Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
J Feline Med Surg ; 26(5): 1098612X241240326, 2024 May.
Article in English | MEDLINE | ID: mdl-38770706

ABSTRACT

OBJECTIVES: The aim of the present study was to assess the effect of gabapentin on blood pressure (BP) in cats with and without chronic kidney disease (CKD). METHODS: A randomized, blinded, placebo-controlled crossover study was performed. A total of 29 cats were included: 13 cats with stable CKD (IRIS stage 2-4) and 16 apparently healthy cats (serum creatinine <1.6 mg/dl and urine specific gravity >1.035). The cats were evaluated twice, approximately 1 week apart, and BP (Doppler sphygmomanometry) was obtained 3 h after cats received either a single dose of gabapentin 10mg/kg PO or placebo. For each cat, BP readings were obtained at each visit using the same Doppler and sphygmomanometer unit, and the same cat holder and Doppler operator, in the same location. RESULTS: After administration of a single dose of gabapentin (10 mg/kg PO), BP was significantly lower (median 122 mmHg, range 82-170) than after administration of the placebo (median 150 mmHg, range 102-191; P = 0.001). In the CKD subgroup, BP was significantly lower after administration of gabapentin (median 129 mmHg, range 96-170) than after administration of the placebo (median 155 mmHg, range 102-191; P = 0.008). In the healthy cat subgroup, BP was significantly lower after administration of gabapentin (median 121 mmHg, range 82-139) than after administration of the placebo (median 137 mmHg, range 102-177; P = 0.002). The median change in BP was -12 mmHg (range -95 to 10) for healthy cats and -12 mmHg (range -43 to 21) for cats with CKD (no significant difference between subgroups). CONCLUSIONS AND RELEVANCE: Gabapentin may decrease arterial BP in cats with and without CKD and these findings should be taken into account when gabapentin is administered to patients in which measurement of BP is needed.


Subject(s)
Blood Pressure , Cat Diseases , Cross-Over Studies , Gabapentin , Renal Insufficiency, Chronic , Animals , Cats , Gabapentin/administration & dosage , Gabapentin/pharmacology , Gabapentin/therapeutic use , Cat Diseases/drug therapy , Renal Insufficiency, Chronic/veterinary , Renal Insufficiency, Chronic/drug therapy , Blood Pressure/drug effects , Male , Female
2.
Biomed Pharmacother ; 174: 116524, 2024 May.
Article in English | MEDLINE | ID: mdl-38574622

ABSTRACT

This study evaluates the antiallodynic and antihyperalgesic effects of LMH-2, a new haloperidol (HAL) analog that acts as sigma-1 receptor (σ1 R) antagonist, in diabetic mice using a model of neuropathic pain induced by chronic hyperglycemia. Additionally, we compared its effects with those of HAL. Hyperglycemia was induced in mice by nicotinamide-streptozotocin administration (NA-STZ, 50-130 mg/kg). Four weeks later, mechanical allodynia was assessed using the up-down method, and hyperalgesia was evoked with formalin 0.5%. We evaluated antiallodynic and antihyperalgesic effects of LMH-2 (5.6-56.2 mg/kg), HAL (0.018-0.18 mg/kg) and gabapentin (GBP, 5.6-56.2 mg/kg). The results showed that LMH-2 had a more significant antiallodynic effect compared to HAL and GBP (90.4±8.7 vs 75.1±3.1 and 41.9±2.3%, respectively; P<0.05), as well as an antihyperalgesic effect (96.3±1.2 vs 86.9±7.41 and 86.9±4.8%, respectively; P<0.05). Moreover, the antiallodynic and antihyperalgesic effect of both LMH-2 and HAL were completely abolished by PRE-084 (σ1 R agonist); and partially by pramipexole (a D2-like receptor agonist). Finally, the effect of all treatments on the rotarod test, barra, open field and exploratory behaviors showed that LMH-2 did not alter the animals' balance or the exploratory behavior, unlike as HAL or GBP. The molecular docking included indicate that LMH-2 has lower affinity to the D2R than HAL. These results provide evidence that LMH-2 exerts its antinociceptive effects as a σ1 R antagonist without the adverse effects induced by HAL or GBP. Consequently, LMH-2 can be considered a good and safe strategy for treating neuropathic pain caused by hyperglycemia in patients with diabetes.


Subject(s)
Analgesics , Diabetes Mellitus, Experimental , Haloperidol , Hyperalgesia , Neuralgia , Receptors, sigma , Sigma-1 Receptor , Animals , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/metabolism , Haloperidol/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Male , Mice , Analgesics/pharmacology , Neuralgia/drug therapy , Hyperalgesia/drug therapy , Diabetic Neuropathies/drug therapy , Molecular Docking Simulation , Streptozocin , Dose-Response Relationship, Drug , Gabapentin/pharmacology
3.
Biomed Pharmacother ; 174: 116472, 2024 May.
Article in English | MEDLINE | ID: mdl-38531121

