Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Pharmacol Sci ; 152(2): 86-89, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37169483

ABSTRACT

Cav3.2, a T-type calcium channel (T-channel) family member, is expressed in the nociceptors and spinal cord, and its activity is largely suppressed by zinc under physiological conditions. In rats, intrathecal and intraplantar administration of a zinc chelator, TPEN, caused T-channel-dependent mechanical hyperalgesia, and the intraplantar, but not intrathecal, TPEN induced Cav3.2 upregulation in the dorsal root ganglion. In mice, intraplantar TPEN also caused mechanical allodynia, which was abolished by T-channel inhibitors or Cav3.2 gene deletion. Together, spinal and peripheral zinc deficiency appears to enhance Cav3.2 activity in the spinal postsynaptic neurons and nociceptors, respectively, thereby promoting pain.


Subject(s)
Calcium Channels, T-Type , Hyperalgesia , Rats , Mice , Animals , Hyperalgesia/chemically induced , Rodentia , Chelating Agents , Zinc , Calcium Channels, T-Type/genetics , Ganglia, Spinal
2.
Biol Pharm Bull ; 46(12): 1699-1705, 2023.
Article in English | MEDLINE | ID: mdl-38044093

ABSTRACT

Community pharmacists may play a key role in promoting deprescribing of potential inappropriate medications (PIMs) that are highly prevalent among community-dwelling elderly with dementia. To characterize PIMs categories that need a special attention for dementia patients, in the present study, we analyzed the anonymized pharmacy claims data of patients aged 65 years and older (n = 333869) who visited nationwide 905 community-based pharmacies of Sugi Pharmacy Co., Ltd. during December 1-31, 2019. A dementia group was defined as patients who received typical dementia medications marketed in Japan, i.e., donepezil, galantamine, memantine or rivastigmine, and a non-dementia group was defined as patients who received no such medications. After propensity score matching on the basis of patients' age, gender and home healthcare insurance usage, the data of 11486 patients in each group were subjected to logistic regression analyses, to identify PIMs categories particularly important for dementia patients. Univariate analysis indicated that the proportions of dementia patients who received 1 and 2≤ of PIMs were significantly (p < 0.001) greater than those of non-dementia patients (odds ratios were 1.35 and 1.47, respectively). Multivariate analyses identified 5 categories of PIMs that were significantly more frequently prescribed in dementia patients, i.e., 'H2 blockers,' 'drugs for overactive bladder,' 'anti-diabetes drugs' and 'sulpiride' listed as PIMs categories for non-specific cases (adjusted odds ratios (aORs): 1.29, 1.91, 1.17, and 1.38, respectively), in addition to 'antipsychotics' listed only for dementia patients (aOR: 4.29). These results provide useful information to establish strategies for pharmacist-led deprescribing of PIMs in dementia patients.


Subject(s)
Dementia , Pharmacies , Pharmacy , Aged , Humans , Potentially Inappropriate Medication List , Inappropriate Prescribing , Dementia/drug therapy
3.
J Pharmacol Sci ; 140(3): 310-312, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31492577

ABSTRACT

We tested whether genetic deletion of Cav3.2 T-type Ca2+ channels abolishes hydrogen sulfide (H2S)-mediated pain signals in mice. In Cav3.2-expressing HEK293 cells, Na2S, an H2S donor, at 100 µM clearly increased Ba2+ currents, as assessed by whole-cell patch-clamp recordings. In wild-type C57BL/6 mice, intraplantar and intracolonic administration of Na2S evoked mechanical allodynia and visceral nociceptive behavior, respectively, which were abolished by TTA-A2, a T-type Ca2+ channel blocker. In Cav3.2-knockout mice of a C57BL/6 background, Na2S caused neither somatic allodynia nor colonic nociception. Our study thus provides definitive evidence for an essential role of Cav3.2 in H2S-dependent somatic and colonic pain.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Hydrogen Sulfide/pharmacology , Nociceptive Pain/metabolism , Signal Transduction/drug effects , Visceral Pain/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , TRPA1 Cation Channel/metabolism
4.
Bioorg Med Chem ; 26(15): 4410-4427, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30031654

