Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 263
1.
Br J Pharmacol ; 181(12): 1734-1756, 2024 Jun.
Article En | MEDLINE | ID: mdl-38157867

BACKGROUND AND PURPOSE: Neuropathic pain affects up to 10% of the global population and is caused by an injury or a disease affecting the somatosensory, peripheral, or central nervous system. NP is characterized by chronic, severe and opioid-resistant properties. Therefore, its clinical management remains very challenging. The N-type voltage-gated calcium channel, Cav2.2, is a validated target for therapeutic intervention in chronic and neuropathic pain. The conotoxin ziconotide (Prialt®) is an FDA-approved drug that blocks Cav2.2 channel but needs to be administered intrathecally. Thus, although being principally efficient, the required application route is very much in disfavour. EXPERIMENTAL APPROACH AND KEY RESULTS: Here, we describe an orally available drug candidate, RD2, which competes with ziconotide binding to Cav2.2 at nanomolar concentrations and inhibits Cav2.2 almost completely reversible. Other voltage-gated calcium channel subtypes, like Cav1.2 and Cav3.2, were affected by RD2 only at concentrations higher than 10 µM. Data from sciatic inflammatory neuritis rat model demonstrated the in vivo proof of concept, as low-dose RD2 (5 mg·kg-1) administered orally alleviated neuropathic pain compared with vehicle controls. High-dose RD2 (50 mg·kg-1) was necessary to reduce pain sensation in acute thermal response assessed by the tail flick test. CONCLUSIONS AND IMPLICATIONS: Taken together, these results demonstrate that RD2 has antiallodynic properties. RD2 is orally available, which is the most convenient application form for patients and caregivers. The surprising and novel result from standard receptor screens opens the room for further optimization into new promising drug candidates, which address an unmet medical need.


Calcium Channel Blockers , Calcium Channels, N-Type , Neuralgia , Animals , Humans , Male , Mice , Rats , Administration, Oral , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/drug effects , Dose-Response Relationship, Drug , Mice, Inbred C57BL , Neuralgia/drug therapy , omega-Conotoxins/administration & dosage , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use , Rats, Inbred Lew
2.
Angew Chem Int Ed Engl ; 62(29): e202302812, 2023 07 17.
Article En | MEDLINE | ID: mdl-37148162

Ziconotide (ω-conotoxin MVIIA) is an approved analgesic for the treatment of chronic pain. However, the need for intrathecal administration and adverse effects have limited its widespread application. Backbone cyclization is one way to improve the pharmaceutical properties of conopeptides, but so far chemical synthesis alone has been unable to produce correctly folded and backbone cyclic analogues of MVIIA. In this study, an asparaginyl endopeptidase (AEP)-mediated cyclization was used to generate backbone cyclic analogues of MVIIA for the first time. Cyclization using six- to nine-residue linkers did not perturb the overall structure of MVIIA, and the cyclic analogues of MVIIA showed inhibition of voltage-gated calcium channels (CaV 2.2) and substantially improved stability in human serum and stimulated intestinal fluid. Our study reveals that AEP transpeptidases are capable of cyclizing structurally complex peptides that chemical synthesis cannot achieve and paves the way for further improving the therapeutic value of conotoxins.


Conotoxins , omega-Conotoxins , Humans , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Conotoxins/pharmacology , Calcium Channels/chemistry , Calcium Channel Blockers/pharmacology
3.
J Ethnopharmacol ; 313: 116531, 2023 Sep 15.
Article En | MEDLINE | ID: mdl-37164254

ETHNOPHARMACOLOGICAL RELEVANCE: Prangos ferulacea (L.) Lindl is an Apiaceae plant, widely used in traditional medicine. Recently, chemical composition and biological activities of its essential oil (Prangroil) have been reported, but there are no studies on possible effects on intestinal contractility. AIMS OF THE STUDY: We investigated the effects of essential oil Sicilian Prangoil on the contractility of rat small (duodenum) and large (colon) intestine and the related action mechanism. MATERIALS AND METHODS: Responses to Prangoil and to its major component ß-ocimen in intestinal segments were assessed in vitro as changes in isometric tension. RESULTS: Prangoil, induced in duodenum, depending upon doses, contraction and/or muscular relaxation. Instead, in colon Prangoil only reduced the phasic contractions and induced muscular relaxation. ß-ocimen, in both segments, produced only reduction of the spontaneous contractions without affecting basal tone. Prangoil contractile effects were abolished by ω-conotoxin, neural N-type Ca2+ channels blocker, atropine, muscarinic receptor antagonist, neostigmine, acetylcholinesterase (AChE) inhibitor, suggesting that Prangoil-induced contraction would be the result of an increase in neuronal cholinergic activity. Prangoil and ß-ocimen inhibitory effects were unaffected by ω-conotoxin, L-NAME, blocker of the NO synthase, ODQ, soluble guanylate cyclase inhibitor, excluding involvement of neurotransmitter release or NO synthesis in the inhibitory effects. Potassium channel blocker did not affect Prangoil or ß-ocimen inhibitory responses. Prangoil or ß-ocimen inhibited the Ca2+ and high-KCl solution -induced contractions and the Carbachol-induced contractions in calcium free solution. CONCLUSION: Prangoil affects the contractility of small and large intestine in rat, with regional differences, via potentiation of neural cholinergic activity, blockade of L-type voltage-gated calcium channel and reduction of Ca2+ release from the intracellular store. The Prangroil main components, ß-ocimen, contributes to the inhibitory effects.


Acetylcholinesterase , omega-Conotoxins , Rats , Animals , Muscle, Smooth , Muscle Contraction , Colon , Cholinergic Agents/pharmacology , Calcium Channels, L-Type/physiology , omega-Conotoxins/pharmacology
4.
Mar Drugs ; 21(4)2023 Apr 01.
Article En | MEDLINE | ID: mdl-37103368

Chronic pain is one of the most prevalent health problems worldwide. An alternative to suppress or alleviate chronic pain is the use of peptide drugs that block N-type Ca2+ channels (Cav2.2), such as ω-conotoxin MVIIA. Nevertheless, the narrow therapeutic window, severe neurological side effects and low stability associated with peptide MVIIA have restricted its widespread use. Fortunately, self-assembly endows the peptide with high stability and multiple functions, which can effectively control its release to prolong its duration of action. Inspired by this, MVIIA was modified with appropriate fatty acid chains to render it amphiphilic and easier to self-assemble. In this paper, an N-terminal myristoylated MVIIA (Myr-MVIIA, medium carbon chain length) was designed and prepared to undergo self-assembly. The present results indicated that Myr-MVIIA can self-assemble into micelles. Self-assembled micelles formed by Myr-MVIIA at higher concentrations than MVIIA can prolong the duration of the analgesic effect and significantly reduce or even eliminate the side effects of tremor and coordinated motor dysfunction in mice.


Chronic Pain , omega-Conotoxins , Mice , Animals , Chronic Pain/drug therapy , Micelles , omega-Conotoxins/pharmacology , Peptides/pharmacology , Calcium Channel Blockers/pharmacology
5.
J Control Release ; 356: 448-462, 2023 04.
Article En | MEDLINE | ID: mdl-36898532

Ziconotide (ZIC) is an N-type calcium channel antagonist for treating severe chronic pain that is intolerable, or responds poorly to the administration of other drugs, such as intrathecal morphine and systemic analgesics. As it can only work in the brain and cerebrospinal fluid, intrathecal injection is the only administration route for ZIC. In this study, borneol (BOR)-modified liposomes (LIPs) were fused with exosomes from mesenchymal stem cells (MSCs) and loaded with ZIC to prepare microneedles (MNs) to improve the efficiency of ZIC across the blood-brain barrier. To evaluate local analgesic effects of MNs, the sensitivity of behavioral pain to thermal and mechanical stimuli was tested in animal models of peripheral nerve injury, diabetes-induced neuropathy pain, chemotherapy-induced pain, and ultraviolet-B (UV-B) radiation-induced neurogenic inflammatory pain. BOR-modified LIPs loaded with ZIC were spherical or nearly spherical, with a particle size of about 95 nm and a Zeta potential of -7.8 mV. After fusion with MSC exosomes, the particle sizes of LIPs increased to 175 nm, and their Zeta potential increased to -3.8 mV. The nano-MNs constructed based on BOR-modified LIPs had good mechanical properties and could effectively penetrate the skin to release drugs. The results of analgesic experiments showed that ZIC had a significant analgesic effect in different pain models. In conclusion, the BOR-modified LIP membrane-fused exosome MNs constructed in this study for delivering ZIC provide a safe and effective administration for chronic pain treatment, as well as great potential for clinical application of ZIC.


Analgesia , Chronic Pain , Exosomes , Neuralgia , omega-Conotoxins , Animals , Liposomes/therapeutic use , Chronic Pain/drug therapy , omega-Conotoxins/therapeutic use , omega-Conotoxins/pharmacology , Analgesics , Neuralgia/drug therapy
6.
Mol Pain ; 18: 17448069221129829, 2022 04.
Article En | MEDLINE | ID: mdl-36113096

Development of chronic pain has been attributed to dysfunctional GABA signaling in the spinal cord. Direct pharmacological interventions on GABA signaling are usually not very efficient and often accompanied by side effects due to the widespread distribution of GABA receptors in CNS. Transplantation of GABAergic neuronal cells may restore the inhibitory potential in the spinal cord. Grafted cells may also release additional analgesic peptides by means of genetic engineering to further enhance the benefits of this approach. Conopeptides are ideal candidates for recombinant expression using cell-based strategies. The omega-conopeptide MVIIA is in clinical use for severe pain marketed as FDA approved Prialt in the form of intrathecal injections. The goal of this study was to develop transplantable recombinant GABAergic cells releasing conopeptide MVIIA and to evaluate the analgesic effect of the grafts in a model of peripheral nerve injury-induced pain. We have engineered and characterized the GABAergic progenitors expressing MVIIA. Recombinant and nonrecombinant cells were intraspinally injected into animals after the nerve injury. Animals were tested weekly up to 12 weeks for the presence of hypersensitivity, followed by histochemical and biochemical analysis of the tissue. We observed beneficial effects of the grafted cells in reducing hypersensitivity in all grafted animals, especially potent in the recombinant group. The level of pain-related cytokines was reduced in the grafted animals and correlation between these pain markers and actual behavior was indicated. This study demonstrated the feasibility of recombinant cell transplantation in the management of chronic pain.


