Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Pain ; 161(7): 1670-1681, 2020 07.
Article En | MEDLINE | ID: mdl-32142016

Pituitary adenylate cyclase activating polypeptide-38 (PACAP38) may play an important role in primary headaches. Preclinical evidence suggests that PACAP38 modulates trigeminal nociceptive activity mainly through PAC1 receptors while clinical studies report that plasma concentrations of PACAP38 are elevated in spontaneous attacks of cluster headache and migraine and normalize after treatment with sumatriptan. Intravenous infusion of PACAP38 induces migraine-like attacks in migraineurs and cluster-like attacks in cluster headache patients. A rodent-specific PAC1 receptor antibody Ab181 was developed, and its effect on nociceptive neuronal activity in the trigeminocervical complex was investigated in vivo in an electrophysiological model relevant to primary headaches. Ab181 is potent and selective at the rat PAC1 receptor and provides near-maximum target coverage at 10 mg/kg for more than 48 hours. Without affecting spontaneous neuronal activity, Ab181 effectively inhibits stimulus-evoked activity in the trigeminocervical complex. Immunohistochemical analysis revealed its binding in the trigeminal ganglion and sphenopalatine ganglion but not within the central nervous system suggesting a peripheral site of action. The pharmacological approach using a specific PAC1 receptor antibody could provide a novel mechanism with a potential clinical efficacy in the treatment of primary headaches.


Migraine Disorders , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Animals , Headache/chemically induced , Headache/drug therapy , Humans , Nociception , Pituitary Adenylate Cyclase-Activating Polypeptide/therapeutic use , Rats
2.
PLoS One ; 13(5): e0196791, 2018.
Article En | MEDLINE | ID: mdl-29723257

Identification of voltage-gated sodium channel NaV1.7 inhibitors for chronic pain therapeutic development is an area of vigorous pursuit. In an effort to identify more potent leads compared to our previously reported GpTx-1 peptide series, electrophysiology screening of fractionated tarantula venom discovered the NaV1.7 inhibitory peptide JzTx-V from the Chinese earth tiger tarantula Chilobrachys jingzhao. The parent peptide displayed nominal selectivity over the skeletal muscle NaV1.4 channel. Attribute-based positional scan analoging identified a key Ile28Glu mutation that improved NaV1.4 selectivity over 100-fold, and further optimization yielded the potent and selective peptide leads AM-8145 and AM-0422. NMR analyses revealed that the Ile28Glu substitution changed peptide conformation, pointing to a structural rationale for the selectivity gains. AM-8145 and AM-0422 as well as GpTx-1 and HwTx-IV competed for ProTx-II binding in HEK293 cells expressing human NaV1.7, suggesting that these NaV1.7 inhibitory peptides interact with a similar binding site. AM-8145 potently blocked native tetrodotoxin-sensitive (TTX-S) channels in mouse dorsal root ganglia (DRG) neurons, exhibited 30- to 120-fold selectivity over other human TTX-S channels and exhibited over 1,000-fold selectivity over other human tetrodotoxin-resistant (TTX-R) channels. Leveraging NaV1.7-NaV1.5 chimeras containing various voltage-sensor and pore regions, AM-8145 mapped to the second voltage-sensor domain of NaV1.7. AM-0422, but not the inactive peptide analog AM-8374, dose-dependently blocked capsaicin-induced DRG neuron action potential firing using a multi-electrode array readout and mechanically-induced C-fiber spiking in a saphenous skin-nerve preparation. Collectively, AM-8145 and AM-0422 represent potent, new engineered NaV1.7 inhibitory peptides derived from the JzTx-V scaffold with improved NaV selectivity and biological activity in blocking action potential firing in both DRG neurons and C-fibers.