ABSTRACT

Since 6-prenylnaringenin (6-PNG) was recently identified as a novel T-type calcium channel blocker with the IC50 value around 1 µM, a series of flavanone derivatives were designed, synthesized and subsequently evaluated for T-channel-blocking activity in HEK293 cells transfected with Cav3.2 T-type channels using a patch-clamp technique. As a result, several new flavanones blocked Cav3.2-dependent T-currents more potently than 6-PNG. In the synthesized compounds, 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(2-hydroxyphenyl)chroman-4-one 8j, 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 11b, 6-(2-cyclopentylideneethyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 11d, and 6-(2-Cyclopentylethyl)-5,7-dihydroxy-2-(4-hydroxyphenyl)chroman-4-one 12c were more potent blocker than 6-PNG with the IC50 value of 0.39, 0.26, 0.46, and 0.50 µM, respectively. Among the above four derivatives, the compound 8j provided the best result in the in vivo experiments; i.e. systemic administration of 8j at the minimum dose completely restored neuropathic pain induced by partial sciatic nerve ligation in mice.


Subject(s)
Analgesics/chemical synthesis , Calcium Channel Blockers/chemical synthesis , Calcium Channels, T-Type/chemistry , Drug Design , Flavonoids/chemistry , Action Potentials/drug effects , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/metabolism , Disease Models, Animal , Flavonoids/pharmacology , Flavonoids/therapeutic use , HEK293 Cells , Humans , Inhibitory Concentration 50 , Male , Mice , Neuralgia/drug therapy , Patch-Clamp Techniques , Structure-Activity Relationship
5.
Redox Biol ; 59: 102579, 2023 02.
Article in English | MEDLINE | ID: mdl-36563535

ABSTRACT

Poly-trans-[(2-carboxyethyl)germasesquioxane] (Ge-132), an organogermanium, is hydrolyzed to 3-(trihydroxygermyl)propanoic acid (THGP) in aqueous solutions, and reduces inflammation, pain and cancer, whereas the underlying mechanisms remain unknown. Sulfides including H2S, a gasotransmitter, generated from l-cysteine by some enzymes including cystathionine-γ-lyase (CSE), are pro-nociceptive, since they enhance Cav3.2 T-type Ca2+ channel activity expressed in the primary afferents, most probably by canceling the channel inhibition by Zn2+ linked via coordinate bonding to His191 of Cav3.2. Given that germanium is reactive to sulfur, we tested whether THGP would directly trap sulfide, and inhibit sulfide-induced enhancement of Cav3.2 activity and sulfide-dependent pain in mice. Using mass spectrometry and 1H NMR techniques, we demonstrated that THGP directly reacted with sulfides including Na2S and NaSH, and formed a sulfur-containing reaction product, which decreased in the presence of ZnCl2. In Cav3.2-transfected HEK293 cells, THGP inhibited the sulfide-induced enhancement of T-type Ca2+ channel-dependent membrane currents. In mice, THGP, administered systemically or locally, inhibited the mechanical allodynia caused by intraplantar Na2S. In the mice with cyclophosphamide-induced cystitis and cerulein-induced pancreatitis, which exhibited upregulation of CSE in the bladder and pancreas, respectively, systemic administration of THGP as well as a selective T-type Ca2+ channel inhibitor suppressed the cystitis-related and pancreatitis-related visceral pain. These data suggest that THGP traps sulfide and inhibits sulfide-induced enhancement of Cav3.2 activity, leading to suppression of Cav3.2-dependent pain caused by sulfide applied exogenously and generated endogenously.


