173 results on '"Atsufumi Kawabata"'
Search Results
2. Role of neuron-derived ATP in paclitaxel-induced HMGB1 release from macrophages and peripheral neuropathy
- Author
-
Hiroki Yamanishi, Masahiro Nishibori, Riki Kamaguchi, Maho Tsubota, Dengli Wang, Risa Domoto, Atsufumi Kawabata, Maiko Iemura, and Fumiko Sekiguchi
- Subjects
Male ,Paclitaxel ,Mice, Inbred Strains ,chemical and pharmacologic phenomena ,Chemotherapy-induced peripheral neuropathy (CIPN) ,RM1-950 ,HMGB1 ,Neutralization ,Mice ,chemistry.chemical_compound ,Adenosine Triphosphate ,medicine ,Animals ,Macrophage depletion ,HMGB1 Protein ,Neuroimmune crosstalk ,Neurons ,Pharmacology ,biology ,Chemistry ,Macrophages ,Peripheral Nervous System Diseases ,Receptor Cross-Talk ,medicine.disease ,Blockade ,Cell biology ,ATP ,RAW 264.7 Cells ,Peripheral neuropathy ,High-mobility group ,medicine.anatomical_structure ,biology.protein ,Molecular Medicine ,Therapeutics. Pharmacology ,Receptors, Purinergic P2X7 ,Neuron ,High mobility group box 1 (HMGB1) ,Receptors, Purinergic P2X4 - Abstract
We examined the role of ATP and high mobility group box 1 (HMGB1) in paclitaxel-induced peripheral neuropathy (PIPN). PIPN in mice was prevented by HMGB1 neutralization, macrophage depletion, and P2X7 or P2X4 blockade. Paclitaxel and ATP synergistically released HMGB1 from macrophage-like RAW264.7 cells, but not neuron-like NG108-15 cells. The paclitaxel-induced HMGB1 release from RAW264.7 cells was accelerated by co-culture with NG108-15 cells in a manner dependent on P2X7 or P2X4. Paclitaxel released ATP from NG108-15 cells, but not RAW264.7 cells. Thus, PIPN is considered to involve acceleration of HMGB1 release from macrophages through P2X7 and P2X4 activation by neuron-derived ATP.
- Published
- 2022
3. Estrogen decline is a risk factor for paclitaxel-induced peripheral neuropathy: Clinical evidence supported by a preclinical study
- Author
-
Shigekatsu Hatanaka, Ayano Kanto, Masanori Fujitani, Atsufumi Kawabata, Hiroki Fukuyama, Maho Tsubota, Shiori Hiramoto, Fumiko Sekiguchi, Yuichi Koizumi, and Tomoyoshi Miyamoto
- Subjects
0301 basic medicine ,Oncology ,Thrombomodulin ,medicine.medical_treatment ,Mice ,chemistry.chemical_compound ,Breast cancer ,0302 clinical medicine ,Cancer Survivors ,Risk Factors ,Medicine ,Chemotherapy-induced peripheral neuropathy ,Aged, 80 and over ,Age Factors ,Peripheral Nervous System Diseases ,Middle Aged ,Postmenopause ,Paclitaxel ,Molecular Medicine ,Female ,medicine.medical_specialty ,medicine.drug_class ,Ovariectomy ,Breast Neoplasms ,Mice, Inbred Strains ,03 medical and health sciences ,Internal medicine ,Postmenopausal estrogen decline ,Animals ,Humans ,Risk factor ,Aged ,Retrospective Studies ,Pharmacology ,Chemotherapy ,business.industry ,lcsh:RM1-950 ,Cancer ,Estrogens ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Rats ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,Peripheral neuropathy ,chemistry ,Estrogen ,business ,030217 neurology & neurosurgery - Abstract
We performed clinical retrospective study in female cancer patients and fundamental experiments in mice, in order to clarify risk factors for paclitaxel-induced peripheral neuropathy (PIPN). In the clinical study, 131 of 189 female outpatients with cancer undergoing paclitaxel-based chemotherapy met inclusion criteria. Breast cancer survivors (n = 40) showed significantly higher overall PIPN (grades 1–4) incidence than non-breast cancer survivors (n = 91). Multivariate sub-analyses of breast cancer survivors showed that 57 years of age or older and endocrine therapy before paclitaxel treatment were significantly associated with severe PIPN (grades 2–4). The age limit was also significantly correlated with overall development of severe PIPN. In the preclinical study, female mice subjected to ovariectomy received repeated administration of paclitaxel, and mechanical nociceptive threshold was assessed by von Frey test. Ovariectomy aggravated PIPN in the mice, an effect prevented by repeated treatment with 17β-estradiol. Repeated administration of thrombomodulin alfa (TMα), known to prevent chemotherapy-induced peripheral neuropathy in rats and mice, also prevented the development of PIPN in the ovariectomized mice. Collectively, breast cancer survivors, particularly with postmenopausal estrogen decline and/or undergoing endocrine therapy, are considered a PIPN-prone subpopulation, and may require non-hormonal pharmacological intervention for PIPN in which TMα may serve as a major candidate.
- Published
- 2021
4. Effects of Bepridil and Pimozide, Existing Medicines Capable of Blocking T-Type Ca2+ Channels, on Visceral Pain in Mice
- Author
-
Fumiko Sekiguchi, Maho Tsubota, Atsufumi Kawabata, Saaya Fukushi, Kazuki Matsui, and Kyoko Okazaki
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,Pharmaceutical Science ,Visceral pain ,General Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pimozide ,Nociception ,030220 oncology & carcinogenesis ,Bepridil ,Hyperalgesia ,medicine ,Nociception assay ,Intractable pain ,medicine.symptom ,Bladder Pain ,business ,medicine.drug - Abstract
T-Type Ca2+ channels (T-channels), particularly Cav3.2, are now considered as therapeutic targets for treatment of intractable pain including visceral pain. Among existing medicines, bepridil, a multi-channel blocker, used for treatment of arrhythmia and angina, and pimozide, a dopamine D2 receptor antagonist, known as a typical antipsychotic, have potent T-channel blocking activity. We thus tested whether bepridil and pimozide could suppress visceral pain in mice. Colonic and bladder pain were induced by intracolonic administration of 2,4,6-trinitrobenzene sulfonic acid (TNBS) and systemic administration of cyclophosphamide (CPA), respectively. Referred hyperalgesia was assessed by von Frey test, and colonic hypersensitivity to distension by a volume load with intracolonic water injection and spontaneous bladder pain were evaluated by observing nociceptive behaviors in conscious mice. The mice exhibited referred hyperalgesia and colonic hypersensitivity to distension on day 6 after TNBS treatment. Systemic administration of bepridil at 10-20 mg/kg or pimozide at 0.1-0.5 mg/kg strongly reduced the referred hyperalgesia on the TNBS-induced referred hyperalgesia and colonic hypersensitivity to distension. CPA treatment caused bladder pain-like nociceptive behavior and referred hyperalgesia, which were reversed by bepridil at 10-20 mg/kg or pimozide at 0.5-1 mg/kg. Our data thus suggest that bepridil and pimozide, existing medicines capable of blocking T-channels, are useful for treatment of colonic and bladder pain, and serve as seeds for the development of new medicines for visceral pain treatment.
- Published
- 2021
5. Relationship between serum bepridil concentration and corrected QT interval
- Author
-
Nobue Terakawa, Kyoichi Wada, Kazuki Matsui, Mitsutaka Takada, Kota Sakakura, Kengo Kusano, Kouichi Hosomi, Naoki Hayakawa, Atsufumi Kawabata, Satoshi Yokoyama, Yutaro Mukai, and Tsutomu Nakamura
- Subjects
medicine.medical_specialty ,Bepridil ,Prolonged QT ,QT interval ,Electrocardiography ,Japan ,Torsades de Pointes ,Internal medicine ,medicine ,Humans ,Pharmacology (medical) ,In patient ,cardiovascular diseases ,Risk factor ,Retrospective Studies ,Pharmacology ,business.industry ,Significant difference ,Corrected qt ,Atrial fibrillation ,medicine.disease ,Long QT Syndrome ,Cardiology ,business ,medicine.drug - Abstract
OBJECTIVE Bepridil prolongs the QT interval and can induce torsade de pointes. Although increased bepridil concentration may be a primary cause of prolonged QT, the relationship between serum bepridil concentration and prolonged QT remains unclear. We investigated the relationship between serum bepridil concentration and the corrected QT (QTc) interval in patients treated with bepridil. MATERIALS AND METHODS A retrospective study was performed at the National Cerebral and Cardiovascular Center in Japan. Patients with atrial fibrillation who were treated with bepridil from January 2014 to December 2015 were enrolled in the study. Serum bepridil concentrations and electrocardiogram data collected more than 21 days after the initiation of bepridil were used for analysis. RESULTS A total of 60 patients were included in this study. There was a significant difference in mean QTc interval before and after initiation of bepridil (p < 0.0001). A significant relationship was observed between bepridil dose (p = 0.014) or serum bepridil concentration (p
- Published
- 2021
6. Tacrolimus, a calcineurin inhibitor, promotes capsaicin-induced colonic pain in mice
- Author
-
Yuka Terada, Kazuki Matsui, Fumiko Sekiguchi, Atsufumi Kawabata, and Maho Tsubota
- Subjects
0301 basic medicine ,Colon ,Calcineurin Inhibitors ,TRPV1 ,TRPV Cation Channels ,chemical and pharmacologic phenomena ,Pharmacology ,Tacrolimus ,Irritable Bowel Syndrome ,Intracolonic ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,business.industry ,lcsh:RM1-950 ,Calcineurin ,lcsh:Therapeutics. Pharmacology ,surgical procedures, operative ,030104 developmental biology ,nervous system ,chemistry ,Hyperalgesia ,Capsaicin ,Nociceptor ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Capsazepine ,business ,Immunosuppressive Agents ,030217 neurology & neurosurgery - Abstract
TRPV1 is phosphorylated and functionally upregulated by protein kinases, and negatively regulated by phosphatases including calcineurin. Since the clinical use of calcineurin-inhibiting immunosuppressants is commonly associated with chronic diarrhea, we examined if tacrolimus, a calcineurin inhibitor, promotes TRPV1-dependent colonic hypersensitivity in mice. Intracolonic administration of capsaicin, a TRPV1 agonist, caused referred hyperalgesia in the lower abdomen, an effect prevented by capsazepine, a TRPV1 blocker. Tacrolimus accelerated the intracolonic capsaicin-induced referred hyperalgesia. Similarly, intracolonic capsaicin caused spinal ERK phosphorylation, a marker for nociceptor excitation, an effect promoted by tacrolimus. Thus, tacrolimus may aggravate TRPV1-related colonic pain accompanying irritable bowel syndrome. Keywords: Tacrolimus, Colonic pain, TRPV1
- Published
- 2020
7. Discovery of pimozide derivatives as novel T-type calcium channel inhibitors with little binding affinity to dopamine D2 receptors for treatment of somatic and visceral pain
- Author
-
Yoshihito Kasanami, Chihiro Ishikawa, Takahiro Kino, Momoka Chonan, Naoki Toyooka, Yasuhiro Takashima, Yuriko Iba, Fumiko Sekiguchi, Maho Tsubota, Tsuyako Ohkubo, Shigeru Yoshida, Atsushi Kawase, Takuya Okada, and Atsufumi Kawabata
- Subjects
Pharmacology ,Organic Chemistry ,Drug Discovery ,General Medicine - Published
- 2022
8. Development of hepatic impairment aggravates chemotherapy-induced peripheral neuropathy following oxaliplatin treatment: Evidence from clinical and preclinical studies
- Author
-
Tomoyoshi Miyamoto, Risa Domoto, Fumiko Sekiguchi, Riki Kamaguchi, Rika Nishimura, Misato Matsuno, Maho Tsubota, Masanori Fujitani, Shigekatsu Hatanaka, Yuichi Koizumi, Dengli Wang, Masahiro Nishibori, and Atsufumi Kawabata
- Subjects
Adult ,Male ,Thrombomodulin alfa ,Hepatic injury ,Antineoplastic Agents ,Mice, Inbred Strains ,Chemotherapy-induced peripheral neuropathy (CIPN) ,RM1-950 ,Severity of Illness Index ,Animals ,Humans ,HMGB1 Protein ,Cells, Cultured ,Aged ,Retrospective Studies ,Pharmacology ,Aged, 80 and over ,Peripheral Nervous System Diseases ,Middle Aged ,digestive system diseases ,Oxaliplatin ,Disease Progression ,Molecular Medicine ,Female ,Therapeutics. Pharmacology ,Chemical and Drug Induced Liver Injury ,High mobility group box 1 (HMGB1) - Abstract
Oxaliplatin often induces peripheral neuropathy, a dose-limiting adverse reaction, and in rare cases leads to sinusoidal obstruction syndrome. We thus conducted a retrospective cohort study to examine the relationship between oxaliplatin-induced peripheral neuropathy (OIPN) and hepatic impairment, and then perform a fundamental study to analyze the underlying mechanisms. Analysis of medical records in cancer patients treated with oxaliplatin indicated that laboratory test parameters of hepatic impairment including AST, ALT and APRI (AST to platelet ratio index) moderately increased during oxaliplatin treatment, which was positively correlated with the severity of OIPN (grades 1–4), and associated with later incidence of survivors with OIPN grades ≥2. In mice, hepatic injury induced by CCl4 or ethanol accelerated OIPN in mice, an effect prevented by inactivation of high mobility group box 1 (HMGB1), known to participate in OIPN, by the neutralizing antibody or thrombomodulin alfa capable of promoting its thrombin-dependent degradation. Oxaliplatin also aggravated the hepatic injury in mice. CCl4 released HMGB1 from cultured hepatic parenchymal cells, and oxaliplatin at clinically achievable concentrations released HMGB1 from hepatic parenchymal and non-parenchymal cells. Our clinical and preclinical data suggest that the development of mild hepatic impairment during oxaliplatin treatment is associated with later aggravation of OIPN.