Analgesics/isolation & purification , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Peptides/chemistry , Sodium Channel Blockers/isolation & purification , Spider Venoms/chemistry , Action Potentials/drug effects , Amino Acid Substitution , Analgesics/pharmacology , Animals , Capsaicin/pharmacology , Cell Line , Drug Evaluation, Preclinical , Ganglia, Spinal/drug effects , Humans , Male , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Nerve Fibers, Unmyelinated/drug effects , Nuclear Magnetic Resonance, Biomolecular , Patch-Clamp Techniques , Physical Stimulation , Protein Engineering , Recombinant Proteins/drug effects , Sodium Channel Blockers/pharmacology , Structure-Activity Relationship , Tetrodotoxin/pharmacology
3.
J Pharmacol Exp Ther ; 362(1): 146-160, 2017 07.
Article En | MEDLINE | ID: mdl-28473457

Potent and selective antagonists of the voltage-gated sodium channel NaV1.7 represent a promising avenue for the development of new chronic pain therapies. We generated a small molecule atropisomer quinolone sulfonamide antagonist AMG8379 and a less active enantiomer AMG8380. Here we show that AMG8379 potently blocks human NaV1.7 channels with an IC50 of 8.5 nM and endogenous tetrodotoxin (TTX)-sensitive sodium channels in dorsal root ganglion (DRG) neurons with an IC50 of 3.1 nM in whole-cell patch clamp electrophysiology assays using a voltage protocol that interrogates channels in a partially inactivated state. AMG8379 was 100- to 1000-fold selective over other NaV family members, including NaV1.4 expressed in muscle and NaV1.5 expressed in the heart, as well as TTX-resistant NaV channels in DRG neurons. Using an ex vivo mouse skin-nerve preparation, AMG8379 blocked mechanically induced action potential firing in C-fibers in both a time-dependent and dose-dependent manner. AMG8379 similarly reduced the frequency of thermally induced C-fiber spiking, whereas AMG8380 affected neither mechanical nor thermal responses. In vivo target engagement of AMG8379 in mice was evaluated in multiple NaV1.7-dependent behavioral endpoints. AMG8379 dose-dependently inhibited intradermal histamine-induced scratching and intraplantar capsaicin-induced licking, and reversed UVB radiation skin burn-induced thermal hyperalgesia; notably, behavioral effects were not observed with AMG8380 at similar plasma exposure levels. AMG8379 is a potent and selective NaV1.7 inhibitor that blocks sodium current in heterologous cells as well as DRG neurons, inhibits action potential firing in peripheral nerve fibers, and exhibits pharmacodynamic effects in translatable models of both itch and pain.


NAV1.7 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neurons/drug effects , Pain/prevention & control , Pain/psychology , Patch-Clamp Techniques , Pruritus/prevention & control , Pruritus/psychology , Quinolones/pharmacology , Small Molecule Libraries , Stereoisomerism , Sulfonamides/pharmacology
4.
Mol Pain ; 122016.
Article En | MEDLINE | ID: mdl-27899696

The transient receptor potential ankyrin 1 (TRPA1) channel has been implicated in pathophysiological processes that include asthma, cough, and inflammatory pain. Agonists of TRPA1 such as mustard oil and its key component allyl isothiocyanate (AITC) cause pain and neurogenic inflammation in humans and rodents, and TRPA1 antagonists have been reported to be effective in rodent models of pain. In our pursuit of TRPA1 antagonists as potential therapeutics, we generated AMG0902, a potent (IC90 of 300 nM against rat TRPA1), selective, brain penetrant (brain to plasma ratio of 0.2), and orally bioavailable small molecule TRPA1 antagonist. AMG0902 reduced mechanically evoked C-fiber action potential firing in a skin-nerve preparation from mice previously injected with complete Freund's adjuvant, supporting the role of TRPA1 in inflammatory mechanosensation. In vivo target coverage of TRPA1 by AMG0902 was demonstrated by the prevention of AITC-induced flinching/licking in rats. However, oral administration of AMG0902 to rats resulted in little to no efficacy in models of inflammatory, mechanically evoked hypersensitivity; and no efficacy was observed in a neuropathic pain model. Unbound plasma concentrations achieved in pain models were about 4-fold higher than the IC90 concentration in the AITC target coverage model, suggesting that either greater target coverage is required for efficacy in the pain models studied or TRPA1 may not contribute significantly to the underlying mechanisms.


Hyperalgesia/metabolism , Inflammation/complications , Sciatica/complications , TRPC Cation Channels/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Amines/therapeutic use , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , CHO Cells , Cricetulus , Cyclohexanecarboxylic Acids/therapeutic use , Exploratory Behavior/drug effects , Freund's Adjuvant/toxicity , Gabapentin , Hyperalgesia/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Male , Mice, Inbred C57BL , Mice, Knockout , Naproxen/pharmacology , Nerve Fibers, Unmyelinated/drug effects , Nerve Fibers, Unmyelinated/physiology , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Sciatica/drug therapy , TRPA1 Cation Channel , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , gamma-Aminobutyric Acid/therapeutic use
5.
Int J Neurosci ; 126(2): 182-92, 2016.
Article En | MEDLINE | ID: mdl-25562420

The nine members of the voltage-gated sodium channel (Nav) family mediate inward sodium currents that depolarize neurons and lead to action potential firing. Increased Nav expression and function in sensory ganglia may drive ectopic action potentials and result in neuropathic pain. Using patch-clamp electrophysiology and molecular biology techniques, experiments were performed to elucidate the contribution of Nav channels to sodium currents in rat dorsal root ganglion (DRG) neurons following the L5/L6 spinal nerve ligation (SNL) model of neuropathic pain. The abundance of DRG neurons with fast, tetrodotoxin sensitive (TTX-S) currents was seven-fold higher whereas the abundance of DRG neurons with slow, tetrodotoxin resistant (TTX-R) currents was nearly thirty-fold lower when comparing ipsilateral (injured) to contralateral (uninjured) neurons. TTX-S currents were elevated in larger neurons while TTX-R currents were reduced in both small and large neurons. Among Nav transcripts encoding TTX-R channels, Scn10a (Nav1.8) and Scn11a (Nav1.9) expression was twenty- to thirty-fold lower, while among Nav transcripts encoding TTX-S channels, Scn3a (Nav1.3) expression was four-fold higher in injured compared to uninjured DRG by qRT-PCR analysis. In summary, the SNL model of neuropathic pain induced a phenotypic switch in Nav expression from TTX-R to TTX-S channels in injured DRG neurons. Transcriptional reprogramming of Nav genes may drive ectopic action potential firing and contribute to neuropathic pain.


Functional Laterality/physiology , Ganglia, Spinal/pathology , Neurons/physiology , Peripheral Nerve Injuries/pathology , Voltage-Gated Sodium Channels/metabolism , Animals , Biophysical Phenomena/drug effects , Electric Stimulation , Gene Expression Regulation/drug effects , Hyperalgesia/etiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Patch-Clamp Techniques , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Voltage-Gated Sodium Channels/genetics
6.
PLoS One ; 9(9): e105895, 2014.
Article En | MEDLINE | ID: mdl-25188265

Clinical genetic studies have shown that loss of Nav1.7 function leads to the complete loss of acute pain perception. The global deletion is reported lethal in mice, however, and studies of mice with promoter-specific deletions of Nav1.7 have suggested that the role of Nav1.7 in pain transduction depends on the precise form of pain. We developed genetic and animal husbandry strategies that overcame the neonatal-lethal phenotype and enabled construction of a global Nav1.7 knockout mouse. Knockouts were anatomically normal, reached adulthood, and had phenotype wholly analogous to human congenital indifference to pain (CIP): compared to littermates, knockouts showed no defects in mechanical sensitivity or overall movement yet were completely insensitive to painful tactile, thermal, and chemical stimuli and were anosmic. Knockouts also showed no painful behaviors resulting from peripheral injection of nonselective sodium channel activators, did not develop complete Freund's adjuvant-induced thermal hyperalgesia, and were insensitive to intra-dermal histamine injection. Tetrodotoxin-sensitive sodium current recorded from cell bodies of isolated sensory neurons and the mechanically-evoked spiking of C-fibers in a skin-nerve preparation each were reduced but not eliminated in tissue from knockouts compared to littermates. Results support a role for Nav1.7 that is conserved between rodents and humans and suggest several possibly translatable biomarkers for the study of Nav1.7-targeted therapeutics. Results further suggest that Nav1.7 may retain its key role in persistent as well as acute forms of pain.


