Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Front Immunol ; 12: 696784, 2021.
Article in English | MEDLINE | ID: mdl-34276687

ABSTRACT

Atopic dermatitis (AD) is a multifaceted, chronic relapsing inflammatory skin disease that affects people of all ages. It is characterized by chronic eczema, constant pruritus, and severe discomfort. AD often progresses from mild annoyance to intractable pruritic inflammatory lesions associated with exacerbated skin sensitivity. The T helper-2 (Th2) response is mainly linked to the acute and subacute phase, whereas Th1 response has been associated in addition with the chronic phase. IL-17, IL-22, TSLP, and IL-31 also play a role in AD. Transient receptor potential (TRP) cation channels play a significant role in neuroinflammation, itch and pain, indicating neuroimmune circuits in AD. However, the Th2-driven cutaneous sensitization of TRP channels is underappreciated. Emerging findings suggest that critical Th2-related cytokines cause potentiation of TRP channels, thereby exaggerating inflammation and itch sensation. Evidence involves the following: (i) IL-13 enhances TRPV1 and TRPA1 transcription levels; (ii) IL-31 sensitizes TRPV1 via transcriptional and channel modulation, and indirectly modulates TRPV3 in keratinocytes; (iii) The Th2-cytokine TSLP increases TRPA1 synthesis in sensory neurons. These changes could be further enhanced by other Th2 cytokines, including IL-4, IL-25, and IL-33, which are inducers for IL-13, IL-31, or TSLP in skin. Taken together, this review highlights that Th2 cytokines potentiate TRP channels through diverse mechanisms under different inflammatory and pruritic conditions, and link this effect to distinct signaling cascades in AD. This review strengthens the notion that interrupting Th2-driven modulation of TRP channels will inhibit transition from acute to chronic AD, thereby aiding the development of effective therapeutics and treatment optimization.


Subject(s)
Cytokines/metabolism , Dermatitis, Atopic/metabolism , Inflammation Mediators/metabolism , Pruritus/metabolism , Skin/metabolism , Th2 Cells/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatitis, Atopic/immunology , Disease Progression , Humans , Membrane Transport Modulators/therapeutic use , Molecular Targeted Therapy , Pruritus/drug therapy , Pruritus/genetics , Pruritus/immunology , Signal Transduction , Skin/drug effects , Skin/immunology , Th2 Cells/immunology , Transcriptional Activation , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
2.
J Immunol Res ; 2021: 8832586, 2021.
Article in English | MEDLINE | ID: mdl-33928172

ABSTRACT

Ion transport modulators are most commonly used to treat various noncommunicable diseases including diabetes and hypertension. They are also known to bind to receptors on various immune cells, but the immunomodulatory properties of most ion transport modulators have not been fully elucidated. We assessed the effects of thirteen FDA-approved ion transport modulators, namely, ambroxol HCl, amiloride HCl, diazoxide, digoxin, furosemide, hydrochlorothiazide, metformin, omeprazole, pantoprazole, phenytoin, verapamil, drug X, and drug Y on superoxide production, nitric oxide production, and cytokine expression by THP-1-derived macrophages that had been stimulated with ethanol-inactivated Mycobacterium bovis BCG. Ambroxol HCl, diazoxide, digoxin, furosemide, hydrochlorothiazide, metformin, pantoprazole, phenytoin, verapamil, and drug Y had an inhibitory effect on nitric oxide production, while all the test drugs had an inhibitory effect on superoxide production. Amiloride HCl, diazoxide, digoxin, furosemide, phenytoin, verapamil, drug X, and drug Y enhanced the expression of IL-1ß and TNF-α. Unlike most immunomodulatory compounds currently in clinical use, most of the test drugs inhibited some inflammatory processes while promoting others. Ion pumps and ion channels could therefore serve as targets for more selective immunomodulatory agents which do not cause overt immunosuppression.


Subject(s)
Inflammation/drug therapy , Macrophages/immunology , Membrane Transport Modulators/therapeutic use , Mycobacterium bovis/immunology , Ambroxol/therapeutic use , Cells, Cultured , Humans , Immunomodulation , Interleukin-1beta/metabolism , Ion Transport , Macrophages/drug effects , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
3.
Am J Psychiatry ; 178(5): 437-446, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33653118