Chronic Pain , Peripheral Nerve Injuries , omega-Conotoxins , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Chronic Pain/drug therapy , Cytokines , Peptides , Peripheral Nerve Injuries/drug therapy , Rats , Receptors, GABA , gamma-Aminobutyric Acid , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
7.
Mol Neurobiol ; 59(7): 4436-4452, 2022 Jul.
Article En | MEDLINE | ID: mdl-35570263

CTK 01512-2 toxin is a recombinant peptide of the Phα1ß version derived from the venom of the Phoneutria nigriventer spider. It acts as an N-type voltage-gated calcium channel (VGCC) blocker and shows a prolonged effect on preventing and reducing nociception. Herein, CTK 01512-2 was tested on two models of persistent pain, the chronic post-ischemia pain (CPIP) and the paclitaxel-induced peripheral neuropathy, to evaluate its systemic, intrathecal, and intracerebroventricular effects on mechanical hypersensitivity and thermal allodynia. Glial cell viability was also investigated using the MTT test. The results showed that CTK 01512-2 intrathecal and systemic treatments reduced the mechanical hypersensitivity induced by CPIP, mainly between 1-4 h after its administration. Additionally, intrathecal treatment reduced the CPIP-induced thermal allodynia. In its turn, the intracerebroventricular treatment showed mechanical antihyperalgesic and thermal antiallodynic effects in the paclitaxel-induced peripheral neuropathy. These data reinforce the therapeutic potential of CTK 01512-2 to treat persistent pain conditions and offer a perspective to use the systemic route. Moreover, CTK 01512-2 increased the glial cell viability in the MTT reduction assay, and it may indicate a new approach to managing chronic pain. The results found in this study help to pave new perspectives of pain relief treatments to patients affected by chronic pain.


Chronic Pain , Spider Venoms , omega-Conotoxins , Animals , Calcium Channel Blockers/pharmacology , Chronic Pain/drug therapy , Disease Models, Animal , Humans , Hyperalgesia/drug therapy , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Spider Venoms/pharmacology , Spider Venoms/therapeutic use , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
8.
Cell Rep ; 37(5): 109931, 2021 11 02.
Article En | MEDLINE | ID: mdl-34731621

N-type voltage-gated calcium (CaV) channels mediate Ca2+ influx at presynaptic terminals in response to action potentials and play vital roles in synaptogenesis, release of neurotransmitters, and nociceptive transmission. Here, we elucidate a cryo-electron microscopy (cryo-EM) structure of the human CaV2.2 complex in apo, ziconotide-bound, and two CaV2.2-specific pore blockers-bound states. The second voltage-sensing domain (VSD) is captured in a resting-state conformation, trapped by a phosphatidylinositol 4,5-bisphosphate (PIP2) molecule, which is distinct from the other three VSDs of CaV2.2, as well as activated VSDs observed in previous structures of CaV channels. This structure reveals the molecular basis for the unique inactivation process of CaV2.2 channels, in which the intracellular gate formed by S6 helices is closed and a W-helix from the domain II-III linker stabilizes closed-state inactivation. The structures of this inactivated, drug-bound complex lay a solid foundation for developing new state-dependent blockers for treatment of chronic pain.


Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Dipeptides/pharmacology , Ion Channel Gating/drug effects , omega-Conotoxins/pharmacology , Action Potentials , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/ultrastructure , Calcium Signaling , Cryoelectron Microscopy , HEK293 Cells , Humans , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Conformation, alpha-Helical , Structure-Activity Relationship
9.
Nature ; 596(7870): 143-147, 2021 08.
Article En | MEDLINE | ID: mdl-34234349

The neuronal-type (N-type) voltage-gated calcium (Cav) channels, which are designated Cav2.2, have an important role in the release of neurotransmitters1-3. Ziconotide is a Cav2.2-specific peptide pore blocker that has been clinically used for treating intractable pain4-6. Here we present cryo-electron microscopy structures of human Cav2.2 (comprising the core α1 and the ancillary α2δ-1 and ß3 subunits) in the presence or absence of ziconotide. Ziconotide is thoroughly coordinated by helices P1 and P2, which support the selectivity filter, and the extracellular loops (ECLs) in repeats II, III and IV of α1. To accommodate ziconotide, the ECL of repeat III and α2δ-1 have to tilt upward concertedly. Three of the voltage-sensing domains (VSDs) are in a depolarized state, whereas the VSD of repeat II exhibits a down conformation that is stabilized by Cav2-unique intracellular segments and a phosphatidylinositol 4,5-bisphosphate molecule. Our studies reveal the molecular basis for Cav2.2-specific pore blocking by ziconotide and establish the framework for investigating electromechanical coupling in Cav channels.


Analgesics, Non-Narcotic/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Cryoelectron Microscopy , omega-Conotoxins/pharmacology , Calcium Channels, N-Type/ultrastructure , Humans , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Protein Conformation/drug effects , Protein Stability/drug effects
10.
Mar Drugs ; 19(2)2021 Feb 11.
Article En | MEDLINE | ID: mdl-33670311

The peripheral effects of ω-conotoxins, selective blockers of N-type voltage-gated calcium channels (CaV2.2), have not been characterised across different clinically relevant pain models. This study examines the effects of locally administered ω-conotoxin MVIIA, GVIA, and CVIF on mechanical and thermal paw withdrawal threshold (PWT) in postsurgical pain (PSP), cisplatin-induced neuropathy (CisIPN), and oxaliplatin-induced neuropathy (OIPN) rodent models. Intraplantar injection of 300, 100 and 30 nM MVIIA significantly (p < 0.0001, p < 0.0001, and p < 0.05, respectively) alleviated mechanical allodynia of mice in PSP model compared to vehicle control group. Similarly, intraplantar injection of 300, 100, and 30 nM MVIIA (p < 0.0001, p < 0.01, and p < 0.05, respectively), and 300 nM and 100 nM GVIA (p < 0.0001 and p < 0.05, respectively) significantly increased mechanical thresholds of mice in OIPN model. The ED50 of GVIA and MVIIA in OIPN was found to be 1.8 pmol/paw and 0.8 pmol/paw, respectively. However, none of the ω-conotoxins were effective in a mouse model of CisIPN. The intraplantar administration of 300 nM GVIA, MVIIA, and CVIF did not cause any locomotor side effects. The intraplantar administration of MVIIA can alleviate incision-induced mechanical allodynia, and GVIA and MVIIA effectively reduce OIPN associated mechanical pain, without locomotor side effects, in rodent models. In contrast, CVIF was inactive in these pain models, suggesting it is unable to block a subset of N-type voltage-gated calcium channels associated with nociceptors in the skin.


Calcium Channel Blockers/pharmacology , Peripheral Nervous System Diseases/drug therapy , omega-Conotoxins/pharmacology , Acute Pain/drug therapy , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channels, N-Type/drug effects , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Hyperalgesia/drug therapy , Injections, Subcutaneous , Male , Mice , Mice, Inbred C57BL , omega-Conotoxin GVIA/administration & dosage , omega-Conotoxin GVIA/pharmacology , omega-Conotoxins/administration & dosage
11.
Eur J Pharmacol ; 891: 173672, 2021 Jan 15.
Article En | MEDLINE | ID: mdl-33190801

Agents that modulate the activity of high-voltage gated calcium channels (HVCCs) exhibit experimentally and clinically significant effect by relieving visceral pain. Among these agents, the toxins Phα1ß and ω-conotoxin MVIIA effectively reduce chronic pain in rodent models. The molecular mechanisms underlying the chronic pain associated with acute pancreatitis (AP) are poorly understood. Hypercalcemia is a risk factor; the role of cytosolic calcium is considered to be a modulator of pancreatitis. Blockade of Ca2+ signals may be useful as a prophylactic treatment of pancreatitis. We explored the pathophysiological roles of three peptide toxins: Phα1ß and its recombinant form CTK 01512-2-blockers of TRPA1 receptor and HVCCs and ω-conotoxin MVIIA, a specific blocker of N-type calcium channels in cerulein-induced AP. Cerulein injection elicits AP in rats, evidenced by an increase in hyperalgesic pain, inflammatory infiltration, amylase and lipase secretion, and reactive oxygen species, TNF-α, and p65 NF-κB levels. These effects of cerulein-induced AP were abolished by Phα1ß and its recombinant form CTK 01512-2, whereas ω-conotoxin MVIIA had no effect on the induced increase in pancreatic enzyme secretion. Our results demonstrate that Phα1ß and CTK 01512-2 toxins-antagonists of HVCCs and TRPA1 receptor presented an effective response profile, in the control of nociception and inflammatory process in the AP model in rats, without causing changes in spontaneous locomotion of the rats.


Abdominal Pain/prevention & control , Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Hyperalgesia/prevention & control , Pain Threshold/drug effects , Pancreatitis/prevention & control , Abdominal Pain/etiology , Abdominal Pain/metabolism , Abdominal Pain/physiopathology , Animals , Behavior, Animal/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Ceruletide , Disease Models, Animal , Exploratory Behavior/drug effects , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Male , Neuropeptides/pharmacology , Pancreas/drug effects , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/physiopathology , Rats, Wistar , Spider Venoms/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , omega-Conotoxins/pharmacology
12.
Behav Brain Res ; 390: 112647, 2020 07 15.
Article En | MEDLINE | ID: mdl-32428635

The lack of oral or injectable formulations of ziconotide (ω-conotoxin peptide), a novel analgesic agent, limits research on potential neurobehavioral protective properties of this substance, including antidepressant-like effects. Here we expose rats to a stress paradigm that induces depression and memory impairment to assess the effects of ziconotide treatment. Ziconotide was administered intracerebroventricular (i.c.v.) to rats undergoing stereotaxic surgery at a single dose (1 µg/rat) or in repeated long-term applications (dosage groups: 0.1, 0.3, and 1 µg/rat). The antidepressant activity and memory-enhancing effects of ziconotide were examined via the forced swimming test, the Morris water maze test, and the passive avoidance learning test. Behavioral results showed that long-term i.c.v. ziconotide administration significantly decreased the immobility time and delayed the latency period to immobility in a dose-dependent manner compared to controls. In the passive avoidance learning test, the latency period increased, and in the Morris water maze test, the platform location latency time decreased. A single dose of ziconotide (1 µg/rat) did not show a significant effect on memory function or depression parameters during the same tests. Animals were sacrificed immediately after behavioral testing, and both hippocampi were removed and prepared for BDNF evaluation. Hippocampal BDNF levels were significantly increased in rats receiving long-term i.c.v. ziconotide compared to controls. Our results suggest that long-term consumption of ziconotide may attenuate the severity of depression-like behavior and could be useful for preventing memory impairments in various learning models by elevating BDNF levels.


Antidepressive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Depression/drug therapy , Hippocampus/drug effects , Memory Disorders/drug therapy , omega-Conotoxins/pharmacology , Animals , Antidepressive Agents/administration & dosage , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Calcium Channel Blockers/administration & dosage , Disease Models, Animal , Hippocampus/metabolism , Male , Maze Learning/drug effects , Rats , Rats, Wistar , omega-Conotoxins/administration & dosage
13.
Pharmacol Rep ; 72(3): 600-611, 2020 Jun.
Article En | MEDLINE | ID: mdl-32399819

BACKGROUND: Orofacial pain is clinically challenging, having therapeutic failures and side effects. This study evaluated the antinociceptive activities of the CTK 01512-2 toxin, the TRPA1 channel antagonist, and the selective inhibitor of the N-type voltage-gated calcium channels (N-type VGCC), in different pain models. MATERIALS AND METHODS: The trigeminal ganglia were stimulated in vitro with capsaicin. The in vivo models received subcutaneous (sc) injections of formalin into the upper lip of the rats, Freund's Complete Adjuvant (FCA) into the temporomandibular joint (TMJ), and infraorbital nerve constrictions (IONC). CTK 01512-2 at concentrations of 30, 100, and 300 pmol/site, intrathecally (ith), and MVIIA at 10, 30, and 100 pmol/site in the formalin test, guided the doses for the models. The glutamate levels in the CSF of the rats that were submitted to IONC were analyzed. RESULTS: CTK 01512-2 decreased the nociceptive behavior in the inflammatory phase of the formalin test (65.94 ± 7.35%) and MVIIA in the neurogenic phase (81.23 ± 3.36%). CTK 01512-2 reduced facial grooming with FCA in the TMJ (96.7 ± 1.6%), and in the IONC neuropathy model, it decreased heat hyperalgesia (100%) and cold hyperalgesia (81.61 ± 9.02%). The levels of glutamate in the trigeminal ganglia in vitro (81.40 ± 8.59%) and in the CSF in vivo (70.0 ± 9.2%) were reduced. CONCLUSIONS: The roles of TRPA1 in pain transduction and the performance of CTK 01512-2 in the inhibition of the N-type VGCCs were reinforced. This dual activity may represent an advantage in clinical treatments.


Analgesics/pharmacology , Facial Pain/drug therapy , TRPA1 Cation Channel/antagonists & inhibitors , omega-Conotoxins/pharmacology , Animals , Calcium Channels, N-Type/metabolism , Capsaicin/pharmacology , Disease Models, Animal , Freund's Adjuvant , Glutamic Acid/metabolism , Hyperalgesia/drug therapy , Male , Neuralgia/drug therapy , Pain Measurement , Rats , Rats, Wistar
14.
Pharmacol Rep ; 72(1): 47-54, 2020 Feb.
Article En | MEDLINE | ID: mdl-32016848

BACKGROUND: Diabetic neuropathy is a common cause of painful diabetic neuropathy (PDN). C-X-C chemokine receptor type 4 (CXCR4) expression is increased in peripheral nerve samples from diabetes patients, suggesting a role for CXCR4 in PDN. Therefore, we evaluated the effects of Phα1ß, ω-conotoxin MVIIA, and AMD3100 in a model of streptozotocin (STZ)-induced PDN in rodents and naïve model of rats with the activation of the CXCR4/stromal cell-derived factor 1 (SDF-1) signal. METHODS: Diabetic neuropathy was induced by intraperitoneal (ip) injection of STZ in Wistar rats. Naïve rats were intrathecally injected with SDF-1 to test the CXCR4/SDF-1 signal. The effects of Phα1ß intrathecal (it), ω-conotoxin MVIIA intrathecal (it), and AMD3100 intraperitoneal (ip) on rat hypersensitivity, IL-6, and the intracellular calcium [Ca2+]i content of diabetic synaptosomes were studied. RESULTS: The drugs reduced the hypersensitivity in diabetic rats. SDF-1 (1.0 µg/it) administration in naïve rats induced hypersensitivity. Phα1ß (100 pmol/it) or AMD3100 (2.5 µg/ip) reduced this hypersensitivity after 2 h treatments, while ω-conotoxin MVIIA did not have an effect. IL-6 and [Ca2+]i content increased in the spinal cord synaptosomes in diabetic rats. The drug treatments reduced IL-6 and the calcium influx in diabetic synaptosomes. CONCLUSIONS: Phα1ß, ω-conotoxin MVIIA, and AMD3100, after 2 h of treatment of STZ-induced PDN, reduced hypersensitivity in diabetic rats. In naïve rats with CXCR4/SDF-1 activation, the induced hypersensitivity decreased after 2 h treatments with Phα1ß or AMD-3100, while ω-conotoxin MVIIA did not affect. The inhibitory effects of Phα1ß on PDN may involve voltage-dependent calcium channels.


Analgesics/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Spider Venoms/pharmacology , Animals , Benzylamines , Calcium/metabolism , Calcium Channels/metabolism , Chemokine CXCL12/metabolism , Cyclams , Diabetes Mellitus, Experimental/complications , Heterocyclic Compounds/pharmacology , Rats , Rats, Wistar , Receptors, CXCR4/metabolism , omega-Conotoxins/pharmacology
15.
Eur J Pharmacol ; 865: 172781, 2019 Dec 15.
Article En | MEDLINE | ID: mdl-31706856

Inhibiting glutamate release can reduce neuronal excitability and is recognized as a key mechanism of anti-epileptic drugs. In this study, by using isolated nerve terminal (synaptosome) and slice preparations, we investigated the effect of asiatic acid, a triterpene isolated from Centella asiatica with antiepileptic activity, on glutamate release in the hippocampus of rats. In hippocampal synaptosomes, application of asiatic acid resulted in a concentration-dependent inhibition of 4-aminopyridine-evoked glutamate release. This inhibitory action was dependent on extracellular calcium, blocked by inhibiting the vesicular transporter, but was unaffected by inhibiting the glutamate transporter. In addition, asiatic acid decreased the 4-aminopyridine-induced increase in the intraterminal calcium and failed to alter the synaptosomal potential. Furthermore, the asiatic acid-mediated release inhibition was significantly suppressed by the N- and P/Q-type calcium channel inhibitor ω-conotoxin MVIIC or protein kinase C inhibitor GF109203X. Western blotting data in synaptosomes also revealed that asiatic acid reduced 4-aminopyridine-induced phosphorylation of protein kinase C. In hippocampal slices, asiatic acid decreased the frequencies of spontaneous excitatory postsynaptic currents without changing their amplitudes and glutamate-activated currents in CA3 pyramidal neurons. We also observed that asiatic acid significantly suppressed 4-aminopyridine-induced burst firing. These data suggest that, in rat hippocampal nerve terminals, asiatic acid attenuates the calcium influx via N- and P/Q-type calcium channels, subsequently suppressing protein kinase C activity and decreasing glutamate release.


Glutamic Acid/metabolism , Hippocampus/drug effects , Pentacyclic Triterpenes/pharmacology , 4-Aminopyridine/pharmacology , Animals , Calcium/metabolism , Centella , Hippocampus/metabolism , Hippocampus/physiology , Indoles/pharmacology , Male , Maleimides/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats, Sprague-Dawley , Synaptosomes/drug effects , Synaptosomes/metabolism , omega-Conotoxins/pharmacology
16.
Neuropharmacology ; 158: 107732, 2019 11 01.
Article En | MEDLINE | ID: mdl-31377198

Neuropeptide Y (NPY) modulates nociception in the spinal cord, but little is known about its mechanisms of release. We measured NPY release in situ using the internalization of its Y1 receptor in dorsal horn neurons. Y1 receptor immunoreactivity was normally localized to the cell surface, but addition of NPY to spinal cord slices increased the number of neurons with Y1 internalization in a biphasic fashion (EC50s of 1 nM and 1 µM). Depolarization with KCl, capsaicin, or the protein kinase A activator 6-benzoyl-cAMP also induced Y1 receptor internalization, presumably by releasing NPY. NMDA receptor activation in the presence of BVT948, an inhibitor of protein tyrosine phosphatases, also released NPY. Electrical stimulation of the dorsal horn frequency-dependently induced NPY release; and this was decreased by the Y1 antagonist BIBO3304, the Nav channel blocker lidocaine, or the Cav2 channel blocker ω-conotoxin MVIIC. Dorsal root immersion in capsaicin, but not its electrical stimulation, also induced NPY release. This was blocked by CNQX, suggesting that part of the NPY released by capsaicin was from dorsal horn neurons receiving synapses from primary afferents and not from the afferent themselves. Mechanical stimulation in vivo, with rub or clamp of the hindpaw, elicited robust Y1 receptor internalization in rats with spared nerve injury but not sham surgery. In summary, NPY is released from dorsal horn interneurons or primary afferent terminals by electrical stimulation and by activation of TRPV1, PKA or NMDA receptors in. Furthermore, NPY release evoked by noxious and tactile stimuli increases after peripheral nerve injury.


Cyclic AMP-Dependent Protein Kinases/metabolism , Neuropeptide Y/metabolism , Nociception/physiology , Peripheral Nerve Injuries/metabolism , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Neuropeptide Y/metabolism , TRPV Cation Channels/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Calcium Channel Blockers/pharmacology , Capsaicin/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hyperalgesia/metabolism , Indoles/pharmacology , Interneurons/drug effects , Interneurons/metabolism , Lidocaine/pharmacology , Male , Microscopy, Confocal , Neuropeptide Y/drug effects , Nociception/drug effects , Posterior Horn Cells/drug effects , Potassium Chloride/pharmacology , Protein Transport/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Rats , Receptors, Neuropeptide Y/drug effects , Sensory System Agents/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/metabolism , Voltage-Gated Sodium Channel Blockers/pharmacology , omega-Conotoxins/pharmacology
17.
Basic Clin Pharmacol Toxicol ; 124(5): 615-620, 2019 May.
Article En | MEDLINE | ID: mdl-30449066

Phα1ß peptide isolated from the venom of the Phoneutria nigriventer spider has shown higher analgesic action in pre-clinical studies than ω-conotoxin MVIIA peptide used to treat severe chronic pain. In view of the great potential for the development of a new Phα1ß-based drug, a Phα1ß recombinant form (CTK 01512-2) has been studied for efficacy and safety. The aim of this study was to evaluate cytotoxic, genotoxic and mutagenic effects of a Phα1ß recombinant form and compare it with native Phα1ß and ω-conotoxin MVIIA. Cytotoxicity was evaluated using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) colourimetric assay in L929 mouse fibroblast cells (0.5-10.0 µmol/L). Genotoxic and mutagenic activities were analysed using the alkaline comet assay in peripheral blood and spinal cord, and the micronucleus test in bone marrow from Wistar rats treated by intrathecal injection of CTK 01512-2 (200, 500 and 1000 pmol/site), native Phα1ß (500 pmol/site) and ω-conotoxin MVIIA (200 pmol/site). CTK 01512-2 decreased the cell viability of the L929, showing IC50 of 3.3 ± 0.1 µmol/L, while the Phα1ß and ω-conotoxin MVIIA did not show cytotoxicity (IC50  > 5.0 µmol/L). Native and recombinant Phα1ß forms induced DNA damage in the spinal cord, but not in peripheral blood. CTK 01512-2 at 1000 pmol/site increased the micronucleus frequency suggesting mutagenic effects. In conclusion, the recombinant form has cytotoxic, genotoxic and mutagenic effects, evidenced in doses five times above the therapeutic dose.


DNA Damage , Neurotoxins/pharmacology , Peptides/pharmacology , Spider Venoms/pharmacology , Spinal Cord/drug effects , Analgesics/pharmacology , Analgesics/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/drug effects , Inhibitory Concentration 50 , Male , Mice , Mutagenicity Tests , Mutagens , Neurotoxins/toxicity , Rats , Rats, Wistar , Spider Venoms/toxicity , omega-Conotoxins/pharmacology
18.
J Cell Physiol ; 234(7): 10977-10989, 2019 07.
Article En | MEDLINE | ID: mdl-30536400

Besides the nerve endings, the soma of trigeminal neurons also respond to membrane depolarizations with the release of neurotransmitters and neuromodulators in the extracellular space within the ganglion, a process potentially important for the cross-communication between neighboring sensory neurons. In this study, we addressed the dependence of somatic release on Ca2+ influx in trigeminal neurons and the involvement of the different types of voltage-gated Ca2+ (Cav) channels in the process. Similar to the closely related dorsal root ganglion neurons, we found two kinetically distinct components of somatic release, a faster component stimulated by voltage but independent of the Ca2+ influx, and a slower component triggered by Ca2+ influx. The Ca2+ -dependent component was inhibited 80% by ω-conotoxin-MVIIC, an inhibitor of both N- and P/Q-type Cav channels, and 55% by the P/Q-type selective inhibitor ω-agatoxin-IVA. The selective L-type Ca2+ channel inhibitor nimodipine was instead without effect. These results suggest a major involvement of N- and P/Q-, but not L-type Cav channels in the somatic release of trigeminal neurons. Thus antinociceptive Cav channel antagonists acting on the N- and P/Q-type channels may exert their function by also modulating the somatic release and cross-communication between sensory neurons.


Calcium/metabolism , Neurons/physiology , Trigeminal Nerve/cytology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/physiology , Mice , Mice, Inbred C57BL , Nimodipine/pharmacology , omega-Conotoxins/pharmacology
19.
Sci Rep ; 8(1): 13397, 2018 09 07.
Article En | MEDLINE | ID: mdl-30194442

Cone snails are a diverse group of predatory marine invertebrates that deploy remarkably complex venoms to rapidly paralyse worm, mollusc or fish prey. ω-Conotoxins are neurotoxic peptides from cone snail venoms that inhibit Cav2.2 voltage-gated calcium channel, demonstrating potential for pain management via intrathecal (IT) administration. Here, we isolated and characterized two novel ω-conotoxins, MoVIA and MoVIB from Conus moncuri, the first to be identified in vermivorous (worm-hunting) cone snails. MoVIA and MoVIB potently inhibited human Cav2.2 in fluorimetric assays and rat Cav2.2 in patch clamp studies, and both potently displaced radiolabeled ω-conotoxin GVIA (125I-GVIA) from human SH-SY5Y cells and fish brain membranes (IC50 2-9 pM). Intriguingly, an arginine at position 13 in MoVIA and MoVIB replaced the functionally critical tyrosine found in piscivorous ω-conotoxins. To investigate its role, we synthesized MoVIB-[R13Y] and MVIIA-[Y13R]. Interestingly, MVIIA-[Y13R] completely lost Cav2.2 activity and MoVIB-[R13Y] had reduced activity, indicating that Arg at position 13 was preferred in these vermivorous ω-conotoxins whereas tyrosine 13 is preferred in piscivorous ω-conotoxins. MoVIB reversed pain behavior in a rat neuropathic pain model, confirming that vermivorous cone snails are a new source of analgesic ω-conotoxins. Given vermivorous cone snails are ancestral to piscivorous species, our findings support the repurposing of defensive venom peptides in the evolution of piscivorous Conidae.


Analgesics/chemistry , Calcium Channel Blockers/chemistry , Evolution, Molecular , omega-Conotoxins/chemistry , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/metabolism , Cell Line, Tumor , Cells, Cultured , Ganglia, Spinal/cytology , Humans , Neuralgia/drug therapy , Neurons, Afferent/drug effects , Rats , Rats, Wistar , Snails , omega-Conotoxins/genetics , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
20.
Mol Neurobiol ; 55(12): 9307-9327, 2018 Dec.
Article En | MEDLINE | ID: mdl-29667130

Voltage-gated calcium channels (VGCCs) play a critical role in neuroinflammatory diseases, such as multiple sclerosis (MS). CTK 01512-2 is a recombinant version of the peptide Phα1ß derived from the spider Phoneutria nigriventer, which inhibits N-type VGCC/TRPA1-mediated calcium influx. We investigated the effects of this molecule in the mouse model of experimental autoimmune encephalomyelitis (EAE). The effects of CTK 01512-2 were compared to those displayed by ziconotide-a selective N-type VGCC blocker clinically used for chronic pain-and fingolimod-a drug employed for MS treatment. The intrathecal (i.t.) treatment with CTK 01512-2 displayed beneficial effects, by preventing nociception, body weight loss, splenomegaly, MS-like clinical and neurological scores, impaired motor coordination, and memory deficits, with an efficacy comparable to that observed for ziconotide and fingolimod. This molecule displayed a favorable profile on EAE-induced neuroinflammatory changes, including inflammatory infiltrate, demyelination, pro-inflammatory cytokine production, glial activation, and glucose metabolism in the brain and spinal cord. The recovery of spatial memory, besides a reduction of serum leptin levels, allied to central and peripheral elevation of the anti-inflammatory cytokine IL-10, was solely modulated by CTK 01512-2, dosed intrathecally. The intravenous (i.v.) administration of CTK 01512-2 also reduced the EAE-elicited MS-like symptoms, similarly to that seen in animals that received fingolimod orally. Ziconotide lacked any significant effect when dosed by i.v. route. Our results indicate that CTK 01512-2 greatly improved the neuroinflammatory responses in a mouse model of MS, with a higher efficacy when compared to ziconotide, pointing out this molecule as a promising adjuvant for MS management.


Calcium Channel Blockers/therapeutic use , Multiple Sclerosis/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Calcium Channel Blockers/pharmacology , Chemokines/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Inflammation/pathology , Inflammation Mediators/metabolism , Injections, Spinal , Mice, Inbred C57BL , Motor Activity/drug effects , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Nociception/drug effects , Peptide Fragments/metabolism , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use
...