NAV1.7 Voltage-Gated Sodium Channel/deficiency , Pain Insensitivity, Congenital/etiology , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/physiology , Nerve Fibers, Unmyelinated/physiology , Nervous System/pathology , Nervous System/physiopathology , Olfaction Disorders/genetics , Olfaction Disorders/physiopathology , Pain Insensitivity, Congenital/genetics , Pain Insensitivity, Congenital/physiopathology , Pain Threshold/physiology , Phenotype , Sensory Receptor Cells/physiology
7.
J Am Chem Soc ; 133(40): 15810-3, 2011 Oct 12.
Article En | MEDLINE | ID: mdl-21913658

When upconversion nanophosphors were incorporated into an azotolane-containing cross-linked liquid-crystal polymer film, the resulting composite film generated fast bending upon exposure to continuous-wave near-IR light at 980 nm. This occurs because the upconversion luminescence of the nanophosphors leads to trans-cis photoisomerization of the azotolane units and an alignment change of the mesogens. The bent film completely reverted to the initial flat state after the light source was removed.

8.
Bioorg Med Chem Lett ; 20(12): 3573-8, 2010 Jun 15.
Article En | MEDLINE | ID: mdl-20493696

The membrane bound large-conductance, calcium-activated potassium channel (BKCa) is an important regulator of neuronal activity. Here we describe the identification and structure-activity relationship of a novel class of potent tetrahydroquinoline BKCa agonists. An example from this class of BKCa agonists was shown to depress the spontaneous neuronal discharges in an electrophysiological model of migraine.


Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Neurons/drug effects , Quinolines/chemistry , Quinolines/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Electrophysiological Phenomena/drug effects , Humans , Migraine Disorders/drug therapy , Migraine Disorders/pathology , Models, Biological , Rats , Structure-Activity Relationship , Trigeminal Nuclei/cytology
9.
Mol Pharmacol ; 64(4): 798-810, 2003 Oct.
Article En | MEDLINE | ID: mdl-14500736

In the present study, we describe the characterization of a positive allosteric modulator at metabotropic glutamate subtype 2 receptors (mGluR2). N-(4-(2-Methoxyphenoxy)-phenyl-N-(2,2,2-trifluoroethylsulfonyl)-pyrid-3-ylmethylamine (LY487379) is a selective positive allosteric modulator at human mGluR2 and is without activity at human mGluR3. Furthermore, LY487379 has no intrinsic agonist or antagonist activity at hmGluR2, as determined by functional guanosine 5'(gamma-[35S]thio)triphosphate ([35S]GTPgammaS) binding, single-cell Ca2+ imaging, and electrophysiological studies. However, LY487379 markedly potentiated glutamate-stimulated [35S]GTPgammaS binding in a concentration-dependent manner at hmGluR2, shifting the glutamate dose-response curve leftward by 3-fold and increasing the maximum levels of [35S]GTPgammaS stimulation. This effect of LY487479 was also observed to a greater extent on the concentration-response curves to selective hmGluR2/3 agonists. In radioligand binding studies to rat cortical membranes, LY487379 increased the affinity of the radiolabeled agonist, [3H]DCG-IV, without affecting the binding affinity of the radiolabeled antagonist, [3H]LY341495. In rat hippocampal slices, coapplication of LY487379 potentiated synaptically evoked mGluR2 responses. Finally, to elucidate the site of action, we systematically exchanged segments and single amino acids between hmGluR2 and hmGluR3. Substitution of Ser688 and/or Gly689 in transmembrane IV along with Asn735 located in transmembrane segment V, with the homologous amino acids of hmGluR3, completely eliminated LY487379 allosteric modulation of hmGluR2. We propose that this allosteric binding site defines a pocket that is different from the orthosteric site located in the amino terminal domain.


Glycine/analogs & derivatives , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Sulfonamides/pharmacology , Allosteric Regulation , Amino Acid Sequence , Amino Acids/pharmacology , Animals , Binding Sites , CHO Cells , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cerebral Cortex/cytology , Cricetinae , Cyclopropanes/pharmacology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glycine/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Molecular Sequence Data , Perforant Pathway/drug effects , Perforant Pathway/metabolism , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Tritium , Xanthenes/pharmacology
...