ABSTRACT

OBJECTIVE: Preclinical studies point to the KCNQ2/3 potassium channel as a novel target for the treatment of depression and anhedonia, a reduced ability to experience pleasure. The authors conducted the first randomized placebo-controlled trial testing the effect of the KCNQ2/3 positive modulator ezogabine on reward circuit activity and clinical outcomes in patients with depression. METHODS: Depressed individuals (N=45) with elevated levels of anhedonia were assigned to a 5-week treatment period with ezogabine (900 mg/day; N=21) or placebo (N=24). Participants underwent functional MRI during a reward flanker task at baseline and following treatment. Clinical measures of depression and anhedonia were collected at weekly visits. The primary endpoint was the change from baseline to week 5 in ventral striatum activation during reward anticipation. Secondary endpoints included depression and anhedonia severity as measured using the Montgomery-Åsberg Depression Rating Scale (MADRS) and the Snaith-Hamilton Pleasure Scale (SHAPS), respectively. RESULTS: The study did not meet its primary neuroimaging endpoint. Participants in the ezogabine group showed a numerical increase in ventral striatum response to reward anticipation following treatment compared with participants in the placebo group from baseline to week 5. Compared with placebo, ezogabine was associated with a significantly larger improvement in MADRS and SHAPS scores and other clinical endpoints. Ezogabine was well tolerated, and no serious adverse events occurred. CONCLUSIONS: The study did not meet its primary neuroimaging endpoint, although the effect of treatment was significant on several secondary clinical endpoints. In aggregate, the findings may suggest that future studies of the KCNQ2/3 channel as a novel treatment target for depression and anhedonia are warranted.


Subject(s)
Anhedonia , Carbamates/therapeutic use , Depressive Disorder, Major/drug therapy , KCNQ2 Potassium Channel , KCNQ3 Potassium Channel , Membrane Transport Modulators/therapeutic use , Phenylenediamines/therapeutic use , Reward , Ventral Striatum/diagnostic imaging , Adult , Depressive Disorder/diagnostic imaging , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/physiopathology , Double-Blind Method , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Ventral Striatum/physiopathology
4.
J Pharmacol Exp Ther ; 377(2): 293-304, 2021 05.
Article in English | MEDLINE | ID: mdl-33602875

ABSTRACT

In failing hearts, Na+/Ca2+ exchanger (NCX) overactivity contributes to Ca2+ depletion, leading to contractile dysfunction. Inhibition of NCX is expected to normalize Ca2+ mishandling, to limit afterdepolarization-related arrhythmias, and to improve cardiac function in heart failure (HF). SAR340835/SAR296968 is a selective NCX inhibitor for all NCX isoforms across species, including human, with no effect on the native voltage-dependent calcium and sodium currents in vitro. Additionally, it showed in vitro and in vivo antiarrhythmic properties in several models of early and delayed afterdepolarization-related arrhythmias. Its effect on cardiac function was studied under intravenous infusion at 250,750 or 1500 µg/kg per hour in dogs, which were either normal or submitted to chronic ventricular pacing at 240 bpm (HF dogs). HF dogs were infused with the reference inotrope dobutamine (10 µg/kg per minute, i.v.). In normal dogs, NCX inhibitor increased cardiac contractility (dP/dtmax) and stroke volume (SV) and tended to reduce heart rate (HR). In HF dogs, NCX inhibitor significantly and dose-dependently increased SV from the first dose (+28.5%, +48.8%, and +62% at 250, 750, and 1500 µg/kg per hour, respectively) while significantly increasing dP/dtmax only at 1500 (+33%). Furthermore, NCX inhibitor significantly restored sympathovagal balance and spontaneous baroreflex sensitivity (BRS) from the first dose and reduced HR at the highest dose. In HF dogs, dobutamine significantly increased dP/dtmax and SV (+68.8%) but did not change HR, sympathovagal balance, or BRS. Overall, SAR340835, a selective potent NCX inhibitor, displayed a unique therapeutic profile, combining antiarrhythmic properties, capacity to restore systolic function, sympathovagal balance, and BRS in HF dogs. NCX inhibitors may offer new therapeutic options for acute HF treatment. SIGNIFICANCE STATEMENT: HF is facing growing health and economic burden. Moreover, patients hospitalized for acute heart failure are at high risk of decompensation recurrence, and no current acute decompensated HF therapy definitively improved outcomes. A new potent, Na+/Ca2+ exchanger inhibitor SAR340835 with antiarrhythmic properties improved systolic function of failing hearts without creating hypotension, while reducing heart rate and restoring sympathovagal balance. SAR340835 may offer a unique and attractive pharmacological profile for patients with acute heart failure as compared with current inotrope, such as dobutamine.


Subject(s)
Heart Failure/drug therapy , Membrane Transport Modulators/therapeutic use , Sodium-Calcium Exchanger/antagonists & inhibitors , Vagus Nerve/drug effects , Animals , Baroreflex , Dogs , Heart/drug effects , Heart Rate , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/pharmacology , Myocardial Contraction , Myocardium/metabolism , Swine
5.
J Pharmacol Exp Ther ; 377(1): 20-28, 2021 04.
Article in English | MEDLINE | ID: mdl-33431609