ABSTRACT

The Voltage-Gated Calcium Channel (VGCC) auxiliary subunit Cavα2δ-1 (CACNA2D1) is the target/receptor of gabapentinoids which are known therapeutics in epilepsy and neuropathic pain. Following damage to the peripheral sensory nervous system, Cavα2δ-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of chronic neuropathic pain. Gabapentinoids, such as gabapentin and pregabalin, engage with Cavα2δ-1 via binding an arginine residue (R241) within an RRR motif located at the N-terminus of human Cavα2δ-1. A novel, next generation gabapentinoid, engineered not to penetrate the brain, was able to generate a strong analgesic response in Chronic Constriction Injury animal model of chronic neuropathic pain and showed binding specificity for Cavα2δ-1 versus the Cavα2δ-2 subunit. This novel non-brain penetrant gabapentinoid, binds to R241 and a novel binding site on Cavα2δ-1, which is located within the VGCC_α2 domain, identified as a lysine residue within an IKAK amino acid motif (K634). The overall whole cell current amplitudes were diminished by the compound, with these inhibitory effects being diminished in R241A mutant Cavα2δ-1 subunits. The functional effects occurred at lower concentrations than those needed for inhibition by gabapentin or pregabalin, which apparently bound the Cavα2δ-1 subunit only on the R241 and not on the K634 residue. Our work sets the stage for the identification and characterisation of novel compounds with therapeutic properties in neuropathic pain and possibly in other disorders and conditions which require engagement of the Cavα2δ-1 target.


Subject(s)
Calcium Channels, L-Type , Neuralgia , Neuralgia/drug therapy , Neuralgia/metabolism , Animals , Ligands , Humans , Male , Calcium Channels/metabolism , Calcium Channels/genetics , Gabapentin/pharmacology , Rats, Sprague-Dawley , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Rats , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/genetics , Analgesics/pharmacology , Disease Models, Animal , Pregabalin/pharmacology
4.
Neuropharmacology ; 250: 109892, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38428481

ABSTRACT

KCNQ5 encodes the voltage-gated potassium channel KV7.5, a member of the KV7 channel family, which conducts the M-current. This current is a potent regulator of neuronal excitability by regulating membrane potential in the subthreshold range of action potentials and mediating the medium and slow afterhyperpolarization. Recently, we have identified five loss-of-function variants in KCNQ5 in patients with genetic generalized epilepsy. Using the most severe dominant-negative variant (R359C), we set out to investigate pharmacological therapeutic intervention by KV7 channel openers on channel function and neuronal firing. Retigabine and gabapentin increased R359C-derived M-current amplitudes in HEK cells expressing homomeric or heteromeric mutant KV7.5 channels. Retigabine was most effective in restoring K+ currents. Ten µM retigabine was sufficient to reach the level of WT currents without retigabine, whereas 100 µM of gabapentin showed less than half of this effect and application of 50 µM ZnCl2 only significantly increased M-current amplitude in heteromeric channels. Overexpression of KV7.5-WT potently inhibited neuronal firing by increasing the M-current, whereas R359C overexpression had the opposite effect and additionally decreased the medium afterhyperpolarization current. Both aforementioned drugs and Zn2+ reversed the effect of R359C expression by reducing firing to nearly normal levels at high current injections. Our study shows that a dominant-negative variant with a complete loss-of-function in KV7.5 leads to largely increased neuronal firing which may explain a neuronal hyperexcitability in patients. KV7 channel openers, such as retigabine or gabapentin, could be treatment options for patients currently displaying pharmacoresistant epilepsy and carrying loss-of-function variants in KCNQ5.


Subject(s)
Epilepsy , KCNQ2 Potassium Channel , Phenylenediamines , Humans , Gabapentin/pharmacology , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/metabolism , Epilepsy/drug therapy , Epilepsy/genetics , Carbamates/pharmacology , Carbamates/therapeutic use
5.
Eur J Pharmacol ; 967: 176398, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38350591

ABSTRACT

OBJECTIVES: Inflammation regulates ventricular remodeling after myocardial infarction (MI), and gabapentin exerts anti-inflammatory effects. We investigated the anti-inflammatory role and mechanism of gabapentin after MI. METHODS: Rats were divided into the sham group (n = 12), MI group (n = 20), and MI + gabapentin group (n = 16). MI was induced by left coronary artery ligation. The effects of gabapentin on THP-1-derived macrophages were examined in vitro. RESULTS: In vivo, 1 week after MI, gabapentin significantly reduced inducible nitric oxide synthase (iNOS; M1 macrophage marker) expression and decreased pro-inflammatory factors (tumor necrosis factor [TNF]-α and interleukin [IL]-1ß). Gabapentin upregulated the M2 macrophage marker arginase-1, as well as CD163 expression, and increased the expression of anti-inflammatory factors, including chitinase-like 3, IL-10, and transforming growth factor-ß. Four weeks after MI, cardiac function, infarct size, and cardiac fibrosis improved after gabapentin treatment. Gabapentin inhibited sympathetic nerve activity and decreased ventricular electrical instability in rats after MI. Tyrosine hydroxylase and growth-associated protein 43 were suppressed after gabapentin treatment. Gabapentin downregulated nerve growth factor (NGF) and reduced pro-inflammatory factors (iNOS, TNF-α, and IL-1ß). In vitro, gabapentin reduced NGF, iNOS, TNF-α, and IL-1ß expression in lipopolysaccharide-stimulated macrophages. Mechanistic studies revealed that the peroxisome proliferator-activated receptor-γ antagonist GW9662 attenuated the effects of gabapentin. Moreover, gabapentin reduced α2δ1 expression in the macrophage plasma membrane and reduced the calcium content of macrophages. CONCLUSION: Gabapentin attenuates cardiac remodeling by inhibiting inflammation via peroxisome proliferator-activated receptor-γ activation and preventing calcium overload.