- Published
- 2021
9. NNC 55-0396, a T-type calcium channel blocker, protects against the brain injury induced by middle cerebral artery occlusion and reperfusion in mice
- Author
-
Atsufumi Kawabata, Sachi Matsuda, Shogo Tokuyama, Hiroyuki Nishikawa, Fumiko Sekiguchi, Maho Tsubota, Anna Fukatsu, and Yuko Kurokawa
- Subjects
0301 basic medicine ,Cyclopropanes ,Male ,medicine.medical_specialty ,Time Factors ,Ischemia ,Mice, Inbred Strains ,Brain damage ,Naphthalenes ,03 medical and health sciences ,Calcium Channels, T-Type ,0302 clinical medicine ,Neuronal damage ,Internal medicine ,Occlusion ,Medicine ,Animals ,Infusions, Parenteral ,Middle cerebral artery occlusion ,cardiovascular diseases ,Pharmacology ,business.industry ,Calcium channel ,lcsh:RM1-950 ,T-type calcium channel ,Infarction, Middle Cerebral Artery ,medicine.disease ,Calcium Channel Blockers ,030104 developmental biology ,Infusions, Intraventricular ,lcsh:Therapeutics. Pharmacology ,Blood-Brain Barrier ,Brain Injuries ,Reperfusion Injury ,Systemic administration ,Cardiology ,Molecular Medicine ,Benzimidazoles ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
We tested whether NNC 55-0396 (NNC), a T-type calcium channel (T-channel) blocker, reduces the brain injury caused by middle cerebral artery occlusion and reperfusion (MCAO/R) in mice. NNC, administered i.c.v. before the occlusion, greatly reduced the MCAO/R-induced brain infarct and neurological dysfunctions, although it, given toward the end of occlusion, was less effective. Systemic administration of NNC before the occlusion also attenuated the infarct and neurological dysfunctions. Our data imply that blood-brain-barrier-permeable T-channel blockers such as NNC are capable of reducing MCAO/R-induced brain damage, and that T-channels are involved in neuronal damage induced by ischemia rather than reperfusion. Keywords: T-type calcium channel, Middle cerebral artery occlusion, Ischemia-reperfusion
- Published
- 2019
10. Critical role of Cav3.2 T-type calcium channels in the peripheral neuropathy induced by bortezomib, a proteasome-inhibiting chemotherapeutic agent, in mice
- Author
-
Takuya Okada, Naoki Toyooka, Tomoyo Deguchi, Shiori Tomita, Takaya Miyazaki, Yuya Ikeda, Maho Tsubota, Fumiko Sekiguchi, Shigeru Yoshida, Atsufumi Kawabata, and Huy Du Nguyen
- Subjects
0301 basic medicine ,Voltage-dependent calcium channel ,Chemistry ,Bortezomib ,Calcium channel ,T-type calcium channel ,Pharmacology ,Toxicology ,Ascorbic acid ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Proteasome ,Chemotherapy-induced peripheral neuropathy ,medicine ,Proteasome inhibitor ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Bortezomib, a first-line agent for treatment of multiple myeloma, exhibits anticancer activity through proteasome inhibition. However, bortezomib-induced peripheral neuropathy (BIPN) is one of the most serious side effects. Since decreased proteasomal degradation of Cav3.2 T-type calcium channels in the primary afferents is involved in persistent pain, we investigated whether BIPN involves increased protein levels of Cav3.2 in mice. Six repeated i.p. administrations of bortezomib for 12 days developed persistent mechanical allodynia. Systemic administration of novel T-type calcium channel blockers, (2R/S)-6-prenylnaringenin and KTt-45, and of TTA-A2, the well-known blocker, reversed the BIPN. Ascorbic acid, known to block Cav3.2, but not Cav3.1 or 3.3, and silencing of Cav3.2 gene also suppressed BIPN. Protein levels of Cav3.2 in the dorsal root ganglion (DRG) at L4-L6 levels increased throughout days 1-21 after the onset of bortezomib treatment. Protein levels of USP5, a deubiquitinating enzyme that specifically inhibits proteasomal degradation of Cav3.2, increased in DRG on days 3-21, but not day 1, in bortezomib-treated mice. In DRG-derived ND7/23 cells, bortezomib increased protein levels of Cav3.2 and T-channel-dependent currents, as assessed by a patch-clamp method, but did not upregulate expression of Cav3.2 mRNA or USP5 protein. MG-132, another proteasome inhibitor, also increased Cav3.2 protein levels in the cultured cells. Given the previous evidence for USP5 induction following nociceptor excitation, our data suggest that BIPN involves the increased protein levels of Cav3.2 in nociceptors through inhibition of proteasomal degradation of Cav3.2 by bortezomib itself and then by USP5 that is upregulated probably in an activity-dependent manner.
- Published
- 2019
11. Role of Cav3.2 T-type Ca2+ channels in prostate cancer cells
- Author
-
Atsufumi Kawabata and Fumiko Sekiguchi
- Subjects
Pharmacology ,Chemistry ,medicine.drug_class ,Cellular differentiation ,Cancer ,medicine.disease ,Androgen ,Androgen deprivation therapy ,Prostate cancer ,Downregulation and upregulation ,LNCaP ,Cancer cell ,medicine ,Cancer research - Abstract
Among voltage-gated Ca2+ channels, T-type Ca2+ channels, which are activated by low voltages, regulate neuronal excitability, spontaneous neurotransmitter release, hormone secretion, etc. and also participate in proliferation of distinct cancer cells. Among three isoforms of T-type Ca2+ channels, Cav3.2 is detectable in 100% of biopsy samples from prostate cancer patients. In general, prostate cancer cells are highly sensitive to androgen deprivation therapy, but often acquire hormone-therapy resistance. The androgen deprivation may trigger neuroendocrine (NE)-like differentiation of some prostate cancer cells. We have analyzed the expression and function of Cav3.2 in human prostate cancer LNCaP cells during NE-like differentiation. NE-like LNCaP cells overexpress Cav3.2 through the CREB/Egr-1 pathway and also cystathionine-γ-lyase (CSE), which generates H2S that enhances the channel activity of Cav3.2. H2S generated by upregulated CSE appears to enhance the activity of upregulated Cav3.2 after the differentiation. The enhanced Cav3.2 activity in NE-like cells may contribute to increased secretion of mitogenic factors essential for androgen-independent proliferation of surrounding prostate cancer cells. It is known that increased extracellular glucose levels enhance Cav3.2 activity through asparagine (N)-linked glycosylation of Cav3.2, which might contribute to diabetic neuropathy. We then found that high glucose accelerates the enhanced channel function and overexpression of Cav3.2 in NE-like LNCaP cells, which might be associated with clinical evidence for diabetes-related poor prognosis of prostate cancer and development of hormone therapy resistance. Thus, Cav3.2 is considered to play a role in the pathophysiology of prostate cancer, and may serve as a therapeutic target.
- Published
- 2019
12. Risk factors and pharmacotherapy for chemotherapy-induced peripheral neuropathy in paclitaxel-treated female cancer survivors: A retrospective study in Japan
- Author
-
Shiori Hiramoto, Hajime Asano, Tomoyoshi Miyamoto, Manabu Takegami, and Atsufumi Kawabata
- Subjects
Clinical Oncology ,Paclitaxel ,Genital Neoplasms, Female ,Science ,Cancer Treatment ,Antineoplastic Agents ,Breast Neoplasms ,Duloxetine Hydrochloride ,Cancer Chemotherapy ,Cancer Survivors ,Drug Therapy ,Japan ,Risk Factors ,Diagnostic Medicine ,Breast Tumors ,Breast Cancer ,Medicine and Health Sciences ,Cancer Detection and Diagnosis ,Humans ,Chemotherapy ,Chemotherapeutic Agents ,Aged ,Retrospective Studies ,Platinum ,Pharmacology ,Multidisciplinary ,Pharmaceutics ,Gynecologic Cancers ,Peripheral Nervous System Diseases ,Obstetrics and Gynecology ,Cancers and Neoplasms ,Drugs ,Middle Aged ,Chemistry ,Oncology ,Physical Sciences ,Women's Health ,Medicine ,Female ,Oncology Agents ,Clinical Medicine ,Research Article ,Chemical Elements - Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting adverse reaction in cancer patients treated with several cytotoxic anticancer agents including paclitaxel. Duloxetine, an antidepressant known as a serotonin-noradrenalin reuptake inhibitor, is the only agent that has moderate evidence for the use to treat painful CIPN. The present retrospective cohort study aimed to analyze risk factors for paclitaxel-induced peripheral neuropathy (PIPN), and investigate ongoing prescription drug use for PIPN in Japan. Female breast and gynecologic cancer patients who underwent paclitaxel-based chemotherapy at a single center in Japan between January 2016 and December 2019 were enrolled in this study. Patients’ information obtained from electronic medical records were statistically analyzed to test possible risk factors on PIPN diagnosis. Patients’ age, total paclitaxel dose, the history of female hormone-related diseases, hypertension and body mass index (BMI), but not additional platinum agents, were significantly associated with increased PIPN diagnosis. Drugs prescribed for PIPN included duloxetine, pregabalin, mecobalamin and Goshajinkigan, a polyherbal medicine, regardless of poor evidence for their effectiveness against CIPN, and were greatly different between breast and gynecologic cancer patients diagnosed with PIPN at the departments of Surgery and Gynecology, respectively. Thus, older age, greater total paclitaxel dose, the history of estrogen-related diseases, hypertension and BMI are considered risk factors for PIPN in paclitaxel-based chemotherapy of female cancer patients. It appears an urgent need to establish a guideline of evidence-based pharmacotherapy for PIPN.
- Published
- 2021
13. Cystitis-Related Bladder Pain Involves ATP-Dependent HMGB1 Release from Macrophages and Its Downstream H2S/Cav3.2 Signaling in Mice
- Author
-
Shiori Hiramoto, Maho Tsubota, Masahiro Nishibori, Atsufumi Kawabata, Huy Du Nguyen, Hidenori Wake, Aya Sakaegi, Kaoru Yamaguchi, Takuya Okada, Junichi Tanaka, Kyoko Okazaki, Naoki Toyooka, Yuki Toriyama, Hiroyasu Ishikura, and Fumiko Sekiguchi
- Subjects
Male ,hydrogen sulfide (H2S) ,cystathionine-γ-lyase (CSE) ,Receptor for Advanced Glycation End Products ,Cystitis, Interstitial ,Pharmacology ,Calcium Channels, T-Type ,Mice ,Adenosine Triphosphate ,Cav3.2 T-type Ca2+ channel ,Acrolein ,HMGB1 Protein ,lcsh:QH301-705.5 ,Adenosine triphosphate (ATP) ,chemistry.chemical_classification ,Mice, Knockout ,biology ,high mobility group box 1 (HMGB1) ,Cell migration ,General Medicine ,Up-Regulation ,Hyperalgesia ,Nociceptor ,cardiovascular system ,Female ,medicine.symptom ,Signal Transduction ,Cell signaling ,chemical and pharmacologic phenomena ,macrophage ,HMGB1 ,reactive oxygen species (ROS) ,Article ,Downregulation and upregulation ,medicine ,Animals ,Sulfites ,Bladder Pain ,Cyclophosphamide ,cyclophosphamide (CPA) ,Reactive oxygen species ,Macrophages ,Cystathionine gamma-Lyase ,interstitial cystitis/bladder pain syndrome (IC/BPS) ,equipment and supplies ,receptor for advanced glycation end products (RAGE) ,Disease Models, Animal ,lcsh:Biology (General) ,chemistry ,biology.protein - Abstract
Cystitis-related bladder pain involves RAGE activation by HMGB1, and increased Cav3.2 T-type Ca2+ channel activity by H2S, generated by upregulated cystathionine-&gamma, lyase (CSE) in mice treated with cyclophosphamide (CPA). We, thus, investigated possible crosstalk between the HMGB1/RAGE and CSE/H2S/Cav3.2 pathways in the bladder pain development. Bladder pain (nociceptive behavior/referred hyperalgesia) and immuno-reactive CSE expression in the bladder were determined in CPA-treated female mice. Cell signaling was analyzed in urothelial T24 and macrophage-like RAW264.7 cells. The CPA-induced bladder pain was abolished by pharmacological inhibition of T-type Ca2+ channels or CSE, and genetic deletion of Cav3.2. The CPA-induced CSE upregulation, as well as bladder pain was prevented by HMGB1 inactivation, inhibition of HMGB1 release from macrophages, antagonists of RAGE or P2X4/P2X7 receptors, and N-acetylcysteine, an antioxidant. Acrolein, a metabolite of CPA, triggered ATP release from T24 cells. Adenosine triphosphate (ATP) stimulated cell migration via P2X7/P2X4, and caused HMGB1 release via P2X7 in RAW264.7 cells, which was dependent on p38MAPK/NF-&kappa, B signaling and reactive oxygen species (ROS) accumulation. Together, our data suggest that CPA, once metabolized to acrolein, causes urothelial ATP-mediated, redox-dependent HMGB1 release from macrophages, which in turn causes RAGE-mediated CSE upregulation and subsequent H2S-targeted Cav3.2-dependent nociceptor excitation, resulting in bladder pain.
- Published
- 2020
14. Role of high-mobility group box 1 and its modulation by thrombomodulin/thrombin axis in neuropathic and inflammatory pain
- Author
-
Atsufumi Kawabata, Maho Tsubota, Fumiko Sekiguchi, and Ryuichi Tsujita
- Subjects
0301 basic medicine ,Thrombomodulin ,Receptor for Advanced Glycation End Products ,Pain ,chemical and pharmacologic phenomena ,Pharmacology ,HMGB1 ,RAGE (receptor) ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,HMGB1 Protein ,biology ,business.industry ,Thrombin ,Visceral pain ,030104 developmental biology ,Allodynia ,Neuropathic pain ,Hyperalgesia ,TLR4 ,biology.protein ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
High-mobility group box 1 (HMGB1), a nuclear protein, once released to the extracellular space, facilitates pain signals as well as inflammation. Intraplantar or intraspinal application of HMGB1 elicits hyperalgesia/allodynia in rodents by activating the advanced glycosylation end-product specific receptor (receptor for advanced glycation end-products; RAGE) or Toll-like receptor 4 (TLR4). Endogenous HMGB1 derived from neurons, perineuronal cells or immune cells accumulating in the dorsal root ganglion or sensory nerves participates in somatic and visceral pain consisting of neuropathic and/or inflammatory components. Endothelial thrombomodulin (TM) and recombinant human soluble TM, TMα, markedly increase thrombin-dependent degradation of HMGB1, and systemic administration of TMα prevents and reverses various HMGB1-dependent pathological pain. Low MW compounds that directly inactivate HMGB1 or antagonize HMGB1-targeted receptors would be useful to reduce various forms of intractable pain. Thus, HMGB1 and its receptors are considered to serve as promising targets in developing novel agents to prevent or treat pathological pain. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
- Published
- 2020
15. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: Evidence for a neuroimmune crosstalk
- Author
-
Maho Tsubota, Atsufumi Kawabata, Masahiro Nishibori, Kana Nakashima, Fumiko Sekiguchi, Hidenori Wake, Risa Domoto, Hiroki Yamanishi, and Daichi Yamasoba
- Subjects
Male ,0301 basic medicine ,Pyridines ,Thrombomodulin ,Receptor for Advanced Glycation End Products ,Minocycline ,Stimulation ,Pharmacology ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Ganglia, Spinal ,p300-CBP Transcription Factors ,HMGB1 Protein ,Phosphorylation ,Cells, Cultured ,Neurons ,biology ,Imidazoles ,Peripheral Nervous System Diseases ,Sciatic Nerve ,Recombinant Proteins ,Up-Regulation ,Allodynia ,Paclitaxel ,Chemotherapy-induced peripheral neuropathy ,Hyperalgesia ,medicine.symptom ,Receptors, CXCR4 ,Proline ,p38 mitogen-activated protein kinases ,chemical and pharmacologic phenomena ,HMGB1 ,Antibodies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Downregulation and upregulation ,Thiocarbamates ,medicine ,Animals ,Pyruvates ,Macrophages ,Membrane Proteins ,Phosphoproteins ,medicine.disease ,Coculture Techniques ,Acetylcysteine ,030104 developmental biology ,Peripheral neuropathy ,chemistry ,biology.protein ,Clodronic Acid ,Reactive Oxygen Species ,030217 neurology & neurosurgery - Abstract
Given our recent evidence for the role of high mobility group box 1 (HMGB1) in chemotherapy-induced peripheral neuropathy (CIPN) in rats, we examined the origin of HMGB1 and the upstream and downstream mechanisms of HMGB1 release involved in paclitaxel-induced neuropathy in mice. Paclitaxel treatment developed mechanical allodynia in mice, as assessed by von Frey test, which was prevented by an anti-HMGB1-neutralizing antibody or thrombomodulin alfa capable of inactivating HMGB1. RAGE or CXCR4 antagonists, ethyl pyruvate or minocycline, known to inhibit HMGB1 release from macrophages, and liposomal clodronate, a macrophage depletor, prevented the paclitaxel-induced allodynia. Paclitaxel caused upregulation of RAGE and CXCR4 in the dorsal root ganglia and macrophage accumulation in the sciatic nerve. In macrophage-like RAW264.7 cells, paclitaxel evoked cytoplasmic translocation of nuclear HMGB1 followed by its extracellular release, and overexpression of CBP and PCAF, histone acetyltransferases (HATs), known to cause acetylation and cytoplasmic translocation of HMGB1, which were suppressed by ethyl pyruvate, N-acetyl- l -cysteine, an anti-oxidant, and SB203580 and PDTC, inhibitors of p38 MAP kinase (p38MAPK) and NF-κB, respectively. Paclitaxel increased accumulation of reactive oxygen species (ROS) and phosphorylation of p38MAPK, NF-κB p65 and I-κB in RAW264.7 cells. In mice, N-acetyl- l -cysteine or PDTC prevented the paclitaxel-induced allodynia. Co-culture of neuron-like NG108-15 cells or stimulation with their conditioned medium promoted paclitaxel-induced HMGB1 release from RAW264.7 cells. Our data indicate that HMGB1 released from macrophages through the ROS/p38MAPK/NF-κB/HAT pathway participates in the paclitaxel-induced peripheral neuropathy in mice, and unveils an emerging therapeutic avenue targeting a neuroimmune crosstalk in CIPN.
- Published
- 2018
16. Involvement of Voltage-Gated Calcium Channels in Inflammation and Inflammatory Pain
- Author
-
Atsufumi Kawabata, Maho Tsubota, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,Pain ,Pharmaceutical Science ,Inflammation ,Pharmacology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Irritable bowel syndrome ,Neurogenic inflammation ,Voltage-dependent calcium channel ,business.industry ,Interstitial cystitis ,Visceral pain ,General Medicine ,medicine.disease ,030104 developmental biology ,Neuropathic pain ,Pancreatitis ,Calcium Channels ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Voltage-gated calcium channels (VGCCs) are classified into high-voltage-activated (HVA) channels and low-voltage-activated channels consisting of Cav3.1-3.3, known as T ("transient")-type VGCC. There is evidence that certain types of HVA channels are involved in neurogenic inflammation and inflammatory pain, in agreement with reports indicating the therapeutic effectiveness of gabapentinoids, ligands for the α2δ subunit of HVA, in treating not only neuropathic, but also inflammatory, pain. Among the Cav3 family members, Cav3.2 is abundantly expressed in the primary afferents, regulating both neuronal excitability at the peripheral terminals and spontaneous neurotransmitter release at the spinal terminals. The function and expression of Cav3.2 are modulated by a variety of inflammatory mediators including prostanoids and hydrogen sulfide (H2S), a gasotransmitter. The increased activity of Cav3.2 by H2S participates in colonic, bladder and pancreatic pain, and regulates visceral inflammation. Together, VGCCs are involved in inflammation and inflammatory pain, and Cav3.2 T-type VGCC is especially a promising therapeutic target for the treatment of visceral inflammatory pain in patients with irritable bowel syndrome, interstitial cystitis/bladder pain syndrome, pancreatitis, etc., in addition to neuropathic pain.
- Published
- 2018
17. Design and synthesis of novel anti-hyperalgesic agents based on 6-prenylnaringenin as the T-type calcium channel blockers
- Author
-
Takuya Okada, Maho Tsubota, Hiroyuki Nishikawa, Yamato Horaguchi, Huy Du Nguyen, Fumiko Sekiguchi, Shun Kitamura, Shigeru Yoshida, Yoshihito Kasanami, Atsufumi Kawabata, Sakura Yamaoka, and Naoki Toyooka
- Subjects
Male ,0301 basic medicine ,Patch-Clamp Techniques ,medicine.drug_class ,Clinical Biochemistry ,Action Potentials ,Pharmaceutical Science ,Calcium channel blocker ,Pharmacology ,01 natural sciences ,Biochemistry ,Calcium Channels, T-Type ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Biology ,IC50 ,Flavonoids ,Analgesics ,010405 organic chemistry ,Organic Chemistry ,T-type calcium channel ,Transfection ,Calcium Channel Blockers ,0104 chemical sciences ,Disease Models, Animal ,HEK293 Cells ,030104 developmental biology ,chemistry ,Drug Design ,Systemic administration ,Neuralgia ,Molecular Medicine ,Sciatic nerve ,Flavanone - 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.
- Published
- 2018
18. Involvement of the cystathionine-γ-lyase/Cav3.2 pathway in substance P-induced bladder pain in the mouse, a model for nonulcerative bladder pain syndrome
- Author
-
Yasumasa Okawa, Mariko Maeda, Maho Tsubota, Tomoka Ozaki, Yuhei Irie, Hiroyasu Ishikura, Fumiko Sekiguchi, and Atsufumi Kawabata
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cyclophosphamide ,Urology ,Substance P ,urologic and male genital diseases ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Edema ,Medicine ,Bladder Pain ,Pharmacology ,business.industry ,Interstitial cystitis ,medicine.disease ,030104 developmental biology ,Nociception ,chemistry ,Hyperalgesia ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Hemorrhagic cystitis ,medicine.drug - Abstract
Hydrogen sulfide (H2S) formed by cystathionine-γ-lyase (CSE) enhances the activity of Cav3.2 T-type Ca2+ channels, contributing to the bladder pain accompanying hemorrhagic cystitis caused by systemic administration of cyclophosphamide (CPA) in mice. Given clinical and fundamental evidence for the involvement of the substance P/NK1 receptor systems in bladder pain syndrome (BPS)/interstitial cystitis (IC), we created an intravesical substance P-induced bladder pain model in mice and analyzed the possible involvement of the CSE/Cav3.2 pathway. Bladder pain/cystitis was induced by i.p. CPA or intravesical substance P in female mice. Bladder pain was evaluated by counting nociceptive behavior and by detecting referred hyperalgesia in the lower abdomen and hindpaw. The isolated bladder tissue was weighed to estimate bladder swelling and subjected to histological observation and Western blotting. Intravesical substance P caused profound referred hyperalgesia accompanied by little bladder swelling or edema 6-24 h after the administration, in contrast to i.p. CPA-induced nociceptive behavior/referred hyperalgesia with remarkable bladder swelling/edema and urothelial damage. The bladder pain and/or cystitis symptoms caused by substance P or CPA were prevented by the NK1 receptor antagonist. CSE in the bladder was upregulated by substance P or CPA, and the NK1 antagonist prevented the CPA-induced CSE upregulation. A CSE inhibitor, a T-type Ca2+ channel blocker and gene silencing of Cav3.2 abolished the intravesical substance P-induced referred hyperalgesia. The intravesical substance P-induced pain in mice is useful as a model for nonulcerative BPS, and involves the activation of the NK1 receptor/CSE/H2S/Cav3.2 cascade.
- Published
- 2018
19. Prostanoid-dependent bladder pain caused by proteinase-activated receptor-2 activation in mice: Involvement of TRPV1 and T-type Ca2+ channels
- Author
-
Hiroyuki Nishikawa, Yasumasa Okawa, Ayaka Fujimura, Maho Tsubota, Tomoka Ozaki, Fumiko Sekiguchi, Yuko Hayashi, and Atsufumi Kawabata
- Subjects
0301 basic medicine ,Prostaglandin E2 ,TRPV1 ,Stimulation ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Channel blocker ,Bladder Pain ,Receptor ,Chemistry ,lcsh:RM1-950 ,Prostanoid ,PAR2 ,Bladder pain ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,Hyperalgesia ,Molecular Medicine ,medicine.symptom ,030217 neurology & neurosurgery ,medicine.drug - Abstract
We studied the pronociceptive role of proteinase-activated receptor-2 (PAR2) in mouse bladder. In female mice, intravesical infusion of the PAR2-activating peptide, SLIGRL-amide (SL), caused delayed mechanical hypersensitivity in the lower abdomen, namely ‘referred hyperalgesia’, 6–24 h after the administration. The PAR2-triggered referred hyperalgesia was prevented by indomethacin or a selective TRPV1 blocker, and restored by a T-type Ca2+ channel blocker. In human urothelial T24 cells, SL caused delayed prostaglandin E2 production and COX-2 upregulation. Our data suggest that luminal PAR2 stimulation in the bladder causes prostanoid-dependent referred hyperalgesia in mice, which involves the activation of TRPV1 and T-type Ca2+ channels.
- Published
- 2018
20. Genetic deletion of Cav3.2 T-type calcium channels abolishes H2S-dependent somatic and visceral pain signaling in C57BL/6 mice
- Author
-
Maho Tsubota, Mitsutaka Takada, Saaya Fukushi, Akira Oita, Hiroshi Masuda, Kazuki Matsui, Fumiko Sekiguchi, Yutaro Mukai, Shigeru Yoshida, Takaya Miyazaki, Atsufumi Kawabata, Yoshihito Kasanami, Nene Koike, and Tsuyako Ohkubo
- Subjects
0301 basic medicine ,Pharmacology ,C57BL/6 ,biology ,Chemistry ,Somatic cell ,lcsh:RM1-950 ,T-type calcium channel ,Visceral pain ,biology.organism_classification ,Intracolonic ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Nociception ,Allodynia ,lcsh:Therapeutics. Pharmacology ,medicine ,Molecular Medicine ,Channel blocker ,medicine.symptom ,030217 neurology & neurosurgery - 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. Keywords: Hydrogen sulfide, Cav3.2 T-type calcium channel, Pain
- Published
- 2019
21. Macrophage-derived HMGB1 as a Pain Mediator in the Early Stage of Acute Pancreatitis in Mice: Targeting RAGE and CXCL12/CXCR4 Axis
- Author
-
Hiroyasu Ishikura, Maho Tsubota, Keyue Liu, Yuhei Irie, Fumiko Sekiguchi, Masahiro Nishibori, Atsufumi Kawabata, Yuka Terada, and Toshifumi Tsujiuchi
- Subjects
Male ,0301 basic medicine ,Receptors, CXCR4 ,medicine.medical_specialty ,Receptor for Advanced Glycation End Products ,Immunology ,Neuroscience (miscellaneous) ,Pain ,chemical and pharmacologic phenomena ,HMGB1 ,RAGE (receptor) ,Mice ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,HMGB1 Protein ,Pharmacology ,biology ,Microglia ,business.industry ,Macrophages ,medicine.disease ,Chemokine CXCL12 ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Pancreatitis ,Hyperalgesia ,biology.protein ,TLR4 ,Acute pancreatitis ,medicine.symptom ,Pancreas ,business ,Signal Transduction - Abstract
Extracellular high mobility group box 1 (HMGB1) activates the receptor for advanced glycation end products (RAGE) or Toll-like receptor 4 (TLR4) and forms a heterocomplex with CXCL12 that strongly activates CXCR4, promoting inflammatory and pain signals. In the present study, we investigated the role of HMGB1 in pancreatic pain accompanying cerulein-induced acute pancreatitis in mice. Abdominal referred hyperalgesia accompanying acute pancreatitis occurred within 1 h after 6 hourly injections of cerulein. The anti-HMGB1 neutralizing antibody or recombinant human soluble thrombomodulin (rhsTM), known to inactivate HMGB1, abolished the cerulein-induced referred hyperalgesia, but not pancreatitis itself. Plasma or pancreatic HMGB1 levels did not change, but macrophage infiltration into the pancreas occurred 1 h after cerulein treatment. Minocycline, a macrophage/microglia inhibitor, ethyl pyruvate that inhibits HMGB1 release from macrophages, or liposomal clodronate that depletes macrophages prevented the referred hyperalgesia, but not pancreatitis. Antagonists of RAGE or CXCR4, but not TLR4, strongly suppressed the cerulein-induced referred hyperalgesia, but not pancreatitis. Upregulation of RAGE, CXCR4 and CXCL12, but not TLR4, were detected in the pancreas 1 h after cerulein treatment. Our data suggest that HMGB1 regionally secreted by macrophages mediates pancreatic pain by targeting RAGE and CXCL12/CXCR4 axis in the early stage of acute pancreatitis.
- Published
- 2017
22. Circadian pharmacokinetics and limited sampling strategy of everolimus in heart transplant patients
- Author
-
Mitsutaka Takada, Kyoichi Wada, Atsufumi Kawabata, Seiko Nakajima, Yuka Terada, Takuya Watanabe, Takeshi Kuwahara, Sachi Matsuda, Masanobu Yanase, Norihide Fukushima, Osamu Seguchi, Takuma Sato, and Takeshi Nakatani
- Subjects
Adult ,Male ,medicine.medical_specialty ,Period (gene) ,medicine.medical_treatment ,Cmax ,02 engineering and technology ,Pharmacology ,Gastroenterology ,Pharmacokinetics ,Internal medicine ,0202 electrical engineering, electronic engineering, information engineering ,medicine ,Humans ,Pharmacology (medical) ,Everolimus ,Circadian rhythm ,Retrospective Studies ,Whole blood ,060201 languages & linguistics ,Heart transplantation ,business.industry ,Drug Chronotherapy ,06 humanities and the arts ,Area Under Curve ,Data Interpretation, Statistical ,0602 languages and literature ,Heart Transplantation ,Female ,020201 artificial intelligence & image processing ,Drug Monitoring ,business ,Immunosuppressive Agents ,medicine.drug ,Blood sampling - Abstract
Objective To evaluate circadian changes in everolimus (EVL) pharmacokinetics and to identify the time point of blood sampling with the strongest correlation with the area under the blood concentration-time curve (AUC) of EVL in heart transplant patients. Methods Heart transplant patients receiving the same dose of EVL twice a day were reviewed. In 28 patients enrolled, whole blood samples were collected before (C0), and 1, 2, 4, 6, 8, and 12 hours after each administration of EVL. Blood concentrations of EVL were compared between active (9:00 AM to 9:00 PM) and resting periods (9:00 PM to 9:00 AM). Results AUC0-4h, peak concentration (Cmax), Cmax/minimum concentration, and peak-trough fluctuation in the resting period were significantly lower than those in the active period (p = 0.008, 0.017, 0.022, and 0.011, respectively). Half-life and mean residence time were significantly longer in the resting period than in the active period (p = 0.002 and 0.002, respectively). AUC0-12h in the active period was similar (p = 0.154) and correlated with that in the resting period (r2 = 0.93). Two-point blood samplings, C0 and C2, correlated more strongly with AUC0-12h for EVL, compared with C0 alone (0.92 vs. 0.79, respectively, for r2 in the active period). Conclusions EVL pharmacokinetics showed circadian changes, suggesting delayed absorption and decreased metabolic activity at rest. However, the circadian changes did not affect AUC0-12h. A 2-time-point model that included C0 and C2 was more accurate for predicting the AUC0-12h of EVL than C0 alone in heart transplant patients. .
- Published
- 2017
23. Contents Vol. 99, 2017
- Author
-
Akira Oita, Josué A. Velázquez-Moyado, Mohamed Habib Grissa, Zhi Dong, Nizar Fredj, Adel Sekma, Adelfo Reyes-Ramírez, Takayo Haruna, Kaoru Ogawa, Tin Sandar Zaw, Hiiragi Sugo, Semir Nouira, Wen-Bin Wu, Katsuaki Dan, Hiroshi Sakonjo, Juan Carlos Castillo-Hernández, Martín González-Andrade, Mi Tang, Kinichi Imura, Tomio Okamura, Jun Hayakawa, Masashi Deguchi, Yuka Terada, Menglin Rao, Kamel Monastiri, Haruka Saeki, Uy Dong Sohn, Takashi Shimosato, Hideki Hasegawa, Akinori Arimura, Yasuhide Morioka, Chi Zhang, Mitsutaka Takada, Elena G. Ramírez-López, Emna Kerkeni, Malek Mzali, Kohei Wakitani, Kyoichi Wada, Saki Hiruma, Yuichi Uwai, Tsong-Long Hwang, Takaya Miyazaki, Yujie Cheng, Jihua Ma, Fumiko Sekiguchi, Phyu Phyu Khin, Keita Takanashi, Nejia Tounsi, Sho Kanzaki, Huey-Ming Lo, Masashi Matsumoto, Jie Hao, Junlian Xing, Wanzhen Jiang, Druckerei Stückle, Masashi Tawa, Yoshinori Funakami, Wahiba Douki, Kenji Watanabe, Zhipei Liu, Maho Tsubota, Yoko Furue, Tomoyoshi Miyamoto, Imen Trabelsi, Atsufumi Kawabata, Mina Yamamoto, Kiyoshi Yasui, Sha Liu, Ilhem Hellara, Andrés Navarrete, Peihua Zhang, Xinrong Fan, Masahiko Soga, Antao Luo, Tomohiro Nabekura, and Tatsuya Kawasaki
- Subjects
Pharmacology ,General Medicine - Published
- 2017
24. Tacrolimus Triggers Transient Receptor Potential Vanilloid-1-Dependent Relapse of Pancreatitis-Related Pain in Mice
- Author
-
Mitsutaka Takada, Yuka Terada, Atsufumi Kawabata, Kyoichi Wada, Maho Tsubota, Hiiragi Sugo, Fumiko Sekiguchi, Kohei Wakitani, and Akira Oita
- Subjects
Cyclopropanes ,Male ,0301 basic medicine ,medicine.medical_specialty ,TRPV1 ,Pain ,TRPV Cation Channels ,Naphthalenes ,Tacrolimus ,Mice ,03 medical and health sciences ,Transient receptor potential channel ,0302 clinical medicine ,Recurrence ,Internal medicine ,medicine ,Animals ,Anilides ,Protein kinase A ,Pharmacology ,business.industry ,General Medicine ,medicine.disease ,Calcineurin ,surgical procedures, operative ,030104 developmental biology ,Endocrinology ,Pancreatitis ,nervous system ,Cinnamates ,Hyperalgesia ,Nociceptor ,Benzimidazoles ,medicine.symptom ,business ,Ceruletide ,030217 neurology & neurosurgery - Abstract
Transient receptor potential vanilloid-1 (TRPV1) expressed in nociceptors is directly phosphorylated and activated by protein kinase C, and involved in the signaling of pancreatic pain. On the other hand, Cav3.2 T-type Ca2+ channels expressed in nociceptors are functionally upregulated by phosphorylation with protein kinase A and also play a role in pancreatitis-related pain. Calcineurin, a phosphatase, negatively regulates various channel functions including TRPV1, and calcineurin inhibitor-induced pain syndrome by tacrolimus, a calcineurin inhibitor, used as an immunosuppressant, has been a clinical problem. We thus examined the effect of tacrolimus on pancreatitis-related pain in mice. Repeated treatment with cerulein caused referred hyperalgesia accompanying acute pancreatitis, which was unaffected by tacrolimus. Pancreatitis-related symptoms disappeared in 24 h, whereas the referred hyperalgesia recurred following the administration of tacrolimus, which was abolished by the blockers of TRPV1 but not T-type Ca2+ channels. Thus, tacrolimus appears to cause the TRPV1-dependent relapse of pancreatitis-related pain, suggesting the involvement of calcineurin in the termination of pancreatic pain.
- Published
- 2017
25. Hydrogen Sulfide and T-Type Ca2+ Channels in Pain Processing, Neuronal Differentiation and Neuroendocrine Secretion
- Author
-
Kazuki Fukami, Fumiko Sekiguchi, and Atsufumi Kawabata
- Subjects
0301 basic medicine ,Pharmacology ,chemistry.chemical_classification ,medicine.medical_specialty ,Voltage-dependent calcium channel ,Chemistry ,Hydrogen sulfide ,Cellular differentiation ,Neuronal differentiation ,General Medicine ,Pain processing ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Enzyme ,Endocrinology ,Internal medicine ,medicine ,Ca2 channels ,Secretion ,030217 neurology & neurosurgery - Abstract
Background: Hydrogen sulfide (H2S), a gasotransmitter, is generated from L-cysteine by mainly 3 enzymes, cystathionine-γ-lyase (CSE), cystathionine-β-synthase, and 3-mercaptopyruvate sulfurtransferase in cooperation with cysteine aminotransferase. The H2S-forming enzymes, particularly CSE, are overexpressed under the pathological conditions such as inflammation, neuronal or neuroendocrine differentiation and cancer development. Given that Cav3.2 T-type Ca2+ channels mediate some of the biological activity of H2S, we focus on the role of the H2S/Cav3.2 pathway in regulating the neuronal and neuroendocrine function. Summary: In the neuronal system, H2S regulates the activity of various ion channels including Cav3.2. Exogenous and endogenous H2S enhances the Cav3.2 channel activity, promoting somatic and visceral pain signaling. The H2S/Cav3.2 pathway also facilitates neuritogenesis or neuronal differentiation. Interestingly, endogenous H2S formed by CSE regulates secretory function by enhancing Cav3.2 channel activity in neuroendocrine-differentiated prostate cancer cells or carotid glomus cells. Key Messages: The H2S/Cav3.2 pathway may serve as therapeutic targets for treatment of intractable pain, neuronal injury, androgen-independent prostate cancer, cardiovascular diseases, etc.
- Published
- 2016
26. [HMGB1 as a target for prevention of chemotherapy-induced peripheral neuropathy]
- Author
-
Atsufumi Kawabata, Maho Tsubota, Ryuichi Tsujita, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Thrombomodulin ,Antineoplastic Agents ,HMGB1 ,03 medical and health sciences ,Drug treatment ,0302 clinical medicine ,Japan ,Internal medicine ,Medicine ,Humans ,Molecular Targeted Therapy ,HMGB1 Protein ,Pharmacology ,Disseminated intravascular coagulation ,biology ,business.industry ,Cancer ,Peripheral Nervous System Diseases ,medicine.disease ,Discontinuation ,030104 developmental biology ,Peripheral neuropathy ,Chemotherapy-induced peripheral neuropathy ,biology.protein ,business ,030217 neurology & neurosurgery - Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) considerably impairs cancer patients' QOL, and may lead to discontinuation of drug treatment of cancer. Currently, there is no effective strategy against CIPN. Therefore, it is an urgent issue to develop clinically available drugs that prevent or treat CIPN. We have shown that high mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) molecule, plays an essential role in the development of CIPN. Most interestingly, thrombomodulin α, approved as a medicine for treatment of disseminated intravascular coagulation (DIC) in Japan, causes thrombin-dependent degradation of extracellular HMGB1 that is released in response to chemotherapeutics, and prevents CIPN. Thus, we expect that targeting HMGB1 or its receptors would lead to prevention of CIPN in cancer patients in near future.
- Published
- 2019
27. Role of non-macrophage cell-derived HMGB1 in oxaliplatin-induced peripheral neuropathy and its prevention by the thrombin/thrombomodulin system in rodents: negative impact of anticoagulants
- Author
-
Toshiyuki Araki, Ryotaro Fukuda, Fumiko Sekiguchi, Maho Tsubota, Nene Koike, Takaya Miyazaki, Yuka Ujiie, Shuji Wakatsuki, Atsufumi Kawabata, Rika Yamashita, Shin Ueda, Masahiro Nishibori, Hidenori Wake, and Yusuke Hayashi
- Subjects
Male ,Thrombomodulin ,Immunology ,Antineoplastic Agents ,Rodentia ,Chemotherapy-induced peripheral neuropathy (CIPN) ,Pharmacology ,lcsh:RC346-429 ,Argatroban ,Mice ,Cellular and Molecular Neuroscience ,Thrombin ,medicine ,Animals ,Nociception assay ,HMGB1 Protein ,Rats, Wistar ,Receptor ,lcsh:Neurology. Diseases of the nervous system ,Chemistry ,Research ,General Neuroscience ,Anticoagulants ,Peripheral Nervous System Diseases ,medicine.disease ,Rats ,Oxaliplatin ,RAW 264.7 Cells ,Peripheral neuropathy ,Allodynia ,Neurology ,medicine.symptom ,High mobility group box 1 (HMGB1) ,medicine.drug - Abstract
Background Macrophage-derived high mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) protein, plays a key role in the development of chemotherapy-induced peripheral neuropathy (CIPN) caused by paclitaxel in rodents. Endothelial thrombomodulin (TM) promotes thrombin-induced degradation of HMGB1, and TMα, a recombinant human soluble TM, abolishes peripheral HMGB1-induced allodynia in mice. We thus examined whether HMGB1, particularly derived from macrophages, contributes to oxaliplatin-induced neuropathy in mice and analyzed the anti-neuropathic activity of the TM/thrombin system. Methods CIPN models were created by the administration of oxaliplatin in mice and rats, and the nociceptive threshold was assessed by von Frey test or paw pressure test. Macrophage-like RAW264.7 cells were stimulated with oxaliplatin in vitro. Proteins were detected and/or quantified by Western blotting, immunostaining, or enzyme-linked immunosorbent assay. Results Intraperitoneal administration of an anti-HMGB1-neutralizing antibody (AB) at 1 mg/kg prevented the oxaliplatin-induced allodynia in mice and rats. Antagonists of Toll-like receptor (TLR) 4, receptor for advanced glycation end products (RAGE) and CXCR4 among the HMGB1-targeted pro-nociceptive receptors, also mimicked the anti-neuropathic activity of AB in mice. Macrophage accumulation in the sciatic nerve was observed in mice treated with paclitaxel, but not oxaliplatin, and neither macrophage depletion nor inhibitors of macrophage activation affected oxaliplatin-induced allodynia. Oxaliplatin was 10- to 100-fold less potent than paclitaxel in releasing HMGB1 from macrophage-like RAW264.7 cells. Like AB, TMα at 10 mg/kg prevented the oxaliplatin-induced allodynia in mice as well as rats, an effect abolished by argatroban at 10 mg/kg, a thrombin inhibitor. The anti-neuropathic activity of TMα in oxaliplatin-treated mice was suppressed by oral anticoagulants such as warfarin at 1 mg/kg, dabigatran at 75 mg/kg, and rivaroxaban at 10 mg/kg, but not antiplatelet agents such as aspirin at 50 mg/kg and clopidogrel at 10 mg/kg. Repeated administration of the anticoagulants gradually developed neuropathic allodynia and elevated plasma HMGB1 levels in mice treated with a subeffective dose of oxaliplatin. Conclusions Our data thus suggests a causative role of HMGB1 derived from non-macrophage cells in oxaliplatin-induced peripheral neuropathy and a thrombin-dependent anti-neuropathic activity of exogenous TMα and, most probably, endogenous TM.
- Published
- 2019
28. Dietary ascorbic acid restriction in GNL/SMP30-knockout mice unveils the role of ascorbic acid in regulation of somatic and visceral pain sensitivity
- Author
-
Atsufumi Kawabata, Kenta Uebo, Akihiko Ishigami, Maho Tsubota, Koki Miki, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,Male ,Pain Threshold ,Biophysics ,Ascorbic Acid ,Pharmacology ,Biochemistry ,03 medical and health sciences ,Calcium Channels, T-Type ,0302 clinical medicine ,medicine ,Animals ,Channel blocker ,Molecular Biology ,chemistry.chemical_classification ,Mice, Knockout ,Vitamin C ,Chemistry ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Visceral pain ,Cell Biology ,Visceral Pain ,Ascorbic acid ,Mice, Inbred C57BL ,030104 developmental biology ,Enzyme ,Allodynia ,Hyperalgesia ,030220 oncology & carcinogenesis ,Knockout mouse ,Ascorbic Acid Deficiency ,medicine.symptom - Abstract
Cav3.2 T-type Ca2+ channels are expressed in the primary afferents and play a pronociceptive role. The activity of Cav3.2 is enhanced by H2S, a gasotransmitter, and suppressed by ascorbic acid (vitamin C) through metal-catalyzed oxidation of the Zn2+-binding His191 in Cav3.2. Since rodents, but not humans, are capable of synthesizing ascorbic acid, the present study examined the role of ascorbic acid in nociceptive processing, using the mice lacking GNL/SMP30, an enzyme essential for ascorbic acid biosynthesis. Intraplantar and intracolonic administration of NaHS, an H2S donor, caused somatic allodynia and referred hyperalgesia, respectively, and repeated treatment with paclitaxel produced neuropathic allodynia in wild-type mice, all of which were suppressed by ascorbic acid or T-type Ca2+ channel blockers. Dietary ascorbic acid restriction caused dramatic decreases in plasma and tissue ascorbic acid levels in GNL/SMP30-knockout, but not wild-type, mice. The ascorbic acid restriction enhanced the somatic and visceral hypersensitivity following intraplantar and intracolonic NaHS, respectively, and paclitaxel-induced neuropathy in GNL/SMP30-knockout mice, while it had no such effect in wild-type mice. Together, our data unveil the critical role of ascorbic acid in regulating somatic and visceral pain sensitivity and support accumulating clinical evidence for the usefulness of ascorbic acid in pain management.
- Published
- 2019
29. Corrigendum to 'Essential role of Cav3.2 T-type calcium channels in butyrate-induced colonic pain and nociceptor hypersensitivity in mice' [Eur. J. Pharmacol. 887 (2020) 173576]
- Author
-
Maho Tsubota, Ken Tomochika, Fumiko Sekiguchi, Atsufumi Kawabata, Maki Nakano, Ayumu Yamagata, Takuya Okada, Saaya Fukushi, Rie Kajitani, Naoki Toyooka, Yuko Ishii, Yuta Nishikawa, and Kazuki Matsui
- Subjects
Pharmacology ,Chemistry ,T-type calcium channel ,Nociceptor ,Butyrate - Published
- 2021
30. Cav3.2 overexpression in L4 dorsal root ganglion neurons after L5 spinal nerve cutting involves Egr-1, USP5 and HMGB1 in rats: An emerging signaling pathway for neuropathic pain
- Author
-
Shiori Tomita, Katsuki Naoe, Maho Tsubota, Fumiko Sekiguchi, Hidenori Wake, Atsufumi Kawabata, Masahiro Nishibori, and Yoshihito Kasanami
- Subjects
0301 basic medicine ,Pharmacology ,medicine.medical_specialty ,biology ,business.industry ,HMGB1 ,RAGE (receptor) ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Endocrinology ,Downregulation and upregulation ,Dorsal root ganglion ,Internal medicine ,Spinal nerve ,Neuropathic pain ,biology.protein ,Medicine ,Signal transduction ,business ,Receptor ,030217 neurology & neurosurgery - 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.
- Published
- 2020
31. The prostaglandin E2/EP4 receptor/cyclic AMP/T-type Ca2+ channel pathway mediates neuritogenesis in sensory neuron-like ND7/23 cells
- Author
-
Yukari Tanaka, Takashi Maeda, Atsufumi Kawabata, Shigeru Yoshida, Kenji Mitani, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Sensory Receptor Cells ,medicine.medical_treatment ,Neuronal Outgrowth ,EP4 Receptor ,Dinoprostone ,Adenylyl cyclase ,Calcium Channels, T-Type ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Cyclic AMP ,medicine ,Animals ,Prostaglandin E2 ,Protein kinase A ,Neuritogenesis ,Cells, Cultured ,Pharmacology ,Sensory neuron ,Forskolin ,Activator (genetics) ,lcsh:RM1-950 ,Rats ,Cell biology ,T-type calcium channel ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Receptors, Prostaglandin E, EP4 Subtype ,030217 neurology & neurosurgery ,Signal Transduction ,Prostaglandin E ,medicine.drug - Abstract
We investigated mechanisms for the neuritogenesis caused by prostaglandin E 2 (PGE 2 ) or intracellular cyclic AMP (cAMP) in sensory neuron-like ND7/23 cells. PGE 2 caused neuritogenesis, an effect abolished by an EP4 receptor antagonist or inhibitors of adenylyl cyclase (AC) or protein kinase A (PKA) and mimicked by the AC activator forskolin, dibutyryl cAMP (db-cAMP), and selective activators of PKA or Epac. ND7/23 cells expressed both Ca v 3.1 and Ca v 3.2 T-type Ca 2+ channels (T-channels). The neuritogenesis induced by db-cAMP or PGE 2 was abolished by T-channel blockers. T-channels were functionally upregulated by db-cAMP. The PGE 2 /EP4/cAMP/T-channel pathway thus appears to mediate neuritogenesis in sensory neurons.
- Published
- 2016
32. Essential role of Cav3.2 T-type calcium channels in butyrate-induced colonic pain and nociceptor hypersensitivity in mice
- Author
-
Ayumu Yamagata, Saaya Fukushi, Ken Tomochika, Yuta Nishikawa, Naoki Toyooka, Kazuki Matsui, Rie Kajitani, Maho Tsubota, Takuya Okada, Maki Nakano, Atsufumi Kawabata, Yuko Ishii, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,TRPV1 ,Visceral pain ,Butyrate ,Distension ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Nociception ,Hyperalgesia ,Nociceptor ,Medicine ,Nociception assay ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Given the role of Cav3.2 isoform among T-type Ca2+ channels (T-channels) in somatic and visceral nociceptive processing, we analyzed the contribution of Cav3.2 to butyrate-induced colonic pain and nociceptor hypersensitivity in mice, to evaluate whether Cav3.2 could serve as a target for treatment of visceral pain in irritable bowel syndrome (IBS) patients. Mice of ddY strain, and wild-type and Cav3.2-knockout mice of a C57BL/6J background received intracolonic administration of butyrate twice a day for 3 days. Referred hyperalgesia in the lower abdomen was assessed by von Frey test, and colonic hypersensitivity to distension by a volume load or chemicals was evaluated by counting nociceptive behaviors. Spinal phosphorylated ERK was detected by immunohistochemistry. Cav3.2 knockdown was accomplished by intrathecal injection of antisense oligodeoxynucleotides. Butyrate treatment caused referred hyperalgesia and colonic hypersensitivity to distension in ddY mice, which was abolished by T-channel blockers and/or Cav3.2 knockdown. Butyrate also increased the number of spinal phosphorylated ERK-positive neurons following colonic distension in the anesthetized ddY mice. The butyrate-treated ddY mice also exhibited T-channel-dependent colonic hypersensitivity to intracolonic Na2S, known to enhance Cav3.2 activity, and TRPV1, TRPA1 or proteinase-activated receptor 2 (PAR2) agonists. Wild-type, but not Cav3.2-knockout, mice of a C57BL/6J background, after treated with butyrate, mimicked the T-channel-dependent referred hyperalgesia and colonic hypersensitivity in butyrate-treated ddY mice. Our study provides definitive evidence for an essential role of Cav3.2 in the butyrate-induced colonic pain and nociceptor hypersensitivity, which might serve as a target for treatment of visceral pain in IBS patients.
- Published
- 2020
33. Blockade of T-type calcium channels by 6-prenylnaringenin, a hop component, alleviates neuropathic and visceral pain in mice
- Author
-
Tomoyo Fujita, Sakura Yamaoka, Fumiko Sekiguchi, Ken Tomochika, Nene Koike, Sumire Ono, Hiroyuki Nishikawa, Maki Ichii, Naoki Toyooka, Yamato Horaguchi, Mio Ichikawa, Takahiro Deguchi, Takuya Okada, Shigeru Yoshida, Hideaki Matsuda, Tsuyako Ohkubo, Kazuya Murata, Tadatoshi Tanino, Yumiko Ueno, Maho Tsubota, Huy Du Nguyen, and Atsufumi Kawabata
- Subjects
0301 basic medicine ,Naringenin ,Male ,Sophoraflavanone G ,Phytochemicals ,Mice, Transgenic ,Pharmacology ,Contractility ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Calcium Channels, T-Type ,Random Allocation ,0302 clinical medicine ,parasitic diseases ,medicine ,Animals ,Humans ,Rats, Wistar ,Humulus ,Flavonoids ,Plant Extracts ,T-type calcium channel ,Visceral pain ,Visceral Pain ,Analgesics, Non-Narcotic ,Calcium Channel Blockers ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Allodynia ,HEK293 Cells ,chemistry ,Hyperalgesia ,Neuropathic pain ,Neuralgia ,Sciatic nerve ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Since Cav3.2 T-type Ca2+ channels (T-channels) expressed in the primary afferents and CNS contribute to intractable pain, we explored T-channel-blocking components in distinct herbal extracts using a whole-cell patch-clamp technique in HEK293 cells stably expressing Cav3.2 or Cav3.1, and purified and identified sophoraflavanone G (SG) as an active compound from SOPHORAE RADIX (SR). Interestingly, hop-derived SG analogues, (2S)-6-prenylnaringenin (6-PNG) and (2S)-8-PNG, but not naringenin, also blocked T-channels; IC50 (μM) of SG, (2S)-6-PNG and (2S)-8-PNG was 0.68–0.75 for Cav3.2 and 0.99–1.41 for Cav3.1. (2S)-6-PNG and (2S)-8-PNG, but not SG, exhibited reversible inhibition. The racemic (2R/S)-6-PNG as well as (2S)-6-PNG potently blocked Cav3.2, but exhibited minor effect on high-voltage-activated Ca2+ channels and voltage-gated Na+ channels in differentiated NG108-15 cells. In mice, the mechanical allodynia following intraplantar (i.pl.) administration of an H2S donor was abolished by oral or i.p. SR extract and by i.pl. SG, (2S)-6-PNG or (2S)-8-PNG, but not naringenin. Intraperitoneal (2R/S)-6-PNG strongly suppressed visceral pain and spinal ERK phosphorylation following intracolonic administration of an H2S donor in mice. (2R/S)-6-PNG, administered i.pl. or i.p., suppressed the neuropathic allodynia induced by partial sciatic nerve ligation or oxaliplatin, an anti-cancer agent, in mice. (2R/S)-6-PNG had little or no effect on open-field behavior, motor performance or cardiovascular function in mice, and on the contractility of isolated rat aorta. (2R/S)-6-PNG, but not SG, was detectable in the brain after their i.p. administration in mice. Our data suggest that 6-PNG, a hop component, blocks T-channels, and alleviates neuropathic and visceral pain with little side effects.
- Published
- 2018
34. Functional upregulation of the H2S/Cav3.2 channel pathway accelerates secretory function in neuroendocrine-differentiated human prostate cancer cells
- Author
-
Mai Ueda, Atsufumi Kawabata, Shigeru Yoshida, Erina Asano, Fumiko Sekiguchi, Ryuji Kasamatsu, Kazuki Fukami, and Miku Yasukawa
- Subjects
Male ,medicine.medical_specialty ,Cellular differentiation ,Biology ,Biochemistry ,Calcium Channels, T-Type ,Neuroendocrine Cells ,Downregulation and upregulation ,Cell Line, Tumor ,Internal medicine ,LNCaP ,medicine ,Humans ,Secretion ,Hydrogen Sulfide ,Neurotensin ,Cell Proliferation ,Early Growth Response Protein 1 ,Pharmacology ,Cell growth ,Parathyroid Hormone-Related Protein ,T-type calcium channel ,Prostatic Neoplasms ,Cell Differentiation ,Up-Regulation ,Cell biology ,Repressor Proteins ,Endocrinology ,Prostatic acid phosphatase ,Cancer cell - Abstract
Neuroendocrine-differentiated prostate cancer cells may contribute to androgen-independent proliferation of surrounding cells through Ca(2+)-dependent secretion of mitogenic factors. Human prostate cancer LNCaP cells, when neuroendocrine-differentiated, overexpress Cav3.2 T-type Ca(2+) channels that contribute to Ca(2+)-dependent secretion. Given evidence for the acceleration of Cav3.2 activity by hydrogen sulfide (H2S), we examined the roles of the H2S/Cav3.2 pathway and then analyzed the molecular mechanisms of the Cav3.2 overexpression in neuroendocrine-differentiated LNCaP cells. LNCaP cells were differentiated by dibutyryl cyclic AMP. Protein levels and T-type Ca(2+) channel-dependent currents (T-currents) were measured by immunoblotting and whole-cell pacth-clamp technique, respectively. Spontaneous release of prostatic acid phosphatase (PAP) was monitored to evaluate secretory function. The differentiated LNCaP cells exhibited neurite outgrowth, androgen-independent proliferation and upregulation of mitogenic factors, and also showed elevation of Cav3.2 expression or T-currents. Expression of cystathionine-γ-lyase (CSE) and cystathionine-β-synthase (CBS), H2S-forming enzymes, and spontaneous secretion of PAP increased following the differentiation. The augmented T-currents were enhanced by H2S donors and suppressed by inhibitors of CSE, but not CBS. The PAP secretion was reduced by inhibition of CSE or T-type Ca(2+) channels. During differentiation, Egr-1 and REST, positive and negative transcriptional regulators for Cav3.2, were upregulated and downregulated, respectively, and Egr-1 knockdown prevented the Cav3.2 overexpression. Our data suggest that, in neuroendocrine-differentiated LNCaP cells, H2S formed by the upregulated CSE promotes the activity of the upregulated Cav3.2, leading to the elevated secretory functions. The overexpression of Cav3.2 appears to involve upregulation of Egr-1 and downregulation of REST.