ABSTRACT

Treatment of chronic pain remains an unmet medical need. The neuronal voltage-gated potassium Kv7/KCNQ/M channel has been implicated as a therapeutic target for chronic pain. However, whether pharmacological activation of the Kv7 channel can alleviate pain remains elusive. In this study, we show that selective activation of native M-currents by a novel channel opener SCR2682 reduces repetitive firings of dorsal root ganglia (DRG) sensory neurons. Intraperitoneal administration of SCR2682 relieves mechanical allodynia and thermal hyperalgesia in rat models of pain induced by complete Freund's adjuvant (CFA) or spared nerve injury (SNI) in a dose-dependent manner without affecting locomotor activity. The antinociceptive efficacy of SCR2682 can be reversed by the channel-specific blocker XE991. Furthermore, SCR2682 increases Kv7.2/KCNQ2 mRNA and protein expression in DRG neurons from rats in the SNI model of neuropathic pain. Taken together, pharmacological activation of neuronal Kv7 channels by opener SCR2682 can alleviate pain in rats, thus possessing therapeutic potential for chronic pain or hyperexcitability-related neurologic disorders. SIGNIFICANCE STATEMENT: A novel voltage-gated potassium Kv7 channel opener SCR2682 inhibits action potential firings in dorsal root ganglia sensory neurons and exhibits efficacy in antinociception, thus possessing a developmental potential for treatment of chronic pain or epilepsy.


Subject(s)
Analgesics/therapeutic use , Chronic Pain/drug therapy , KCNQ2 Potassium Channel/metabolism , Membrane Transport Modulators/therapeutic use , Pyridines/therapeutic use , Action Potentials , Analgesics/pharmacology , Animals , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiology , KCNQ2 Potassium Channel/agonists , Male , Membrane Transport Modulators/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
6.
J Pharmacol Exp Ther ; 377(1): 75-85, 2021 04.
Article in English | MEDLINE | ID: mdl-33504590

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory diseases of the gastrointestinal tract arising from abnormal responses of the innate and adaptative immune systems. Interleukin (IL)-10-producing CD4+CD25+ regulatory T (Treg) cells play a protective role in the recovery phase of IBD. In the present study, the effects of the administration of the selective Ca2+-activated K+ channel KCa3.1 inhibitor TRAM-34 on disease activities were examined in chemically induced IBD model mice. IBD disease severity, as assessed by diarrhea, visible fecal blood, inflammation, and crypt damage in the colon, was significantly lower in mice administered 1 mg/kg TRAM-34 than in vehicle-administered mice. Quantitative real-time polymerase chain reaction examinations showed that IL-10 expression levels in the recovery phase were markedly increased by the inhibition of KCa3.1 in mesenteric lymph node (mLN) Treg cells of IBD model mice compared with vehicle-administered mice. Among several positive and negative transcriptional regulators (TRs) for IL-10, three positive TRs-E4BP4, KLF4, and Blimp1-were upregulated by the inhibition of KCa3.1 in the mLN Treg cells of IBD model mice. In mouse peripheral CD4+CD25+ Treg cells induced by lectin stimulation, IL-10 expression and secretion were enhanced by the treatment with TRAM-34, together with the upregulation of E4BP4, KLF4, and Blimp1. Collectively, the present results demonstrated that the pharmacological inhibition of KCa3.1 decreased IBD symptoms in the IBD model by increasing IL-10 production in peripheral Treg cells and that IL-10high Treg cells produced by the treatment with KCa3.1 inhibitor may contribute to efficient Treg therapy for chronic inflammatory disorders, including IBD. SIGNIFICANCE STATEMENT: Pharmacological inhibition of Ca2+-activated K+ channel KCa3.1 increased IL-10 expression in peripheral Treg cells, together with the upregulation of the transcriptional regulators of IL-10: Krüppel-like factor 4, E4 promoter-binding protein 4, and/or B lymphocyte-induced maturation protein 1. The manipulation of IL-10high-producing Treg cells by the pharmacological inhibition of KCa3.1 may be beneficial in the treatment of chronic inflammatory diseases such as inflammatory bowel disease.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interleukin-10/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Membrane Transport Modulators/pharmacology , Pyrazoles/pharmacology , T-Lymphocytes, Regulatory/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Female , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , T-Lymphocytes, Regulatory/drug effects
7.
J Cardiovasc Pharmacol ; 77(4): 430-449, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33416260

ABSTRACT

ABSTRACT: Establishing efficient perfusion into the myocardium is the main purpose in patients with acute coronary syndrome, but the process of reperfusion is not without risk and can damage the myocardium paradoxically. Unfortunately, there is no effective treatment for reperfusion injury, and efforts to find an efficient preventive approach are still ongoing. In the past 3 decades, there have been many successful animal studies on how to prevent reperfusion injury; nonetheless, translation to the clinical setting has almost always proven disappointing. In this article, we review clinical studies on the prevention of reperfusion injury in patients with acute coronary syndrome undergoing primary percutaneous coronary intervention in a pharmacologic-based approach. We categorize all the agents that are evaluated for the prevention of myocardial reperfusion injury based on their mechanisms of action into 5 groups: drugs that can reduce oxidative stress, drugs that can affect cellular metabolism, rheological agents that target microvascular obstruction, anti-inflammatory agents, and agents with mixed mechanisms of action. Then, review all the clinical studies of these agents in the setting of primary percutaneous coronary intervention. Finally, we will discuss the possible reasons for the failure in translation of studies into practice and propose potential solutions to overcome this problem.


Subject(s)
Acute Coronary Syndrome/therapy , Cardiovascular Agents/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Percutaneous Coronary Intervention , Acute Coronary Syndrome/physiopathology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cardiovascular Agents/adverse effects , Humans , Membrane Transport Modulators/therapeutic use , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/physiopathology , Percutaneous Coronary Intervention/adverse effects , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
8.
FEBS J ; 288(12): 3784-3798, 2021 06.
Article in English | MEDLINE | ID: mdl-32654397

ABSTRACT

The pervasion of three daily meals and snacks is a relatively new introduction to our shared experience and is coincident with an epidemic rise in obesity and cardiometabolic disorders of overnutrition. The past two decades have yielded convincing evidence regarding the adaptive, protective effects of calorie restriction (CR) and intermittent fasting (IF) against cardiometabolic, neurodegenerative, proteostatic, and inflammatory diseases. Yet, durable adherence to intensive lifestyle changes is rarely attainable. New evidence now demonstrates that restricting carbohydrate entry into the hepatocyte by itself mimics several key signaling responses and physiological outcomes of IF and CR. This discovery raises the intriguing proposition that targeting hepatocyte carbohydrate transport to mimic fasting and caloric restriction can abate cardiometabolic and perhaps other fasting-treatable diseases. Here, we review the metabolic and signaling fates of a hepatocyte carbohydrate, identify evidence to target the key mediators within these pathways, and provide rationale and data to highlight carbohydrate transport as a broad, proximal intervention to block the deleterious sequelae of hepatic glucose and fructose metabolism.


Subject(s)
Hepatocytes/drug effects , Liver/drug effects , Membrane Transport Modulators/therapeutic use , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Overnutrition/prevention & control , Animals , Biological Transport/drug effects , Caloric Restriction/methods , Carbohydrate Metabolism/drug effects , Carbohydrate Metabolism/genetics , Disease Models, Animal , Fasting/metabolism , Gene Expression , Glucose/antagonists & inhibitors , Glucose/metabolism , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Molecular Targeted Therapy/methods , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Overnutrition/genetics , Overnutrition/metabolism , Overnutrition/pathology
9.
Pharmacol Res ; 159: 105026, 2020 09.
Article in English | MEDLINE | ID: mdl-32562815

ABSTRACT

Central nervous system (CNS) disorders like Alzheimer's disease (AD), Parkinson disease (PD), stroke, epilepsy, depression, and bipolar disorder have a high impact on both medical and social problems due to the surge in their prevalence. All of these neuronal disorders share some common etiologies including disruption of Ca2+ homeostasis and accumulation of misfolded proteins. These misfolded proteins further disrupt the intracellular Ca2+ homeostasis by disrupting the activity of several ion channels including transient receptor potential (TRP) channels. TRP channel families include non-selective Ca2+ permeable channels, which act as cellular sensors activated by various physio-chemical stimuli, exogenous, and endogenous ligands responsible for maintaining the intracellular Ca2+ homeostasis. TRP channels are abundantly expressed in the neuronal cells and disturbance in their activity leads to various neuronal diseases. Under the pathological conditions when the activity of TRP channels is perturbed, there is a disruption of the neuronal homeostasis through increased inflammatory response, generation of reactive oxygen species, and mitochondrial dysfunction. Therefore, there is a potential of pharmacological interventions targeting TRP channels in CNS disorders. This review focuses on the role of TRP channels in neurological diseases; also, we have highlighted the current insights into the pharmacological modulators targeting TRP channels.


Subject(s)
Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/drug therapy , Central Nervous System/drug effects , Membrane Transport Modulators/therapeutic use , Transient Receptor Potential Channels/antagonists & inhibitors , Animals , Calcium Signaling , Central Nervous System/metabolism , Central Nervous System/physiopathology , Central Nervous System Agents/adverse effects , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/physiopathology , Humans , Membrane Transport Modulators/adverse effects , Oxidative Stress , Protein Folding , Reactive Oxygen Species/metabolism , Transient Receptor Potential Channels/metabolism
10.
Eur Respir Rev ; 29(155)2020 Mar 31.
Article in English | MEDLINE | ID: mdl-32198216

ABSTRACT

Drug compounds that augment the production and activity of the cystic fibrosis (CF) transmembrane regulator (CFTR) have revolutionised CF care. Many adults and some children with CF suffer advanced and severe lung disease or await lung transplantation. While the hope is that these drug compounds will prevent lung damage when started early in life, there is an ongoing need to care for people with advanced lung disease. The focus of this review is the accumulating data from clinical trials and case series regarding the benefits of CFTR modulator therapy in people with advanced pulmonary disease. We address the impact of treatment with ivacaftor, lumacaftor/ivacaftor, tezacaftor/ivacaftor and elexacaftor/tezacaftor/ivacaftor on lung function, pulmonary exacerbations, nutrition and quality of life. Adverse events of the different CFTR modulators, as well as the potential for drug-drug interactions, are discussed.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/drug therapy , Lung/drug effects , Membrane Transport Modulators/therapeutic use , Respiratory System Agents/therapeutic use , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Progression , Drug Combinations , Drug Interactions , Humans , Lung/metabolism , Lung/physiopathology , Membrane Transport Modulators/adverse effects , Quality of Life , Recovery of Function , Respiratory System Agents/adverse effects , Treatment Outcome
11.
J Pharmacol Exp Ther ; 373(1): 72-80, 2020 04.
Article in English | MEDLINE | ID: mdl-31969383

ABSTRACT

Spinal cord injury (SCI) usually leads to acute neuronal death and delayed secondary degeneration, resulting in sensory dysfunction, paralysis, and chronic pain. Excessive excitation is one of the critical factors leading to secondary neural damage initiated by various insults. KCNQ/Kv7 channels are highly expressed in spinal neurons and axons and play an important role in controlling their excitability. Enhancing KCNQ channel activity by using its specific opener retigabine could thus be a plausible treatment strategy to reduce the pathology after SCI. We produced contusive SCI at T10 in adult male rats, which then received 10 consecutive days' treatment with retigabine or vehicle starting 3 hours or 3 days after contusion. Two different concentrations and two different delivery methods were applied. Delivery of retigabine via Alzet osmotic pumps, but not intraperitoneal injections 3 hours after contusion, promoted recovery of locomotor function. Remarkably, retigabine delivery in both methods significantly attenuated the development of mechanical stimuli-induced hyperreflexia and spontaneous pain; however, no significant difference in the thermal threshold was observed. Although retigabine delivered 3 days after contusion significantly attenuated the development of mechanical hypersensitivity and spontaneous pain, the locomotor function is not improved by the delayed treatments. Finally, we found that early application of retigabine attenuates the inflammatory activity in the spinal cord and increases the survival of white matter after SCI. Our results suggest that decreasing neuronal excitability by targeting KCNQ/Kv7 channels at acute stage aids the recovery of locomotor function and attenuates the development of neuropathic pain after SCI. SIGNIFICANCE STATEMENT: Several pharmacological interventions have been proposed for spinal cord injury (SCI) treatment, but none have been shown to be both effective and safe in clinical trials. Necrotic neuronal death and chronic pain are often the cost of pathological neural excitation after SCI. We show that early, brief application of retigabine could aid locomotor and sensory neurobehavioral recovery after SCI, supporting the use of this drug in the clinic to promote motor and sensory function in patients with SCI.


Subject(s)
KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/metabolism , Locomotion/physiology , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Animals , Carbamates/pharmacology , Carbamates/therapeutic use , Locomotion/drug effects , Male , Membrane Transport Modulators/pharmacology , Membrane Transport Modulators/therapeutic use , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Thoracic Vertebrae/injuries
12.
J Pediatr Health Care ; 33(3): 356-364, 2019.
Article in English | MEDLINE | ID: mdl-31029283

ABSTRACT

The literature surrounding the use of cystic fibrosis transmembrane conductance regulator-targeted pharmacotherapies in pediatric patients continues to evolve. These therapies represent a departure from symptom management and infection prevention, which have been the mainstay of cystic fibrosis management in pediatrics, to targeting the genetic defect present within these patients. This article reviews the clinical studies evaluating the safety and efficacy of ivacaftor, ivacaftor/lumacaftor, and ivacaftor/tezacaftor. These medications were initially studied in adults and adolescents but have begun to be studied in younger populations. Further investigation into the use of these drugs with different CFTR mutations and in younger age groups will continue to expand the number of patients who can benefit from these therapies.


Subject(s)
Aminophenols/therapeutic use , Aminopyridines/therapeutic use , Benzodioxoles/therapeutic use , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Indoles/therapeutic use , Membrane Transport Modulators/therapeutic use , Pediatrics , Quinolones/therapeutic use , Child , Cystic Fibrosis/genetics , Cystic Fibrosis/physiopathology , Drug Combinations , Humans , Mutation , Treatment Outcome
13.
Mol Autism ; 10: 15, 2019.
Article in English | MEDLINE | ID: mdl-30962870

ABSTRACT

Background: Mutations of the SCN2A gene encoding a voltage-gated sodium channel alpha-II subunit Nav1.2 are associated with neurological disorders such as epilepsy, autism spectrum disorders, intellectual disability, and schizophrenia. However, causal relationships and pathogenic mechanisms underlying these neurological defects, especially social and psychiatric features, remain to be elucidated. Methods: We investigated the behavior of mice with a conventional or conditional deletion of Scn2a in a comprehensive test battery including open field, elevated plus maze, light-dark box, three chambers, social dominance tube, resident-intruder, ultrasonic vocalization, and fear conditioning tests. We further monitored the effects of the positive allosteric modulator of AMPA receptors CX516 on these model mice. Results: Conventional heterozygous Scn2a knockout mice (Scn2aKO/+) displayed novelty-induced exploratory hyperactivity and increased rearing. The increased vertical activity was reproduced by heterozygous inactivation of Scn2a in dorsal-telencephalic excitatory neurons but not in inhibitory neurons. Moreover, these phenotypes were rescued by treating Scn2aKO/+ mice with CX516. Additionally, Scn2aKO/+ mice displayed mild social behavior impairment, enhanced fear conditioning, and deficient fear extinction. Neuronal activity was intensified in the medial prefrontal cortex of Scn2aKO/+ mice, with an increase in the gamma band. Conclusions: Scn2aKO/+ mice exhibit a spectrum of phenotypes commonly observed in models of schizophrenia and autism spectrum disorder. Treatment with the CX516 ampakine, which ameliorates hyperactivity in these mice, could be a potential therapeutic strategy to rescue some of the disease phenotypes.


Subject(s)
Anxiety/genetics , Autism Spectrum Disorder/genetics , Memory , NAV1.2 Voltage-Gated Sodium Channel/genetics , Psychomotor Agitation/genetics , Social Behavior , Animals , Anxiety/drug therapy , Autism Spectrum Disorder/drug therapy , Dioxoles/therapeutic use , Gamma Rhythm , Haploinsufficiency , Male , Membrane Transport Modulators/therapeutic use , Mice , Mice, Inbred C57BL , Phenotype , Piperidines/therapeutic use , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Psychomotor Agitation/drug therapy
14.
Exp Neurol ; 315: 52-59, 2019 05.
Article in English | MEDLINE | ID: mdl-30738808

ABSTRACT

Patients with myotonia congenita suffer from muscle stiffness caused by muscle hyperexcitability. Although loss-of-function mutations in the ClC-1 muscle chloride channel have been known for 25 years to cause myotonia congenita, this discovery has led to little progress on development of therapy. Currently, treatment is primarily focused on reducing hyperexcitability by blocking Na+ current. However, other approaches such as increasing K+ currents might also be effective. For example, the K+ channel activator retigabine, which opens KCNQ channels, is effective in treating epilepsy because it causes hyperpolarization of the resting membrane potential in neurons. In this study, we found that retigabine greatly reduced the duration of myotonia in vitro. Detailed study of its mechanism of action revealed that retigabine had no effect on any of the traditional measures of muscle excitability such as resting potential, input resistance or the properties of single action potentials. Instead it appears to shorten myotonia by activating K+ current during trains of action potentials. Retigabine also greatly reduced the severity of myotonia in vivo, which was measured using a muscle force transducer. Despite its efficacy in vivo, retigabine did not improve motor performance of mice with myotonia congenita. There are a number of potential explanations for the lack of motor improvement in vivo including central nervous system side effects. Nonetheless, the striking effectiveness of retigabine on muscle itself suggests that activating potassium currents is an effective method to treat disorders of muscle hyperexcitability.


Subject(s)
Carbamates/therapeutic use , Membrane Transport Modulators/therapeutic use , Myotonia Congenita/drug therapy , Phenylenediamines/therapeutic use , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Chloride Channels/genetics , Chloride Channels/metabolism , In Vitro Techniques , KCNQ Potassium Channels/drug effects , Membrane Potentials/drug effects , Mice , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Myotonia Congenita/psychology , Psychomotor Performance/drug effects
15.
Clin Pharmacol Ther ; 105(4): 943-953, 2019 04.
Article in English | MEDLINE | ID: mdl-30447156

ABSTRACT

Balanced multi-ion channel-blocking drugs have low torsade risk because they block inward currents. The Comprehensive In Vitro Proarrhythmia Assay (CiPA) initiative proposes to use an in silico cardiomyocyte model to determine the presence of balanced block, and absence of heart rate corrected J-Tpeak (J-Tpeak c) prolongation would be expected for balanced blockers. This study included three balanced blockers in a 10-subject-per-drug parallel design; lopinavir/ritonavir and verapamil met the primary end point of ΔΔJ-Tpeak c upper bound < 10 ms, whereas ranolazine did not (upper bounds of 8.8, 6.1, and 12.0 ms, respectively). Chloroquine, a predominant blocker of the potassium channel encoded by the ether-à-go-go related gene (hERG), prolonged ΔΔQTc and ΔΔJ-Tpeak c by ≥ 10 ms. In a separate crossover design, diltiazem (calcium block) did not shorten dofetilide-induced ΔQTc prolongation, but shortened ΔJ-Tpeak c and prolonged ΔTpeak -Tend . Absence of J-Tpeak c prolongation seems consistent with balanced block; however, small sample size (10 subjects) may be insufficient to characterize concentration-response in some cases.


Subject(s)
Biomarkers/metabolism , Electrocardiography/drug effects , Ion Channels/antagonists & inhibitors , Membrane Transport Modulators/therapeutic use , Pharmaceutical Preparations/administration & dosage , Adult , Cross-Over Studies , Female , Heart Rate/drug effects , Humans , Long QT Syndrome/drug therapy , Long QT Syndrome/metabolism , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Torsades de Pointes/drug therapy , Torsades de Pointes/metabolism
16.
Circulation ; 138(17): 1879-1896, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30354657

ABSTRACT

BACKGROUND: Among his major cardiac electrophysiological contributions, Miles Vaughan Williams (1918-2016) provided a classification of antiarrhythmic drugs that remains central to their clinical use. METHODS: We survey implications of subsequent discoveries concerning sarcolemmal, sarcoplasmic reticular, and cytosolic biomolecules, developing an expanded but pragmatic classification that encompasses approved and potential antiarrhythmic drugs on this centenary of his birth. RESULTS: We first consider the range of pharmacological targets, tracking these through to cellular electrophysiological effects. We retain the original Vaughan Williams Classes I through IV but subcategorize these divisions in light of more recent developments, including the existence of Na+ current components (for Class I), advances in autonomic (often G protein-mediated) signaling (for Class II), K+ channel subspecies (for Class III), and novel molecular targets related to Ca2+ homeostasis (for Class IV). We introduce new classes based on additional targets, including channels involved in automaticity, mechanically sensitive ion channels, connexins controlling electrotonic cell coupling, and molecules underlying longer-term signaling processes affecting structural remodeling. Inclusion of this widened range of targets and their physiological sequelae provides a framework for a modernized classification of established antiarrhythmic drugs based on their pharmacological targets. The revised classification allows for the existence of multiple drug targets/actions and for adverse, sometimes actually proarrhythmic, effects. The new scheme also aids classification of novel drugs under investigation. CONCLUSIONS: We emerge with a modernized classification preserving the simplicity of the original Vaughan Williams framework while aiding our understanding and clinical management of cardiac arrhythmic events and facilitating future developments in this area.


Subject(s)
Anti-Arrhythmia Agents/classification , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Heart Conduction System/drug effects , Heart Rate/drug effects , Membrane Transport Modulators/classification , Membrane Transport Modulators/therapeutic use , Terminology as Topic , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Calcium Channel Blockers/classification , Calcium Channel Blockers/therapeutic use , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Membrane Transport Modulators/adverse effects , Neurotransmitter Agents/classification , Neurotransmitter Agents/therapeutic use , Potassium Channel Blockers/classification , Potassium Channel Blockers/therapeutic use , Voltage-Gated Sodium Channel Blockers/classification , Voltage-Gated Sodium Channel Blockers/therapeutic use
17.
Pflugers Arch ; 470(11): 1583-1596, 2018 11.
Article in English | MEDLINE | ID: mdl-30194638

ABSTRACT

Transient receptor potential (TRP) channels comprise a family of cation channels implicated in a variety of cellular processes including light, mechanical or chemical stimuli, temperature, pH, or osmolarity. TRP channel proteins are a diverse family of proteins that are expressed in many tissues. We debated our recent knowledge about the expression, function, and regulation of TRP channels in the different parts of the male urogenital system in health and disease. Emerging evidence suggests that dysfunction of TRP channels significantly contributes to the pathophysiology of urogenital diseases. So far, there are many efforts underway to determine if these channels can be used as drug targets to reverse declines in male urogenital function. Furthermore, developing safe and efficacious TRP channel modulators is warranted for male urogenital disorders in a clinical setting.


Subject(s)
Erectile Dysfunction/metabolism , Membrane Transport Modulators/therapeutic use , TRPV Cation Channels/metabolism , Urinary Bladder, Overactive/metabolism , Animals , Erectile Dysfunction/drug therapy , Humans , Male , Membrane Transport Modulators/pharmacology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , Urinary Bladder, Overactive/drug therapy
18.
Mol Pain ; 14: 1744806918793229, 2018.
Article in English | MEDLINE | ID: mdl-30027794

ABSTRACT

Background Diabetic neuropathic pain is poorly controlled by analgesics, and the precise molecular mechanisms underlying hyperalgesia remain unclear. The KCNQ2/3/5 channels expressed in dorsal root ganglion neurons are important in pain transmission. The expression and activity of KCNQ2/3/5 channels in dorsal root ganglion neurons in rats with diabetic neuropathic pain were investigated in this study. Methods The mRNA levels of KCNQ2/3/5 channels were analyzed by real-time polymerase chain reaction. The protein levels of KCNQ2/3/5 channels were evaluated by Western blot assay. KCNQ2/3/5 channel expression in situ in dorsal root ganglion neurons was detected by double fluorescent labeling technique. M current (IM) density and neuronal excitability were determined by whole-cell voltage and current clamp recordings. Mechanical allodynia and thermal hyperalgesia were assessed by von Frey filaments and plantar analgesia tester, respectively. Results The mRNA and protein levels of KCNQ2/3/5 channels significantly decreased, followed by the reduction of IM density and elevation of neuronal excitability of dorsal root ganglion neurons from diabetic rats. Activation of KCNQ channels with retigabine reduced the hyperexcitability and inhibition of KCNQ channels with XE991 enhanced the hyperexcitability. Administration of retigabine alleviated both mechanical allodynia and thermal hyperalgesia, while XE991 augmented both mechanical allodynia and thermal hyperalgesia in diabetic neuropathic pain in rats. Conclusion The findings elucidate the mechanisms by which downregulation of the expression and reduction of the activity of KCNQ2/3/5 channels in diabetic rat dorsal root ganglion neurons contribute to neuronal hyperexcitability, which results in hyperalgesia. These data provide intriguing evidence that activation of KCNQ2/3/5 channels might be the potential new targets for alleviating diabetic neuropathic pain symptoms.


Subject(s)
Diabetic Neuropathies/pathology , Ganglia, Spinal/pathology , KCNQ Potassium Channels/metabolism , Neurons/metabolism , Animals , Anthracenes/pharmacology , Carbamates/pharmacology , Carbamates/therapeutic use , Cells, Cultured , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/drug therapy , Disease Models, Animal , Gene Expression Regulation/drug effects , KCNQ Potassium Channels/genetics , Membrane Transport Modulators/pharmacology , Membrane Transport Modulators/therapeutic use , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Pain Threshold/drug effects , Pain Threshold/physiology , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Potassium Channel Blockers/pharmacology , RNA, Messenger/metabolism , Rats , Streptozocin/toxicity , TRPV Cation Channels/metabolism
19.
Diabetes ; 67(9): 1847-1857, 2018 09.
Article in English | MEDLINE | ID: mdl-29959129

ABSTRACT

Previous studies by us and others have indicated that renal epidermal growth factor receptors (EGFR) are activated in models of diabetic nephropathy (DN) and that inhibition of EGFR activity protects against progressive DN in type 1 diabetes. In this study we examined whether inhibition of EGFR activation would affect the development of DN in a mouse model of accelerated type 2 diabetes (BKS db/db with endothelial nitric oxide knockout [eNOS-/-db/db]). eNOS-/-db/db mice received vehicle or erlotinib, an inhibitor of EGFR tyrosine kinase activity, beginning at 8 weeks of age and were sacrificed at 20 weeks of age. In addition, genetic models inhibiting EGFR activity (waved 2) and transforming growth factor-α (waved 1) were studied in this model of DN in type 2 diabetes. Compared with vehicle-treated mice, erlotinib-treated animals had less albuminuria and glomerulosclerosis, less podocyte loss, and smaller amounts of renal profibrotic and fibrotic components. Erlotinib treatment decreased renal oxidative stress, macrophage and T-lymphocyte infiltration, and the production of proinflammatory cytokines. Erlotinib treatment also preserved pancreas function, and these mice had higher blood insulin levels at 20 weeks, decreased basal blood glucose levels, increased glucose tolerance and insulin sensitivity, and increased blood levels of adiponectin compared with vehicle-treated mice. Similar to the aforementioned results, both waved 1 and waved 2 diabetic mice also had attenuated DN, preserved pancreas function, and decreased basal blood glucose levels. In this mouse model of accelerated DN, inhibition of EGFR signaling led to increased longevity.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/prevention & control , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/therapeutic use , Insulin Resistance , Membrane Transport Modulators/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Albuminuria/etiology , Albuminuria/prevention & control , Animals , Biomarkers/blood , Biomarkers/metabolism , Biomarkers/urine , Crosses, Genetic , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/immunology , Diabetic Nephropathies/physiopathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibrosis , Glomerulonephritis/immunology , Glomerulonephritis/physiopathology , Glomerulonephritis/prevention & control , Hypoglycemic Agents/therapeutic use , Kidney/drug effects , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Mice, Mutant Strains , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
20.
Eur Respir Rev ; 27(148)2018 Jun 30.
Article in English | MEDLINE | ID: mdl-29653946

ABSTRACT

In this article we analyse the current authorised treatments and trends in early drug development for cystic fibrosis (CF) in the European Union for the time period 2000-2016. The analysis indicates a significant improvement in the innovation and development of new potential medicines for CF, shifting from products that act on the symptoms of the disease towards new therapies targeting the cause of CF. However, within these new innovative medicines, results for CF transmembrane conductance regulator (CFTR) modulators indicate that one major challenge for turning a CF concept product into an actual medicine for the benefit of patients resides in the fact that, although pre-clinical models have shown good predictability for certain mutations, a good correlation to clinical end-points or biomarkers (e.g. forced expiratory volume in 1 s and sweat chloride) for all mutations has not yet been achieved. In this respect, the use of alternative end-points and innovative nonclinical models could be helpful for the understanding of those translational discrepancies. Collaborative endeavours to promote further research and development in these areas as well as early dialogue with the regulatory bodies available at the European competent authorities are recommended.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis/drug therapy , Drug Discovery/trends , Lung/drug effects , Membrane Transport Modulators/therapeutic use , Respiratory System Agents/therapeutic use , Translational Research, Biomedical/trends , Animals , Cystic Fibrosis/diagnosis , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drug Approval/legislation & jurisprudence , Drug Discovery/legislation & jurisprudence , Europe , Government Regulation , Humans , Lung/metabolism , Lung/physiopathology , Membrane Transport Modulators/adverse effects , Molecular Targeted Therapy , Policy Making , Respiratory System Agents/adverse effects , Translational Research, Biomedical/legislation & jurisprudence , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...