Subject(s)
Myocardial Infarction , Tumor Necrosis Factor-alpha , Rats , Animals , Gabapentin/pharmacology , Gabapentin/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , PPAR gamma/metabolism , Ventricular Remodeling , Nerve Growth Factor/metabolism , Calcium/metabolism , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Macrophages , Anti-Inflammatory Agents/pharmacology , Inflammation/metabolism
6.
Pain ; 165(6): 1391-1403, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38227559

ABSTRACT

ABSTRACT: Neuropathic corneal pain (NCP) is a new and ill-defined disease characterized by pain, discomfort, aching, burning sensation, irritation, dryness, and grittiness. However, the mechanism underlying NCP remain unclear. Here, we reported a novel rat model of primary NCP induced by long ciliary nerve (LCN) ligation. After sustained LCN ligation, the rats developed increased corneal mechanical and chemical sensitivity, spontaneous blinking, and photophobia, which were ameliorated by intraperitoneal injection of morphine or gabapentin. However, neither tear reduction nor corneal injury was observed in LCN-ligated rats. Furthermore, after LCN ligation, the rats displayed a significant reduction in corneal nerve density, as well as increased tortuosity and beading nerve ending. Long ciliary nerve ligation also notably elevated corneal responsiveness under resting or menthol-stimulated conditions. At a cellular level, we observed that LCN ligation increased calcitonin gene-related peptide (neuropeptide)-positive cells in the trigeminal ganglion (TG). At a molecular level, upregulated mRNA levels of ion channels Piezo2, TRPM8, and TRPV1, as well as inflammatory factors TNF-α, IL-1ß, and IL-6, were also detected in the TG after LCN ligation. Meanwhile, consecutive oral gabapentin attenuated LCN ligation-induced corneal hyperalgesia and increased levels of ion channels and inflammation factors in TG. This study provides a reliable primary NCP model induced by LCN ligation in rats using a simple, minimally invasive surgery technique, which may help shed light on the underlying cellular and molecular bases of NCP and aid in developing a new treatment for the disease.


Subject(s)
Cornea , Disease Models, Animal , Gabapentin , Neuralgia , Rats, Sprague-Dawley , Animals , Neuralgia/etiology , Male , Rats , Gabapentin/pharmacology , Gabapentin/therapeutic use , Ligation , Cornea/innervation , Trigeminal Ganglion/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , gamma-Aminobutyric Acid/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Calcitonin Gene-Related Peptide/metabolism , Amines/pharmacology , Amines/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Eye Pain/etiology , Hyperalgesia/etiology , Hyperalgesia/physiopathology
7.
JCO Oncol Pract ; 20(2): 268-277, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38061003

ABSTRACT

PURPOSE: Opioid prescribing trends in medical oncology are poorly defined past 2017, the year after the CDC updated opioid prescription guidelines in noncancer settings. We aim to characterize pain management by medical oncologists by analyzing opioid and gabapentin prescribing trends from 2013 to 2019, identify physician-related factors associated with prescribing patterns, and assess whether CDC guidelines for nononcologic settings changed prescribing patterns. METHODS: The Centers for Medicare & Medicaid Services (CMS) Medicare Part D Prescribers-by Provider, CMS Medicare Part D Prescribers-by Provider and Drug, and CMS Medicare Physician National Downloadable files from 2013 to 2019 were merged by National Provider Identification. The database included physicians' sex, years of practice, regions, and practice settings. Multivariable binary logistic regression identified significant predictors of total opioid, long-acting opioid, and gabapentin prescriptions. RESULTS: Binary logistic regression modeling revealed no significant difference in mean daily total opioid prescriptions from 2013 to 2017. Daily opioid prescriptions by medical oncologists decreased significantly after 2017 (P < .001). Increased opioid prescribing was associated with physician male sex (P < .001), practicing over 10 years (P < .001), and practice in nonurban areas (P < .001). Opioid prescribing was greatest in the South and Midwest United States (P < .001). The same patterns were observed with total long-acting opioid prescriptions, whereas gabapentin prescribing increased from 2013 to 2019 (P < .001). CONCLUSION: Opioid prescriptions by medical oncologists decreased significantly from 2013 to 2019, but this decrease was most substantial from 2017 to 2019. These results may imply that the 2016 CDC guidelines influenced medical oncologists, particularly more junior physicians in urban settings, to manage chronic cancer pain with alternative therapies.