- Published
- 2015
35. Role of Thrombin in Soluble Thrombomodulin-Induced Suppression of Peripheral HMGB1-Mediated Allodynia in Mice
- Author
-
Yusuke Hayashi, Atsufumi Kawabata, Ryuichi Tsujita, Maho Tsubota, Haruka Saeki, and Fumiko Sekiguchi
- Subjects
0301 basic medicine ,Male ,Neuroimmunomodulation ,Thrombomodulin ,Immunology ,Neuroscience (miscellaneous) ,chemical and pharmacologic phenomena ,Pharmacology ,HMGB1 ,RAGE (receptor) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Thrombin ,medicine ,Immunology and Allergy ,Nociception assay ,Animals ,HMGB1 Protein ,biology ,Chemistry ,030104 developmental biology ,Allodynia ,Hyperalgesia ,biology.protein ,TLR4 ,medicine.symptom ,030217 neurology & neurosurgery ,medicine.drug - Abstract
High mobility group box 1 (HMGB1), a nuclear protein, once released into the extracellular space under pathological conditions, plays a pronociceptive role in redox-dependent distinct active forms, all-thiol HMGB1 (at-HMGB1) and disulfide HMGB1 (ds-HMGB1), that accelerate nociception through the receptor for advanced glycation endproducts (RAGE) and Toll-like receptor 4 (TLR4), respectively. Thrombomodulin (TM), an endothelial membrane protein, and soluble TM, known as TMα, promote thrombin-mediated activation of protein C and also sequester HMGB1, which might facilitate thrombin degradation of HMGB1. The present study aimed at clarifying the role of thrombin in TMα-induced suppression of peripheral HMGB1-dependent allodynia in mice. Thrombin-induced degradation of at-HMGB1 and ds-HMGB1 was accelerated by TMα in vitro. Intraplantar (i.pl.) injection of bovine thymus-derived HMGB1 in an unknown redox state, at-HMGB1, ds-HMGB1 or lipopolysaccharide (LPS), known to cause HMGB1 secretion, produced long-lasting mechanical allodynia in mice, as assessed by von Frey test. TMα, when preadministered i.pl., prevented the allodynia caused by bovine thymus-derived HMGB1, at-HMGB1, ds-HMGB1 or LPS, in a dose-dependent manner. The TMα-induced suppression of the allodynia following i.pl. at-HMGB1, ds-HMGB1 or LPS was abolished by systemic preadministration of argatroban, a thrombin-inhibiting agent, and accelerated by i.pl. co-administered thrombin. Our data clearly indicate that TMα is capable of promoting the thrombin-induced degradation of both at-HMGB1 and ds-HMGB1, and suppresses the allodynia caused by either HMGB1 in a thrombin-dependent manner. Considering the emerging role of HMGB1 in distinct pathological pain models, the present study suggests the therapeutic usefulness of TMα for treatment of intractable and/or persistent pain.
- Published
- 2017
36. Involvement of NF-κB in the upregulation of cystathionine-γ-lyase, a hydrogen sulfide-forming enzyme, and bladder pain accompanying cystitis in mice
- Author
-
Tomoka Ozaki, Maho Tsubota, Fumiko Sekiguchi, and Atsufumi Kawabata
- Subjects
0301 basic medicine ,Curcumin ,Cyclophosphamide ,Proline ,Physiology ,Urinary Bladder ,Pain ,Pharmacology ,urologic and male genital diseases ,03 medical and health sciences ,chemistry.chemical_compound ,Calcium Channels, T-Type ,Mice ,0302 clinical medicine ,Pyrrolidine dithiocarbamate ,Downregulation and upregulation ,Thiocarbamates ,Physiology (medical) ,Cystitis ,medicine ,Nociception assay ,Animals ,heterocyclic compounds ,Hydrogen Sulfide ,Enzyme Inhibitors ,Bladder Pain ,Alanine ,Chemistry ,Cystathionine gamma-Lyase ,NF-kappa B ,Up-Regulation ,030104 developmental biology ,Nociception ,Anesthesia ,Hyperalgesia ,cardiovascular system ,Female ,medicine.symptom ,030217 neurology & neurosurgery ,medicine.drug ,Signal Transduction - Abstract
Hydrogen sulfide (H2 S) is generated from l-cysteine by multiple enzymes including cystathionine-γ-lyase (CSE), and promotes nociception by targeting multiple molecules such as Cav 3.2 T-type Ca2+ channels. Bladder pain accompanying cyclophosphamide (CPA)-induced cystitis in mice has been shown to involve the functional upregulation of the CSE/H2 S/Cav 3.2 pathway. Therefore, we investigated whether NF-κB, as an upstream signal of the CSE/H2 S system, contributes to bladder pain in mice with CPA-induced cystitis. Bladder pain-like nociceptive behaviour was observed in CPA-treated mice, and referred hyperalgesia was evaluated by the von Frey test. Isolated bladder weights were assessed to estimate bladder swelling, and protein levels were measured by Western blotting. CPA, administered intraperitoneally, induced nociceptive behaviour, referred hyperalgesia and increased bladder weights in mice. β-Cyano-l-alanine, a reversible selective CSE inhibitor, prevented CPA-induced nociceptive behaviour, referred hyperalgesia, and, in part, increases in bladder weight. CPA markedly increased phosphorylated NF-κB p65 levels in the bladder, an effect that was prevented by pyrrolidine dithiocarbamate (PDTC), an NF-κB inhibitor. PDTC and curcumin, which inhibits NF-κB signals, abolished CPA-induced nociceptive behaviour, referred hyperalgesia and, in part, increases in bladder weight. CPA caused the overexpression of CSE in the bladder, and this was prevented by PDTC or curcumin. The CPA-induced activation of NF-κB signals appeared to cause CSE overexpression in the bladder, contributing to bladder pain and in part swelling, possibly through H2 S/Cav 3.2 signaling. Therefore, NF-κB-inhibiting compounds including curcumin may be useful for the treatment of cystitis-related bladder pain.
- Published
- 2017
37. Repeated Cold Stress Enhances the Acute Restraint Stress-Induced Hyperthermia in Mice
- Author
-
Nanako Sugimoto, Seiji Ichida, Haruka Saeki, Yoshinori Funakami, Maho Tsubota, Tomoyoshi Miyamoto, Atsufumi Kawabata, Ai Nomura, and Erika Kawashita
- Subjects
0301 basic medicine ,Hyperthermia ,Male ,Restraint, Physical ,medicine.medical_specialty ,Fever ,medicine.drug_class ,Pharmaceutical Science ,Anxiolytic ,Propanolamines ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Receptors, Glucocorticoid ,Adipose Tissue, Brown ,Corticosterone ,Stress, Physiological ,Internal medicine ,Brown adipose tissue ,medicine ,Animals ,Uncoupling Protein 1 ,Pharmacology ,Diazepam ,business.industry ,Antagonist ,General Medicine ,medicine.disease ,Cold Temperature ,Mifepristone ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Anti-Anxiety Agents ,Anesthesia ,Adrenergic beta-3 Receptor Antagonists ,business ,Thermogenesis ,030217 neurology & neurosurgery ,Glucocorticoid ,Stress, Psychological ,medicine.drug - Abstract
The rodents exposed to repeated cold stress according to a specific schedule, known as specific alternation of rhythm in temperature (SART), exhibit autonomic imbalance, and is now used as an experimental model of fibromyalgia. To explore the susceptibility of SART-stressed animals to novel acute stress, we tested whether exposure of mice to SART stress for 1 week alters the extent of acute restraint stress-induced hyperthermia. Mice were subjected to 7-d SART stress sessions; i.e., the mice were alternately exposed to 24 and 4°C at 1-h intervals during the daytime (09:00-16:00) and kept at 4°C overnight (16:00-09:00). SART-stressed and unstressed mice were exposed to acute restraint stress for 20-60 min, during which rectal temperature was monitored. Serum corticosterone levels were measured before and after 60-min exposure to restraint stress. SART stress itself did not alter the body temperature or serum corticosterone levels in mice. Acute restraint stress increased the body temperature and serum corticosterone levels, both responses being greater in SART-stressed mice than unstressed mice. The enhanced hyperthermic responses to acute restraint stress in SART-stressed mice were significantly attenuated by SR59230A, a β3 adrenoceptor antagonist, but unaffected by diazepam, an anxiolytic, mifepristone, a glucocorticoid receptor antagonist, or indomethacin, a cyclooxygenase inhibitor. These results suggest that SART stress enhances the susceptibility of mice to acute restraint stress, characterized by increased hyperthermia and corticosterone secretion, and that the increased hyperthermic responses to acute stress might involve accelerated activation of sympathetic β3 adrenoceptors, known to regulate non-shivering thermogenesis in the brown adipose tissue.
- Published
- 2017
38. High glucose induces N-linked glycosylation-mediated functional upregulation and overexpression of Cav3.2 T-type calcium channels in neuroendocrine-like differentiated human prostate cancer cells
- Author
-
Shigeru Yoshida, Atsufumi Kawabata, Erina Asano, Fumiko Sekiguchi, Kazuki Fukami, and Mai Ueda
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Glycosylation ,Biology ,Neuroendocrine differentiation ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,N-linked glycosylation ,Internal medicine ,LNCaP ,medicine ,Asparagine-linked glycosylation ,Pharmacology ,lcsh:RM1-950 ,T-type calcium channel ,Cav3.2 T-type calcium channel ,medicine.disease ,Cell biology ,carbohydrates (lipids) ,030104 developmental biology ,Endocrinology ,lcsh:Therapeutics. Pharmacology ,chemistry ,Cancer cell ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
Given that Cav3.2 T-type Ca2+ channels were functionally regulated by asparagine (N)-linked glycosylation, we examined effects of high glucose on the function of Cav3.2, known to regulate secretory function, in neuroendocrine-like differentiated prostate cancer LNCaP cells. High glucose accelerated the increased channel function and overexpression of Cav3.2 during neuroendocrine differentiation, the former prevented by enzymatic inhibition of N-glycosylation and cleavage of N-glycans. Hyperglycemia thus appears to induce N-linked glycosylation-mediated functional upregulation and overexpression of Cav3.2 in neuroendocrine-like differentiated prostate cancer cells.