Subject(s)
Calcium Channels, T-Type , Cystitis , Hydrogen Sulfide , Pancreatitis , Visceral Pain , Mice , Humans , Animals , HEK293 Cells , Calcium Channels, T-Type/physiology , Sulfides/pharmacology , Cystitis/chemically induced , Hydrogen Sulfide/metabolism
6.
Eur J Med Chem ; 243: 114716, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36075145

ABSTRACT

T-type Ca2+ channels (T-channels), particularly Cav3.2 and Cav3.1 isoforms, are promising targets for treating various diseases including intractable pain. Given the potent inhibitory activity of pimozide, an antipsychotic, against T-channels, we conducted structure-activity relationship studies of pimozide derivatives, and identified several compounds including 3a, 3s, and 4 that had potency comparable to that of pimozide in inhibiting T-channels, but little binding affinity to dopamine D2 receptors. The introduction of a phenylbutyl group on the benzoimidazole nuclei of pimozide was considered a key structural modification to reduce the binding affinity to D2 receptors. Those pimozide derivatives potently suppressed T-channel-dependent somatic and visceral pain in mice, without causing any motor dysfunctions attributable to D2 receptor blockade, including catalepsy. The present study thus provides an avenue to develop novel selective T-channel inhibitors available for pain management via the structural modification of existing medicines.


Subject(s)
Calcium Channels, T-Type , Visceral Pain , Mice , Animals , Pimozide/pharmacology , Pimozide/therapeutic use , Calcium Channels, T-Type/metabolism , Visceral Pain/drug therapy , Dopamine , Calcium Channel Blockers/chemistry , Receptors, Dopamine/metabolism
7.
Eur J Pharmacol ; 888: 173587, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32971090

ABSTRACT

Overexpression of Cav3.2 T-type Ca2+ channels in L4 dorsal root ganglion (DRG) participates in neuropathic pain after L5 spinal nerve cutting (L5SNC) in rats. The L5SNC-induced neuropathic pain also involves high mobility group box 1 (HMGB1), a damage-associated molecular pattern protein, and its target, the receptor for advanced glycation end-products (RAGE). We thus studied the molecular mechanisms for the L5SNC-induced Cav3.2 overexpression as well as neuropathic pain in rats by focusing on; 1) possible involvement of early growth response 1 (Egr-1), known to regulate transcriptional expression of Cav3.2, and ubiquitin-specific protease 5 (USP5) that protects Cav3.2 from proteasomal degradation, and 2) possible role of HMGB1/RAGE as an upstream signal. Protein levels of Cav3.2 as well as Egr-1 in L4 DRG significantly increased in the early (day 6) and persistent (day 14) phases of neuropathy after L5SNC, while USP5 protein in L4 DRG did not increase on day 6, but day 14. An anti-HMGB1-neutralizing antibody or a low molecular weight heparin, a RAGE antagonist, prevented the development of neuropathic pain and upregulation of Egr-1 and Cav3.2 in L4 DRG after L5SNC. L5SNC increased macrophages accumulating in the sciatic nerves, and the cytoplasm/nuclear ratio of immunoreactive HMGB1 in those macrophages. Our findings suggest that L5SNC-induced Cav3.2 overexpression in L4 DRG and neuropathic pain involves Egr-1 upregulation downstream of the macrophage-derived HMGB1/RAGE pathway, and that the delayed upregulation of USP5 might contribute to the persistent Cav3.2 overexpression and neuropathy.


Subject(s)
Calcium Channels, T-Type/biosynthesis , Early Growth Response Protein 1/biosynthesis , Ganglia, Spinal/metabolism , HMGB1 Protein/biosynthesis , Neuralgia/metabolism , Ubiquitin-Specific Proteases/biosynthesis , Animals , Calcium Channels, T-Type/genetics , Early Growth Response Protein 1/genetics , Ganglia, Spinal/pathology , Gene Expression , HMGB1 Protein/genetics , Lumbar Vertebrae , Male , Neuralgia/genetics , Neuralgia/pathology , Rats , Rats, Wistar , Signal Transduction/physiology , Spinal Nerves/injuries , Spinal Nerves/metabolism , Spinal Nerves/pathology , Ubiquitin-Specific Proteases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL