletal and cardiac muscle this triggers contraction via ryanodine-receptor (RyR)-mediated sarcoplasmic reticular (SR) Ca2+ release. We here review potential feedback actions of intracellular [Ca2+] ([Ca2+]i) on Na+ channel activity, surveying their structural, genetic and cellular and functional implications, translating these to their possible clinical importance. In addition to phosphorylation sites, both Nav1.4 and Nav1.5 possess potentially regulatory binding sites for Ca2+ and/or the Ca2+-sensor calmodulin in their inactivating III-IV linker and C-terminal domains (CTD), where mutations are associated with a range of skeletal and cardiac muscle diseases. We summarize in vitro cell-attached patch clamp studies reporting correspondingly diverse, direct and indirect, Ca2+ effects upon maximal Nav1.4 and Nav1.5 currents (Imax) and their half-maximal voltages (V1/2) characterizing channel gating, in cellular expression systems and isolated myocytes. Interventions increasing cytoplasmic [Ca2+]i down-regulated Imax leaving V1/2 constant in native loose patch clamped, wild-type murine skeletal and cardiac myocytes. They correspondingly reduced action potential upstroke rates and conduction velocities, causing pro-arrhythmic effects in intact perfused hearts. Genetically modified murine RyR2-P2328S hearts modelling catecholaminergic polymorphic ventricular tachycardia (CPVT), recapitulated clinical ventricular and atrial pro-arrhythmic phenotypes following catecholaminergic challenge. These accompanied reductions in action potential conduction velocities. The latter were reversed by flecainide at RyR-blocking concentrations specifically in RyR2-P2328S as opposed to wild-type hearts, suggesting a basis for its recent therapeutic application in CPVT. We finally explore the relevance of these mechanisms in further genetic paradigms for commoner metabolic and structural cardiac disease.


Subject(s)
Calcium Signaling , Calcium/metabolism , Ion Channel Gating , Myocytes, Cardiac/metabolism , NAV1.4 Voltage-Gated Sodium Channel/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium/metabolism , Action Potentials , Animals , Binding Sites , Disease Models, Animal , Flecainide/therapeutic use , Humans , Mice , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Treatment Outcome , Voltage-Gated Sodium Channel Blockers/therapeutic use
11.
Neurotherapeutics ; 18(3): 1490-1499, 2021 07.
Article in English | MEDLINE | ID: mdl-34431030

ABSTRACT

Genetic testing has yielded major advances in our understanding of the causes of epilepsy. Seizures remain resistant to treatment in a significant proportion of cases, particularly in severe, childhood-onset epilepsy, the patient population in which an underlying causative genetic variant is most likely to be identified. A genetic diagnosis can be explanatory as to etiology, and, in some cases, might suggest a therapeutic approach; yet, a clear path from genetic diagnosis to treatment remains unclear in most cases. Here, we discuss theoretical considerations behind the attempted use of small molecules for the treatment of genetic epilepsies, which is but one among various approaches currently under development. We explore a few salient examples and consider the future of the small molecule approach for genetic epilepsies. We conclude that significant additional work is required to understand how genetic variation leads to dysfunction of epilepsy-associated protein targets, and how this impacts the function of diverse subtypes of neurons embedded within distributed brain circuits to yield epilepsy and epilepsy-associated comorbidities. A syndrome- or even variant-specific approach may be required to achieve progress. Advances in the field will require improved methods for large-scale target validation, compound identification and optimization, and the development of accurate model systems that reflect the core features of human epilepsy syndromes, as well as novel approaches towards clinical trials of such compounds in small rare disease cohorts.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsy/genetics , Epilepsy/therapy , Genetic Testing/methods , Genetic Therapy/methods , Voltage-Gated Sodium Channel Blockers/therapeutic use , Clinical Trials as Topic/methods , Epilepsy/diagnosis , Humans , NAV1.6 Voltage-Gated Sodium Channel/genetics , Nerve Tissue Proteins/genetics , Phenytoin/therapeutic use , Potassium Channels, Sodium-Activated/genetics , Quinidine/therapeutic use
12.
J Ethnopharmacol ; 281: 114495, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34364968

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Allium macrostemon Bunge. is an edible Chinese herb traditionally used for the treatment of thoracic pain, stenocardia, heart asthma and diarrhea. Although its biological potential has been extensively proven such as antioxidant activity, antiplatelet aggregation, vasodilation and antidepressant-like activity, there are no reports in the literature regarding its pharmacological analgesic activity. AIM OF THE STUDY: The study was carried out to examine the anti-nociceptive activity of the crude extract of A. macrostemon bulbs and interpret its likely molecular target. MATERIALS AND METHODS: The bulbs of A. macrostemon were gathered, dried-up, and extracted with water (AMWD). AMWD was subjected to activity testing, using chemical-induced (acetic acid and formalin test) and heat-induced (hot plate) pain models. To evaluate the likely mechanistic strategy involved in the analgesic effect of AMWD, whole-cell patch clamp recordings were conducted in acutely dissociated dorsal root ganglion (DRG) neurons and human embryonic kidney 293T (HEK293T) cells expressing pain-related receptors. Electrophysiological methods were employed to detect the action potentials of DRG neurons and potential targets of A. macrostemon. RESULTS: AMWD showed significant palliative effect in all heat and chemical induced pain assays. Moreover, AMWD significantly reduces the excitability of dorsal root ganglion neurons by reducing the firing frequency of action potentials. Further analysis revealed that voltage-gated sodium channel Nav1.7 is the potential target of A. macrostemon for its analgesic activity. CONCLUSION: This study has brought new scientific evidence of preclinical efficacy of A. macrostemon as an anti-nociceptive agent. Apparently, these effects are involved with the inhibition of the voltage-sensitive Nav1.7 channel contributing to the reduction of peripheral neuronal excitability. Our present study justifies the folkloric usage of A. macrostemon as a remedy for several pain states. Furthermore, A. macrostemon is a good resource for the development of analgesic drugs targeting Nav1.7 channel.


Subject(s)
Analgesics/therapeutic use , Chive , Pain/drug therapy , Plant Extracts/therapeutic use , Voltage-Gated Sodium Channel Blockers/therapeutic use , Acetic Acid , Analgesics/pharmacology , Animals , Cell Survival/drug effects , Formaldehyde , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , HEK293 Cells , Hot Temperature , Humans , Locomotion/drug effects , Male , Mice, Inbred C57BL , NAV1.7 Voltage-Gated Sodium Channel/physiology , Nociceptors/physiology , Pain/etiology , Plant Extracts/pharmacology , Plant Roots , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Voltage-Gated Sodium Channel Blockers/pharmacology
13.
Acta Pharmacol Sin ; 42(8): 1235-1247, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34103689

ABSTRACT

Voltage-gated sodium channel Nav1.7 robustly expressed in peripheral nociceptive neurons has been considered as a therapeutic target for chronic pain, but there is no selective Nav1.7 inhibitor available for therapy of chronic pain. Ralfinamide has shown anti-nociceptive activity in animal models of inflammatory and neuropathic pain and is currently under phase III clinical trial for neuropathic pain. Based on ralfinamide, a novel small molecule (S)-2-((3-(4-((2-fluorobenzyl) oxy) phenyl) propyl) amino) propanamide (QLS-81) was synthesized. Here, we report the electrophysiological and pharmacodynamic characterization of QLS-81 as a Nav1.7 channel inhibitor with promising anti-nociceptive activity. In whole-cell recordings of HEK293 cells stably expressing Nav1.7, QLS-81 (IC50 at 3.5 ± 1.5 µM) was ten-fold more potent than its parent compound ralfinamide (37.1 ± 2.9 µM) in inhibiting Nav1.7 current. QLS-81 inhibition on Nav1.7 current was use-dependent. Application of QLS-81 (10 µM) caused a hyperpolarizing shift of the fast and slow inactivation of Nav1.7 channel about 7.9 mV and 26.6 mV, respectively, and also slowed down the channel fast and slow inactivation recovery. In dissociated mouse DRG neurons, QLS-81 (10 µM) inhibited native Nav current and suppressed depolarizing current pulse-elicited neuronal firing. Administration of QLS-81 (2, 5, 10 mg· kg-1· d-1, i.p.) in mice for 10 days dose-dependently alleviated spinal nerve injury-induced neuropathic pain and formalin-induced inflammatory pain. In addition, QLS-81 (10 µM) did not significantly affect ECG in guinea pig heart ex vivo; and administration of QLS-81 (10, 20 mg/kg, i.p.) in mice had no significant effect on spontaneous locomotor activity. Taken together, our results demonstrate that QLS-81, as a novel Nav1.7 inhibitor, is efficacious on chronic pain in mice, and it may hold developmental potential for pain therapy.


Subject(s)
Analgesics/therapeutic use , Fluorobenzenes/therapeutic use , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neuralgia/drug therapy , Voltage-Gated Sodium Channel Blockers/therapeutic use , Action Potentials/drug effects , Animals , Formaldehyde , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Guinea Pigs , HEK293 Cells , Humans , Inflammation/chemically induced , Inflammation/complications , Male , Mice, Inbred C57BL , Neuralgia/chemically induced , Neuralgia/etiology , Neurons/drug effects , Spinal Nerves/injuries
14.
Neurosci Lett ; 753: 135844, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33775738

ABSTRACT

The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.


Subject(s)
Analgesics/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Pain/genetics , Small Fiber Neuropathy/complications , Analgesics/therapeutic use , Animals , Disease Models, Animal , Gain of Function Mutation , Humans , Loss of Function Mutation , Mice , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nociception/drug effects , Pain/drug therapy , Rats , Rats, Transgenic , Small Fiber Neuropathy/drug therapy , Small Fiber Neuropathy/genetics , Voltage-Gated Sodium Channel Agonists/pharmacology , Voltage-Gated Sodium Channel Agonists/therapeutic use , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/therapeutic use
15.
BMC Cardiovasc Disord ; 21(1): 137, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33722203

ABSTRACT

BACKGROUND: Supraventricular tachycardias (SVTs) are common in the first year of life and may be life-threatening. Acute cardioversion is usually effective, with both pharmacological and non-pharmacological procedures. However, as yet no international consensus exists concerning the best drug required for a stable conversion to sinus rhythm (maintenance treatment). Our study intends to describe the experience of a single centre with maintenance drug treatment of both re-entry and automatic SVTs in the first year of life. METHODS: From March 1995 to April 2019, 55 patients under one year of age with SVT were observed in our Centre. The SVTs were divided into two groups: 45 re-entry and 10 automatic tachycardias. As regards maintenance therapy, in re-entry tachycardias, we chose to start with oral flecainide and in case of relapses switched to combined treatment with beta-blockers or digoxin. In automatic tachycardias we first administered a beta-blocker, later combined with flecainide or amiodarone when ineffective. RESULTS: The patients' median follow-up time was 35 months. In re-entry tachycardias, flecainide was effective as monotherapy in 23/45 patients (51.1%) and in 20/45 patients (44.4%) in combination with nadolol, sotalol or digoxin (overall 95.5%). In automatic tachycardias, a beta-blocker alone was effective in 3/10 patients (30.0%), however, the best results were obtained when combined with flecainide: overall 9/10 (90%). CONCLUSIONS: In this retrospective study on pharmacological treatment of SVTs under 1 year of age the combination of flecainide and beta-blockers was highly effective in long-term maintenance of sinus rhythm in both re-entry and automatic tachycardias.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Heart Rate/drug effects , Tachycardia, Supraventricular/drug therapy , Action Potentials , Adrenergic beta-Antagonists/therapeutic use , Age Factors , Anti-Arrhythmia Agents/adverse effects , Digoxin/therapeutic use , Drug Therapy, Combination , Female , Flecainide/therapeutic use , Humans , Infant , Infant, Newborn , Male , Nadolol/therapeutic use , Recurrence , Retrospective Studies , Sotalol/therapeutic use , Tachycardia, Supraventricular/diagnosis , Tachycardia, Supraventricular/physiopathology , Time Factors , Treatment Outcome , Voltage-Gated Sodium Channel Blockers/therapeutic use
16.
Heart Rhythm ; 18(7): 1212-1220, 2021 07.
Article in English | MEDLINE | ID: mdl-33737232

ABSTRACT

BACKGROUND: Although atrial fibrillation ablation is increasingly used for rhythm control therapy, antiarrhythmic drugs (AADs) are commonly used, either alone or in combination with ablation. The effectiveness of AADs is highly variable. Previous work from our group suggests that alterations in atrial resting membrane potential (RMP) induced by low Pitx2 expression could explain the variable effect of flecainide. OBJECTIVE: The purpose of this study was to assess whether alterations in atrial/cardiac RMP modify the effectiveness of multiple clinically used AADs. METHODS: The sodium channel blocking effects of propafenone (300 nM, 1 µM), flecainide (1 µM), and dronedarone (5 µM, 10 µM) were measured in human stem cell-derived cardiac myocytes, HEK293 expressing human NaV1.5, primary murine atrial cardiac myocytes, and murine hearts with reduced Pitx2c. RESULTS: A more positive atrial RMP delayed INa recovery, slowed channel inactivation, and decreased peak action potential (AP) upstroke velocity. All 3 AADs displayed enhanced sodium channel block at more positive atrial RMPs. Dronedarone was the most sensitive to changes in atrial RMP. Dronedarone caused greater reductions in AP amplitude and peak AP upstroke velocity at more positive RMPs. Dronedarone evoked greater prolongation of the atrial effective refractory period and postrepolarization refractoriness in murine Langendorff-perfused Pitx2c+/- hearts, which have a more positive RMP compared to wild type. CONCLUSION: Atrial RMP modifies the effectiveness of several clinically used AADs. Dronedarone is more sensitive to changes in atrial RMP than flecainide or propafenone. Identifying and modifying atrial RMP may offer a novel approach to enhancing the effectiveness of AADs or personalizing AAD selection.


Subject(s)
Atrial Fibrillation/metabolism , Dronedarone/therapeutic use , Flecainide/therapeutic use , Heart Atria/metabolism , Membrane Potentials/drug effects , Propafenone/therapeutic use , Sodium/metabolism , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Disease Models, Animal , Female , Heart Atria/physiopathology , Male , Mice , Voltage-Gated Sodium Channel Blockers/therapeutic use
17.
Naunyn Schmiedebergs Arch Pharmacol ; 394(7): 1579-1588, 2021 07.
Article in English | MEDLINE | ID: mdl-33738513

ABSTRACT

Aloperine (ALO), a quinolizidine alkaloid extracted from Sophora alopecuroides L., modulates hypertension, ventricular remodeling, and myocardial ischemia. However, few studies have evaluated the effects of ALO on other cardiovascular parameters. Accordingly, in this study, we used a rat model of aconitine-induced ventricular arrhythmia to assess the effects of ALO. Notably, ALO pretreatment delayed the onset of ventricular premature and ventricular tachycardia and reduced the incidence of fatal ventricular fibrillation. Moreover, whole-cell patch-clamp assays in rats' ventricular myocyte showed that ALO (3, 10, and 30 µM) significantly reduced the peak sodium current density of voltage-gated Na+ channel currents (INa) in a concentration-dependent manner. The gating kinetics characteristics showed that the steady-state activation and recovery curve were shifted in positive direction along the voltage axis, respectively, and the steady-state inactivation curve was shifted in negative direction along the voltage axis, i.e., which was similar to the inhibitory effects of amiodarone. These results indicated that ALO had anti-arrhythmic effects, partly attributed to INa inhibition. ALO may act as a class I sodium channel anti-arrhythmia agent.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Myocytes, Cardiac/drug effects , Quinolizidines/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/physiology , Animals , Animals, Newborn , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/physiopathology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Female , Heart Ventricles/cytology , Heart Ventricles/drug effects , Male , Myocytes, Cardiac/physiology , Quinolizidines/therapeutic use , Rats , Rats, Sprague-Dawley , Voltage-Gated Sodium Channel Blockers/therapeutic use
19.
Trials ; 22(1): 131, 2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33573681

ABSTRACT

OBJECTIVES: The main objective of this study is to evaluate the effect of intravenous lidocaine on gas exchange and inflammation in acute respiratory distress syndrome due or not to Covid-19 pneumonia. TRIAL DESIGN: This is a prospective monocentric, randomized, quadruple-blinded and placebo-controlled superiority trial. This phase 3 clinical study is based on two parallel groups received either intravenous lidocaine 2% or intravenous NaCl 0.9%. PARTICIPANTS: This study has been conducted at the University Hospitals of Strasbourg (medical and surgical Intensive Care Units in Hautepierre Hospital) since the 4th November 2020. The participants are 18 years-old and older, hospitalized in ICU for a moderate to severe ARDS according to the Berlin definition; they have to be intubated and sedated for mechanical protective ventilation. All participants are affiliated to the French Social security system and a dosage of beta HCG has to be negative for women of child bearing age . For the Covid-19 subgroup, the SARS-CoV2 infection is proved by RT-PCR <7 days before admission and/or another approved diagnostic technique and/or typical CT appearance pneumonia. The data are prospectively collected in e-Case Report Forms and extracted from clinical files. INTERVENTION AND COMPARATOR: The participants are randomised in two parallel groups with a 1:1 ratio. In the experimental group, patients receive intravenous lidocaine 2% (20mg/mL) (from FRESENIUS KABI France); the infusion protocol provide a bolus of 1 mg/kg (ideal weight), followed by 3 mg/kg/h for the first hour, 1.5 mg/kg/h for the second hour, 0.72 mg/kg/h for the next 22 hours and then 0.6 mg/kg/h for 14 days at most or 24 hours after extubation or ventilator-weaning. The patients in the control group receive intravenous NaCl 0.9% (9 mg/mL) (from Aguettant, France) as placebo comparator; the infusion protocol provide a bolus of 0.05 mL/kg (ideal weight), followed by 0.15 mL/kg/h for the first hour, 0.075 mL/kg/h for the second hour, 0.036 mL/kg/h for the next 22 hours, and the 0.03 mL/kg/h for up to 14 days or 24 hours after extubation or ventilator-weaning. Lidocaine level is assessed at H4, D2, D7 and D14 to prevent local anesthetics systemic toxicity. Clinical data and biological samples are collected to assess disease progression. MAIN OUTCOMES: The primary outcome is the evolution of alveolar-capillary gas exchange measured by the PaO2/FiO2 ratio after two days of treatment. The secondary endpoints of the study include the following: Evolution of PaO2/FiO2 ratio at admission and after 21 days of treatment Number of ventilator-free days Anti-inflammatory effects by dosing inflammatory markers at different timepoints (ferritin, bicarbonate, CRP, PCT, LDH, IL-6, Troponin HS, triglycerides, complete blood count, lymphocytes) Anti-thrombotic effects by dosing platelets, aPTT, fibrinogen, D-dimers, viscoelastic testing and identification of all thromboembolic events up to 4 weeks. Plasmatic concentration of lidocaine and albumin Incidence of adverse events like cardiac rhythm disorders, need of vasopressors, any modification of the QRS, QTc or PR intervals every day Ileus recovery time Consumption of hypnotics, opioids, neuromuscular blockers. Lengths of stay in the ICU, incidence of reintubation and complications due to intensive care unit care (mortality until 90 days, pneumothorax, bacterial pneumopathy, bronchospasm, cardiogenic shock, acute renal failure, need of renal dialysis, delirium, atrial fibrillation, stroke (CAM-ICU score), tetraplegia (MCR score)). Incidence of cough and sore throat at extubation or ventilator-weaning and within 24 hours. All these outcomes will be evaluated according to positivity to Sars-Cov-2. RANDOMISATION: The participants who meet the inclusion criteria and have signed written informed consent will be randomly allocated using a computer-generated random number to either intervention group or control group. The distribution ratio of the two groups will be 1:1, with a stratification according to positivity to Sars-Cov-2. BLINDING (MASKING): All participants, care providers, investigator and outcomes assessor are blinded. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): We planned to randomize fifty participants in each group, 100 participants total. TRIAL STATUS: The amended protocol version 2.1 was approved by the Ethics Committee "Comité de Protection des Personnes Sud-Méditerranée II on January 8, 2021 and by the Commission Nationale de l'Informatique et des Libertés (CNIL) on November 10, 2020. The study is currently recruiting participants; the recruitment started in November 2020 and the planned recruitment period is three years. TRIAL REGISTRATION: The trial was registered on clinicaltrials.gov on October 30, 2020 and identified by number NCT04609865 . FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Subject(s)
COVID-19 Drug Treatment , Lidocaine/therapeutic use , Respiratory Distress Syndrome/drug therapy , Voltage-Gated Sodium Channel Blockers/therapeutic use , Administration, Intravenous , COVID-19/blood , COVID-19/physiopathology , Clinical Trials, Phase III as Topic , Equivalence Trials as Topic , Humans , Inflammation/blood , Pulmonary Gas Exchange , Randomized Controlled Trials as Topic , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/physiopathology , SARS-CoV-2 , Treatment Outcome
20.
Muscle Nerve ; 63(3): 371-383, 2021 03.
Article in English | MEDLINE | ID: mdl-33340120