Subject(s)
Medicare Part D , Oncologists , Aged , Male , Humans , United States , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Medicaid , Gabapentin/pharmacology , Gabapentin/therapeutic use , Practice Patterns, Physicians'
8.
Best Pract Res Clin Endocrinol Metab ; 38(1): 101819, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37659918

ABSTRACT

Women are living a significant portion of their adult lives in the post-reproductive phase, and many seek help for debilitating menopausal symptoms. Every individual's experience of menopausal transition is unique. Adopting a holistic approach to managing the menopause using a combination of lifestyle, hormonal, and non-hormonal interventions is key to maximise the quality of life of affected women. However, many opt to use non hormonal options or have contraindications to using hormonal therapy. Studies have shown that several pharmacological non-hormonal medications such as SSRIs, SSRI/SNRIs, Gabapentin, and Pregabalin are effective for managing vasomotor symptoms as well as other menopausal symptoms. Their main side effects are dry mouth, nausea, constipation, reduced libido, and loss of appetite. Clonidine is the only non-hormonal drug which is licenced for control of vasomotor symptoms in the UK, but has several side effects including dizziness and sleep disturbance. Cognitive Behavioural Therapy is recommended as a treatment for anxiety, sleep problems and vasomotor symptoms related to menopausal transition. Evidence for clinical efficacy and safety of herbal remedies and alternative therapies remains weak. Studies with neurokinin receptor 3 antagonists on women with hot flushes have shown improvement in vasomotor symptoms and results of large-scale studies are awaited.


Subject(s)
Complementary Therapies , Quality of Life , Adult , Female , Humans , Menopause , Gabapentin/therapeutic use , Gabapentin/pharmacology , Hot Flashes/drug therapy
9.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 947-958, 2024 02.
Article in English | MEDLINE | ID: mdl-37548662

ABSTRACT

PURPOSE: Globally, sepsis, which is a major health issue resulting from severe infection-induced inflammation, is the fifth biggest cause of death. This research aimed to evaluate, for the first time, the molecular effects of gabapentin's possible nephroprotective potential on septic rats by cecal ligation and puncture (CLP). METHODS: Sepsis was produced by CLP in male Wistar rats. Evaluations of histopathology and renal function were conducted. MDA, SOD, GSH, TNF-α, IL-1ß, and IL-6 levels were measured. qRT-PCR was utilized to determine the expression of Bax, Bcl-2, and NF-kB genes. The expression of Nrf-2 and HO-1 proteins was examined by western blotting. RESULTS: CLP caused acute renal damage, elevated the blood levels of creatinine, BUN, TNF-α, IL-1ß, and IL-6, reduced the expression of Nrf-2 and HO-1 proteins and the Bcl-2 gene expression, and upregulated NF-kB and Bax genes. Nevertheless, gabapentin dramatically diminished the degree of the biochemical, molecular, and histopathological alterations generated by CLP. Gabapentin reduced the levels of proinflammatory mediators and MDA, improved renal content of GSH and SOD, raised the expression of Nrf-2 and HO-1 proteins and Bcl-2 gene, and reduced the renal expression of NF-kB and Bax genes. CONCLUSION: Gabapentin mitigated the CLP-induced sepsis-related acute kidney injury through up-regulating Nrf-2/HO-1 pathway, repressing apoptosis, and attenuating the oxidative stress status by reducing the levels of the proinflammatory mediators and enhancing the antioxidant status.


Subject(s)
Acute Kidney Injury , Sepsis , Rats , Male , Animals , NF-kappa B/metabolism , Gabapentin/pharmacology , Tumor Necrosis Factor-alpha/metabolism , bcl-2-Associated X Protein/metabolism , Interleukin-6/metabolism , Rats, Wistar , Signal Transduction , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Oxidative Stress , Sepsis/complications , Sepsis/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis , Superoxide Dismutase/metabolism
10.
J Am Vet Med Assoc ; 262(3): 359-363, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38134455

ABSTRACT

OBJECTIVE: To compare the effects of oral pregabalin versus gabapentin on sedation quality and anesthesia recovery times in cats in a typical perioperative setting. ANIMALS: 50 healthy cats with > 1 kg body weight presenting for elective surgery. METHODS: In this randomized, prospective clinical trial, cats presenting to the University of California-Davis Veterinary Medical Teaching Hospital were assigned to receive buprenorphine 0.02 mg/kg IM followed by 1 of 2 oral sedation treatments: pregabalin 4 mg/kg or gabapentin 10 mg/kg. Cats were then anesthetized using a standardized protocol. Physical examination parameters and behavioral scores were measured by 2 treatment-blinded veterinarians to compare sedation levels before and after drug administration. Inadequate sedation for handling or IV catheter placement was addressed by dexmedetomidine administration. After surgery was completed, anesthesia recovery times and quality were assessed by the same veterinarians. The effects of pregabalin versus gabapentin on body temperature, respiratory rate, and heart rate were analyzed using Student t tests; behavioral assessments were analyzed using Wilcoxon signed-rank tests; and drug treatment effects on dexmedetomidine sedation rescue and frequency of delirium during anesthetic recovery were analyzed using Fisher exact tests. A P < .05 indicated statistical significance. RESULTS: There was no significant difference in change of physiologic parameters or sedation scores before and after sedation between groups. The need for rescue sedation for IV catheter placement and the incidence of emergence delirium were infrequent and similar for both treatments. CLINICAL RELEVANCE: At the doses studied, oral pregabalin and gabapentin produced indistinguishable effects as adjunctive perioperative sedation agents in cats.


Subject(s)
Anesthesia , Dexmedetomidine , Cats , Animals , Gabapentin/pharmacology , Pregabalin/pharmacology , Pregabalin/therapeutic use , Dexmedetomidine/pharmacology , Prospective Studies , Anesthesia/veterinary , Heart Rate
11.
Cells ; 12(23)2023 11 25.
Article in English | MEDLINE | ID: mdl-38067133

ABSTRACT

Gabapentin (GBP), a GABA analogue, is primarily used as an anticonvulsant for the treatment of partial seizures and neuropathic pain. Whereas a majority of the side effects are associated with the nervous system, emerging evidence suggests there is a high risk of heart diseases in patients taking GBP. In the present study, we first used a preclinical model of rats to investigate, firstly, the acute cardiovascular responses to GBP (bolus i.v. injection, 50 mg/kg) and secondly the effects of chronic GBP treatment (i.p. 100 mg/kg/day × 7 days) on cardiovascular function and the myocardial proteome. Under isoflurane anesthesia, rat blood pressure (BP), heart rate (HR), and left ventricular (LV) hemodynamics were measured using Millar pressure transducers. The LV myocardium and brain cortex were analyzed by proteomics, bioinformatics, and western blot to explore the molecular mechanisms underlying GBP-induced cardiac dysfunction. In the first experiment, we found that i.v. GBP significantly decreased BP, HR, maximal LV pressure, and maximal and minimal dP/dt, whereas it increased IRP-AdP/dt, Tau, systolic, diastolic, and cycle durations (* p < 0.05 and ** p < 0.01 vs. baseline; n = 4). In the second experiment, we found that chronic GBP treatment resulted in hypotension, bradycardia, and LV systolic dysfunction, with no change in plasma norepinephrine. In the myocardium, we identified 109 differentially expressed proteins involved in calcium pathways, cholesterol metabolism, and galactose metabolism. Notably, we found that calmodulin, a key protein of intracellular calcium signaling, was significantly upregulated by GBP in the heart but not in the brain. In summary, we found that acute and chronic GBP treatments suppressed cardiovascular function in rats, which is attributed to abnormal calcium signaling in cardiomyocytes. These data reveal a novel side effect of GBP independent of the nervous system, providing important translational evidence to suggest that GBP can evoke adverse cardiovascular events by depression of myocardial function.


Subject(s)
Anticonvulsants , Heart , Humans , Rats , Animals , Gabapentin/pharmacology , Anticonvulsants/pharmacology , Hemodynamics , Blood Pressure
12.
ACS Chem Neurosci ; 14(23): 4208-4215, 2023 12 06.
Article in English | MEDLINE | ID: mdl-37947793

ABSTRACT

Gabapentin, a selective ligand for the α2δ subunit of voltage-dependent calcium channels, is an anticonvulsant medication used in the treatment of neuropathic pain, epilepsy, and other neurological conditions. We recently described two radiofluorinated derivatives of gabapentin (trans-4-[18F]fluorogabapentin, [18F]tGBP4F, and cis-4-[18F]fluorogabapentin, [18F]cGBP4F) and showed that these compounds accumulate in the injured nerves in a rodent model of neuropathic pain. Given the use of gabapentin in brain diseases, here we investigate whether these radiofluorinated derivatives of gabapentin can be used for imaging α2δ receptors in the brain. Specifically, we developed automated radiosynthesis methods for [18F]tGBP4F and [18F]cGBP4F and conducted dynamic PET imaging in adult rhesus macaques with and without preadministration of pharmacological doses of gabapentin. Both radiotracers showed very high metabolic stability, negligible plasma protein binding, and slow accumulation in the brain. [18F]tGBP4F, the isomer with higher binding affinity, showed low brain uptake and could not be displaced, whereas [18F]cGBP4F showed moderate brain uptake and could be partially displaced. Kinetic modeling of brain regional time-activity curves using a metabolite-corrected arterial input function shows that a one-tissue compartment model accurately fits the data. Graphical analysis using Logan or multilinear analysis 1 produced similar results as compartmental modeling, indicating robust quantification. This study advances our understanding of how gabapentinoids work and provides an important advancement toward imaging α2δ receptors in the brain.


Subject(s)
Neuralgia , Positron-Emission Tomography , Animals , Gabapentin/pharmacology , Gabapentin/metabolism , Macaca mulatta , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Brain/metabolism , Neuralgia/metabolism
13.
Eur J Pharmacol ; 959: 176078, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37805133

ABSTRACT

OBJECTIVE: The anti-tussive effect of gabapentin and its underlying neuromodulatory mechanism were investigated via a modified guinea pig model of gastroesophageal reflux-related cough (GERC). METHODS: Intra-esophageal perfusion with hydrochloric acid (HCl) was performed every other day 12 times to establish the GERC model. High-dose gabapentin (48 mg/kg), low-dose gabapentin (8 mg/kg), or saline was orally administered for 2 weeks after modeling. Cough sensitivity, airway inflammation, lung and esophagus histology, levels of substance P (SP), and neurokinin-1 (NK1)-receptors were monitored. RESULTS: Repeated intra-esophageal acid perfusion aggravated the cough sensitivity in guinea pigs in a time-dependent manner. The number of cough events was significantly increased after 12 times HCl perfusion, and the hypersensitivity period was maintained for 2 weeks. The SP levels in BALF, trachea, lung, distal esophagus, and vagal ganglia were increased in guinea pigs receiving HCl perfusion. The intensity of cough hypersensitivity in the GERC model was significantly correlated with increased SP expression in the airways. Both high and low doses of gabapentin administration could reduce cough hypersensitivity exposed to HCl perfusion, attenuate airway inflammatory damage, and inhibit neurogenic inflammation by reducing SP expression from the airway and vagal ganglia. CONCLUSIONS: Gabapentin can desensitize the cough sensitivity in the GERC model of guinea pig. The anti-tussive effect is associated with the alleviated peripheral neurogenic inflammation as reflected in the decreased level of SP.


Subject(s)
Cough , Gastroesophageal Reflux , Guinea Pigs , Animals , Cough/drug therapy , Cough/metabolism , Neurogenic Inflammation/complications , Neurogenic Inflammation/metabolism , Gabapentin/pharmacology , Lung/metabolism , Gastroesophageal Reflux/metabolism , Hydrochloric Acid/metabolism , Substance P/metabolism , Receptors, Neurokinin-1/metabolism , Perfusion
14.
Vet Rec ; 193(11): e3558, 2023 Dec 02.
Article in English | MEDLINE | ID: mdl-37886849

ABSTRACT

BACKGROUND: This study aimed to investigate the effects of gabapentin on various ocular parameters in New Zealand White rabbits. METHODS: A randomised, placebo-controlled crossover study design was employed. Eight New Zealand White rabbits were randomly assigned to receive either oral gabapentin at a dosage of 15 mg/kg or an oral placebo, with a 1-week washout period between treatments. Intraocular pressure, tear production and horizontal pupil diameter were measured at baseline (T0) and at 30, 60, 90, 120, 180, 240 and 360 minutes after drug administration. Physiological and behavioural changes were also recorded for both treatments following drug administration. RESULTS: The administration of gabapentin did not have any significant effects on the ocular parameters measured in this study. However, the rabbits exhibited some muscle relaxation with partially closed eyes during handling, and they were slightly easier to remove from the cage when treated with gabapentin compared to the placebo treatment. LIMITATIONS: In this study, the ocular effects of gabapentin were assessed in only a small number of healthy rabbits. These effects may differ in rabbits with pre-existing eye conditions or in those receiving other medications. CONCLUSIONS: Our findings suggest that gabapentin treatment does not have a significant impact on intraocular pressure, tear production or horizontal pupil diameter in rabbits.


Subject(s)
Eye Diseases , Intraocular Pressure , Rabbits , Animals , Pupil , Gabapentin/pharmacology , Cross-Over Studies , Tonometry, Ocular , Eye Diseases/veterinary
15.
J Am Vet Med Assoc ; 261(11): 1660-1665, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37495226

ABSTRACT

OBJECTIVE: To evaluate sedative and behavioral effects of a client-administered preappointment protocol with PO gabapentin and melatonin and oral-transmucosal acepromazine (GMA protocol). ANIMALS: 45 client-owned dogs between 1 and 12 years old that underwent standardize examinations between February and August 2021. METHODS: In this clinical trial, dogs with a history of anxiety, fearfulness, and/or aggression during hospital visits were assessed and videotaped before (baseline) and after administration of the GMA protocol. For the second visit, owners administered PO gabapentin (20 to 25 mg/kg) in the evening prior to the next visit and PO gabapentin (20 to 25 mg/kg), PO melatonin (3 to 5 mg/dog), and oral-transmucosal acepromazine (0.05 mg/kg) 90 to 120 minutes prior to the second appointment. Examinations were performed, and behavioral stress and sedation levels were evaluated with semiquantitative rating scales. Randomized videos were analyzed, and a paired t test was used to compare stress and sedation scores between baseline and GMA. A Pearson correlation coefficient was used to evaluate the effect of age on the scores. RESULTS: Stress scores were significantly lower after the GMA protocol, and sedation scores were significantly higher when compared to baseline (21.84 vs 27.11 and 1.39 vs 0.68, respectively). A significant correlation between increasing age and lower stress scores post-GMA and higher sedation scores post-GMA were observed. CLINICAL RELEVANCE: Preappointment administration of the GMA protocol reduced signs of stress, fear, and fear-based aggression during hospital visits and provided sedation in this dog population. This protocol could represent an adjunct tool for veterinarians to improve quality of care and reduce animal-related injury.


Subject(s)
Acepromazine , Melatonin , Humans , Dogs , Animals , Gabapentin/pharmacology , Gabapentin/therapeutic use , Acepromazine/pharmacology , Acepromazine/therapeutic use , Melatonin/pharmacology , Melatonin/therapeutic use , Aggression , Prospective Studies , Hypnotics and Sedatives/pharmacology , Hypnotics and Sedatives/therapeutic use , Anxiety , Hospitals
16.
Drug Des Devel Ther ; 17: 1793-1803, 2023.
Article in English | MEDLINE | ID: mdl-37346999

ABSTRACT

Background and Objective: Gabapentin is a commonly prescribed antiepileptic agent for seizures, which is also used for pain and addiction management. Due to growing evidence of its abuse liability, there has been an incentive to synthesise potentially useful gabapentin derivatives devoid of adverse effects. A gabapentin adduct with a fluoxetine moiety, GBP1F, was assessed for any sedative, cognitive, anxiolytic, or antidepressant-like actions in murine behavioral models. Materials and Methods: Selected groups of mice were used for each behavioral paradigm, and the effect of GBP1F (5, 10, and 15 mg/kg) was assessed using spontaneous locomotor activity, the tail suspension test, elevated plus maze test, and the Y maze test models. Immediately following behavioral experiments, postmortem striatal and hippocampal tissues were evaluated for the effect of GBP1F on concentrations of dopamine, DOPAC, HVA, serotonin, 5-HIAA, vitamin C, and noradrenaline using high performance liquid chromatography with electrochemical detection. Results: GBP1F induced a mild suppression of locomotor activity, ameliorated anxiety and depression-like behavior, did not alter cognitive behavior, and raised serotonin and 5-HIAA concentrations in the hippocampus and striatum. GBP1F also positively enhanced dopamine and vitamin C tissue levels in the striatum. Thus, GBP1F represents a compound with anxiolytic- and antidepressant-like effects though further studies are warranted at the molecular level to focus on the precise mechanism(s) of action.


Subject(s)
Anti-Anxiety Agents , Fluoxetine , Mice , Animals , Fluoxetine/pharmacology , Gabapentin/pharmacology , Dopamine/pharmacology , Depression/drug therapy , Serotonin , Anti-Anxiety Agents/pharmacology , Hydroxyindoleacetic Acid/pharmacology , Disease Models, Animal , Antidepressive Agents/pharmacology , Anxiety , Cognition , Ascorbic Acid/pharmacology , Behavior, Animal
17.
J Pain ; 24(9): 1681-1695, 2023 09.
Article in English | MEDLINE | ID: mdl-37169156

ABSTRACT

Spinal cord injury (SCI)-induced neuropathic pain (SCI-NP) develops in up to 60 to 70% of people affected by traumatic SCI, leading to a major decline in quality of life and increased risk for depression, anxiety, and addiction. Gabapentin and pregabalin, together with antidepressant drugs, are commonly prescribed to treat SCI-NP, but their efficacy is unsatisfactory. The limited efficacy of current pharmacological treatments for SCI-NP likely reflects our limited knowledge of the underlying mechanism(s) responsible for driving the maintenance of SCI-NP. The leading hypothesis in the field supports a major role for spontaneously active injured nociceptors in driving the maintenance of SCI-NP. Recent data from our laboratory provided additional support for this hypothesis and identified the T-type calcium channels as key players in driving the spontaneous activity of SCI-nociceptors, thus providing a rational pharmacological target to treat SCI-NP. To test whether T-type calcium channels contribute to the maintenance of SCI-NP, male and female SCI and sham rats were treated with TTA-P2 (a blocker of T-type calcium channels) to determine its effects on mechanical hypersensitivity (as measured with the von Frey filaments) and spontaneous ongoing pain (as measured with the conditioned place preference paradigm), and compared them to the effects of gabapentin, a blocker of high voltage-activated calcium channels. We found that both TTA-P2 and gabapentin reduced mechanical hypersensitivity in male and females SCI rats, but surprisingly only TTA-P2 reduced spontaneous ongoing pain in male SCI rats. PERSPECTIVES: SCI-induced neuropathic pain, and in particular the spontaneous ongoing pain component, is notoriously very difficult to treat. Our data provide evidence that inhibition of T-type calcium channels reduces spontaneous ongoing pain in SCI rats, supporting a clinically relevant role for T-type channels in the maintenance of SCI-induced neuropathic pain.


Subject(s)
Calcium Channels, T-Type , Neuralgia , Spinal Cord Injuries , Rats , Male , Female , Animals , Gabapentin/pharmacology , Calcium Channels, T-Type/pharmacology , Calcium Channels, T-Type/therapeutic use , Rats, Sprague-Dawley , Quality of Life , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Neuralgia/drug therapy , Neuralgia/etiology , Spinal Cord
18.
Nature ; 619(7969): 410-419, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37196677

ABSTRACT

Voltage-gated ion channels (VGICs) comprise multiple structural units, the assembly of which is required for function1,2. Structural understanding of how VGIC subunits assemble and whether chaperone proteins are required is lacking. High-voltage-activated calcium channels (CaVs)3,4 are paradigmatic multisubunit VGICs whose function and trafficking are powerfully shaped by interactions between pore-forming CaV1 or CaV2 CaVα1 (ref. 3), and the auxiliary CaVß5 and CaVα2δ subunits6,7. Here we present cryo-electron microscopy structures of human brain and cardiac CaV1.2 bound with CaVß3 to a chaperone-the endoplasmic reticulum membrane protein complex (EMC)8,9-and of the assembled CaV1.2-CaVß3-CaVα2δ-1 channel. These structures provide a view of an EMC-client complex and define EMC sites-the transmembrane (TM) and cytoplasmic (Cyto) docks; interaction between these sites and the client channel causes partial extraction of a pore subunit and splays open the CaVα2δ-interaction site. The structures identify the CaVα2δ-binding site for gabapentinoid anti-pain and anti-anxiety drugs6, show that EMC and CaVα2δ interactions with the channel are mutually exclusive, and indicate that EMC-to-CaVα2δ hand-off involves a divalent ion-dependent step and CaV1.2 element ordering. Disruption of the EMC-CaV complex compromises CaV function, suggesting that the EMC functions as a channel holdase that facilitates channel assembly. Together, the structures reveal a CaV assembly intermediate and EMC client-binding sites that could have wide-ranging implications for the biogenesis of VGICs and other membrane proteins.


Subject(s)
Calcium Channels, L-Type , Endoplasmic Reticulum , Membrane Proteins , Humans , Binding Sites , Brain , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/ultrastructure , Cryoelectron Microscopy , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Gabapentin/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Myocardium/chemistry
19.
Nature ; 617(7961): 599-607, 2023 May.
Article in English | MEDLINE | ID: mdl-37138086

ABSTRACT

Gliomas synaptically integrate into neural circuits1,2. Previous research has demonstrated bidirectional interactions between neurons and glioma cells, with neuronal activity driving glioma growth1-4 and gliomas increasing neuronal excitability2,5-8. Here we sought to determine how glioma-induced neuronal changes influence neural circuits underlying cognition and whether these interactions influence patient survival. Using intracranial brain recordings during lexical retrieval language tasks in awake humans together with site-specific tumour tissue biopsies and cell biology experiments, we find that gliomas remodel functional neural circuitry such that task-relevant neural responses activate tumour-infiltrated cortex well beyond the cortical regions that are normally recruited in the healthy brain. Site-directed biopsies from regions within the tumour that exhibit high functional connectivity between the tumour and the rest of the brain are enriched for a glioblastoma subpopulation that exhibits a distinct synaptogenic and neuronotrophic phenotype. Tumour cells from functionally connected regions secrete the synaptogenic factor thrombospondin-1, which contributes to the differential neuron-glioma interactions observed in functionally connected tumour regions compared with tumour regions with less functional connectivity. Pharmacological inhibition of thrombospondin-1 using the FDA-approved drug gabapentin decreases glioblastoma proliferation. The degree of functional connectivity between glioblastoma and the normal brain negatively affects both patient survival and performance in language tasks. These data demonstrate that high-grade gliomas functionally remodel neural circuits in the human brain, which both promotes tumour progression and impairs cognition.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Pathways , Humans , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Thrombospondin 1/antagonists & inhibitors , Gabapentin/pharmacology , Gabapentin/therapeutic use , Disease Progression , Cognition , Survival Rate , Wakefulness , Biopsy , Cell Proliferation/drug effects
20.
Altern Ther Health Med ; 29(5): 380-385, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37235490

ABSTRACT

Context: Sensory nervous-system diseases are chronic diseases that injury or disease of the somatosensory nervous system causes. Sleep disorders usually accompany these diseases, and in turn, worsen their conditions and form a vicious circle that brings great difficulties in clinical treatment. Objective: The study intended to systematically evaluate the clinical efficacy and safety of gabapentin in improving the sleep quality of patients with sensory nervous-system diseases using a meta-analysis, so as to provide evidence-based medical evidence for clinical treatment. Design: The research team performed a comprehensive narrative review by searching the China National Knowledge Infrastructure (CNKI), Chinese Scientific Journal (VIP), WANFANG, Chinese Biomedical Database (CBM), PubMed, Embase, Cochrane Library, and ClinicalTrials.gov databases. The search terms included gabapentin, 1-(aminomethyl)-cyclohexaneacetic acid, gabapentin hexal, gabapentin-ratiopharm, sleep, and insomnia. Setting: The review took place in the Department of Neurology at the First People's Hospital of Linping District in Hangzhou, China. Outcome Measures: The research team extracted the data from the studies meeting the inclusion criteria and then transferred them into the Review Manager 5.3 software for meta-analysis. The outcome measures included scores: (1) for the improvement in the degree of sleep interference score; (2) for the improvement in sleep quality; (3) for the rate of poor sleep quality; (4) for the rate awakenings of >5 per night; and (5) for the incidence of adverse reactions. Results: The research team found eight RCTs with 1269 participants, including 637 participants in a gabapentin test group and 632 participants in the placebo control group. The meta-analysis showed that the decrease in the degree of sleep interference [mean deviation (MD) = -0.86, 95% CI: (-0.91, -0.82), P < .00001] and the improvement in sleep quality [odds ratio (OR) = 2.64, 95% CI: (1.90, 3.67), P < .00001] in gabapentin group were significantly higher than those in placebo group (P < .05), while the rate of poor sleep quality [OR = 0.43, 95% CI: (0.23, 0.79), P = .007] and the rate of > 5 night awakenings [OR = 0.01, 95% CI: (0.05, 0.70), P = .01] in gabapentin group were significantly lower than those in placebo group (P < .05). No statistically significant differences existed in the incidence of adverse reactions between the two groups. Conclusions: Gabapentin is safe and effective in improving the sleep quality of patients with sensory nervous-system diseases. Due to the limitations of sample size and types of diseases in the current study, the field needs multicenter, large-sample, and high-quality RCTs for further validation in the future.


Subject(s)
Sleep Initiation and Maintenance Disorders , Sleep Quality , Humans , Gabapentin/therapeutic use , Gabapentin/pharmacology , Sleep/physiology , Sleep Initiation and Maintenance Disorders/drug therapy , Chronic Disease , Multicenter Studies as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...