ABSTRACT

BACKGROUND: To collect preliminary data on the effects of mexiletine on cortical and axonal hyperexcitability in sporadic amyotrophic lateral sclerosis (ALS) in a phase 2 double-blind randomized controlled trial. METHODS: Twenty ALS subjects were randomized to placebo and mexiletine 300 or 600 mg daily for 4 wk and assessed by transcranial magnetic stimulation and axonal excitability studies. The primary endpoint was change in resting motor threshold (RMT). RESULTS: RMT was unchanged with 4 wk of mexiletine (combined active therapies) as compared to placebo, which showed a significant increase (P = .039). Reductions of motor evoked potential (MEP) amplitude (P = .013) and accommodation half-time (P = .002), secondary outcome measures of cortical and axonal excitability, respectively, were also evident at 4 wk on mexiletine. CONCLUSIONS: The relative stabilization of RMT in the treated subjects was unexpected and could be attributed to unaccounted sources of error or chance. However, a possible alternative cause is neuromodulation preventing an increase. The change in MEP amplitude and accommodation half-time supports the reduction of cortical and axonal hyperexcitability with mexiletine.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Axons , Cortical Excitability , Mexiletine/therapeutic use , Voltage-Gated Sodium Channel Blockers/therapeutic use , Adult , Aged , Amyotrophic Lateral Sclerosis/physiopathology , Double-Blind Method , Electrodiagnosis , Electromyography , Evoked Potentials, Motor/physiology , Female , Humans , Male , Median Nerve/physiopathology , Middle Aged , Neural Conduction/physiology , Preliminary Data , Transcranial Magnetic Stimulation
SELECTION OF CITATIONS
SEARCH DETAIL
...