- Published
- 2017
- Full Text
- View/download PDF
39. Bladder pain relief by HMGB1 neutralization and soluble thrombomodulin in mice with cyclophosphamide-induced cystitis
- Author
-
Kaoru Yamaguchi, Yukari Seki, Junichi Tanaka, Toshifumi Tsujiuchi, Fumiko Sekiguchi, Akira Murai, Maho Tsubota, Atsufumi Kawabata, Hiroyasu Ishikura, and Takehiro Umemura
- Subjects
Lipopolysaccharides ,Cyclophosphamide ,Thrombomodulin ,Urinary Bladder ,Pain ,chemical and pharmacologic phenomena ,Rhodobacter sphaeroides ,Pharmacology ,HMGB1 ,Nociceptive Pain ,Mice ,Cellular and Molecular Neuroscience ,Cystitis ,Animals ,Humans ,Immunologic Factors ,Medicine ,HMGB1 Protein ,Bladder Pain ,Analgesics ,biology ,Heparin ,business.industry ,Visceral pain ,Organ Size ,Antibodies, Neutralizing ,Nociception ,Hyperalgesia ,Anesthesia ,biology.protein ,TLR4 ,Female ,Pain, Referred ,Urothelium ,medicine.symptom ,business ,medicine.drug - Abstract
High mobility group box 1 (HMGB1), one of damage-associated molecular patterns (DAMPs), plays roles in not only inflammation but also processing of somatic pain. Given that no evidence for roles of HMGB1 in visceral pain signaling is available, we asked if HMGB1 participates in bladder pain accompanying cystitis caused by cyclophosphamide in mice, using the anti-HMGB1 neutralizing antibody and recombinant human soluble thrombomodulin (rhsTM) that sequesters HMGB1 and promotes its degradation by thrombin. Cyclophosphamide, administered i.p., caused bladder pain-like nociceptive behavior and referred hyperalgesia accompanying cystitis symptoms including increased bladder weight, an indicator of edema, in mice. The cyclophosphamide-induced bladder pain and referred hyperalgesia, but not increased bladder weight, were prevented by i.p. preadministration of the anti-HMGB1 neutralizing antibody or rhsTM. HMGB1, given i.p., facilitated the bladder pain and referred hyperalgesia caused by a subeffective dose of cyclophosphamide, an effect blocked by rhsTM. In the cyclophosphamide-treated mice, HMGB1 levels greatly decreased in the bladder tissue, particularly in the urothelial cells, but did not change in the plasma. Low molecular weight heparin, known to inhibit the receptor for advanced glycation end products (RAGE), but not lipopolysaccharide from Rhodobacter sphaeroides, an inhibitor of toll-like receptor 4 (TLR4), blocked the cyclophosphamide-induced bladder pain and referred hyperalgesia. Thus, our data indicate involvement of HMGB1 in the cyclophosphamide-induced bladder pain signaling, but not cystitis itself, and suggest that targeting HMGB1 with rhsTM or blocking RAGE might serve as a novel therapeutic strategy for the management of bladder pain.
- Published
- 2014
40. Repeated Cold Stress Reduces Cyclophosphamide-Induced Cystitis/Bladder Pain and Macrophage Activity in Mice
- Author
-
Maho Tsubota, Tomoyoshi Miyamoto, Haruka Saeki, Takaya Miyazaki, Fumiko Sekiguchi, Atsufumi Kawabata, Saki Hiruma, and Yoshinori Funakami
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,medicine.medical_specialty ,Cyclophosphamide ,medicine.medical_treatment ,Urinary Bladder ,Pain ,Inflammation ,Minocycline ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Stress, Physiological ,Internal medicine ,Cystitis ,medicine ,Macrophage ,Animals ,heterocyclic compounds ,Bladder Pain ,Cold stress ,Pharmacology ,business.industry ,Macrophages ,Low dose ,General Medicine ,Macrophage Activation ,Cold Temperature ,030104 developmental biology ,Endocrinology ,Cytokine ,Immunology ,cardiovascular system ,Cytokines ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
We examined the effect of repeated cold (RC) stress on cyclophosphamide (CPA)-induced cystitis/bladder pain in mice, in relation to macrophage activity. CPA, given i.p. at 400 mg/kg, caused bladder pain symptoms accompanying cystitis in both unstressed and RC-stressed mice, which were prevented by the macrophage inhibitor minocycline. A low dose, that is, 200 mg/kg, of CPA still produced bladder pain symptoms in unstressed but not RC-stressed mice. Lipopolysaccharide-induced cytokine production in peritoneal macrophages from RC-stressed mice was less than that from unstressed mice. Thus, RC stress appears to reduce CPA-induced bladder pain in mice, which may be associated with the decreased macrophage activity.
- Published
- 2016
41. Prenylflavanones as Novel T-Type Calcium Channel Blockers Useful for Pain Therapy
- Author
-
Huy Du Nguyen, Hiroyuki Nishikawa, Atsufumi Kawabata, Maho Tsubota, Takuya Okada, Naoki Toyooka, and Fumiko Sekiguchi
- Subjects
Pharmacology ,0303 health sciences ,business.industry ,T-type calcium channel ,Plant Science ,General Medicine ,Mechanical Allodynia ,03 medical and health sciences ,0302 clinical medicine ,Complementary and alternative medicine ,Drug Discovery ,Neuropathic pain ,Medicine ,business ,030217 neurology & neurosurgery ,030304 developmental biology ,Pain therapy - Abstract
Prenylated flavonoids have attracted much attention due to their promising and diverse bioactivities on multitarget tissues. To the best of our knowledge, our recent studies demonstrated first that (2 S)-6-prenylnaringenin (6-PNG), a hop component, blocks Cav3.2 T-type calcium channels (T-channels) and alleviates neuropathic and visceral pain with little side effects; it also indicated first that other natural prenylflavanones (PFVNs), such as sophoraflavanone G and (2 S)-8-PNG, or synthetic 6-PFVNs including (2 R/S)-6-PNG and its derivatives are capable of blocking T-channels and useful for pain therapy. Through the structure-activity relationship studies on the synthetic 6-PFVNs, we identified 6-(3-ethylpent-2-enyl)-5,7-dihydroxy-2-(2-hydroxyphenyl)chroman-4-one (8j or KTt-45) as the most potent blocker of Cav3.2 T-channels. It is interesting to recognize a prenylated flavonoid, belonging to other sub-classes, as a novel T-channel blocker. Therefore, this article will review some of our recent studies to introduce a new branch to researchers studying on prenylated flavonoids.
- Published
- 2019
42. Origins and targets of HMGB1 essential for bortezomib-induced peripheral neuropathy in mice: distinct profiles in the development phase and sustained period
- Author
-
Yuya Ikeda, Shiori Tomita, Masahiro Nishibori, Takaya Miyazaki, Maho Tsubota, Atsufumi Kawabata, and Fumiko Sekiguchi
- Subjects
biology ,Bortezomib ,business.industry ,Applied Mathematics ,General Mathematics ,Period (gene) ,Pharmacology ,HMGB1 ,medicine.disease ,Peripheral neuropathy ,medicine ,biology.protein ,business ,medicine.drug - Published
- 2019
43. Regulation of Cav3.2-mediated pain signals by hydrogen sulfide
- Author
-
Maho Tsubota and Atsufumi Kawabata
- Subjects
0301 basic medicine ,Pharmacology ,Voltage-dependent calcium channel ,business.industry ,Applied Mathematics ,General Mathematics ,Visceral pain ,Endogeny ,equipment and supplies ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Nociception ,Downregulation and upregulation ,Neuropathic pain ,Neuralgia ,medicine ,Pancreatitis ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Irritable bowel syndrome - Abstract
Hydrogen sulfide (H2S), an endogenous gasotransmitter, is generated from L-cysteine by 3 distinct enzymes including cystathionine-γ-lyase (CSE), and targets multiple molecules, thereby playing various roles in health and disease. H2S triggers or accelerates somatic pain and visceral nociceptive signals in the pancreas, colon and bladder by enhancing the activity of Cav3.2 T-type calcium channels. H2S also activates TRPA1, which participates in H2S-induced somatic pain signaling. However, Cav3.2 predominantly mediates colonic nociception by H2S, because genetic deletion of TRPA1 does not reduce H2S-induced colonic pain. The functional upregulation of the CSE/H2S/Cav3.2 system is involved in neuropathic pain and visceral pain accompanying pancreatitis and cystitis. Cav3.2 also appears to participate in irritable bowel syndrome (IBS), although the role of endogenous H2S generation by CSE in IBS is still open to question. In this review, we describe how H2S regulates pain signals, particularly by interacting with Cav3.2.
- Published
- 2019
44. Recombinant human soluble thrombomodulin prevents peripheral HMGB1-dependent hyperalgesia in rats
- Author
-
Maho Tsubota, Takehiro Umemura, Atsufumi Kawabata, Akira Murai, Kaoru Yamaguchi, Hiroyasu Ishikura, Junichi Tanaka, Yukari Seki, and Fumiko Sekiguchi
- Subjects
Pharmacology ,Lipopolysaccharide ,business.industry ,chemical and pharmacologic phenomena ,Thrombomodulin ,nervous system diseases ,chemistry.chemical_compound ,medicine.anatomical_structure ,Nociception ,Dorsal root ganglion ,chemistry ,Edema ,Anesthesia ,Randall–Selitto test ,Hyperalgesia ,Systemic administration ,medicine ,medicine.symptom ,business - Abstract
Background and Purpose High-mobility group box 1 (HMGB1), a nuclear protein, is actively or passively released during inflammation. Recombinant human soluble thrombomodulin (rhsTM), a medicine for treatment of disseminated intravascular coagulation (DIC), sequesters HMGB1 and promotes its degradation. Given evidence for involvement of HMGB1 in pain signalling, we determined if peripheral HMGB1 causes hyperalgesia, and then asked if rhsTM modulates the HMGB1-dependent hyperalgesia. Experimental Approach Mechanical nociceptive threshold and swelling in rat hindpaw were determined by the paw pressure test and by measuring paw thickness, respectively, and HMGB1 levels in rat hindpaw plantar tissue, dorsal root ganglion (DRG) and serum were determined by Western blotting or elisa. Key Results Intraplantar (i.pl.) administration of HMGB1 rapidly evoked paw swelling and gradually caused hyperalgesia in rats. Systemic administration of rhsTM abolished HMGB1-induced hyperalgesia, and partially blocked paw swelling. LPS, administered i.pl., rapidly produced mild paw swelling, and gradually caused hyperalgesia. The anti-HMGB1 neutralizing antibody abolished LPS-induced hyperalgesia, but partially inhibited paw swelling. rhsTM at a high dose, 10 mg kg−1, prevented both hyperalgesia and paw swelling caused by LPS. In contrast, rhsTM at low doses, 0.001–1 mg kg−1, abolished the LPS-induced hyperalgesia, but not paw swelling. HMGB1 levels greatly decreased in the hindpaw, but not DRG. Serum HMGB1 tended to increase after i.pl. LPS in rats pretreated with vehicle, but not rhsTM. Conclusion and Implications These data suggest that peripheral HMGB1 causes hyperalgesia, and that rhsTM abolishes HMGB1-dependent hyperalgesia, providing novel evidence for therapeutic usefulness of rhsTM as an analgesic.
- Published
- 2013
45. AKAP-dependent sensitization of Cav3.2 channels via the EP4receptor/cAMP pathway mediates PGE2-induced mechanical hyperalgesia
- Author
-
Yuta Nishimoto, Maho Tsubota-Matsunami, Fumiko Sekiguchi, Yuka Aoki, Maiko Nakagawa, Rumi Yamanaka, Shigeru Yoshida, Atsufumi Kawabata, and Daiki Kanaoka
- Subjects
Pharmacology ,medicine.medical_specialty ,Voltage-dependent calcium channel ,Chemistry ,Stimulation ,KT5720 ,Cell biology ,chemistry.chemical_compound ,medicine.anatomical_structure ,Endocrinology ,Dorsal root ganglion ,Internal medicine ,Hyperalgesia ,medicine ,cAMP-dependent pathway ,medicine.symptom ,Receptor ,Sensitization - Abstract
Background and Purpose The Cav3.2 isoform of T-type Ca2+ channels (T channels) is sensitized by hydrogen sulfide, a pro-nociceptive gasotransmitter, and also by PKA that mediates PGE2-induced hyperalgesia. Here we examined and analysed Cav3.2 sensitization via the PGE2/cAMP pathway in NG108-15 cells that express Cav3.2 and produce cAMP in response to PGE2, and its impact on mechanical nociceptive processing in rats. Experimental Approach In NG108-15 cells and rat dorsal root ganglion (DRG) neurons, T-channel-dependent currents (T currents) were measured with the whole-cell patch-clamp technique. The molecular interaction of Cav3.2 with A-kinase anchoring protein 150 (AKAP150) and its phosphorylation were analysed by immunoprecipitation/immunoblotting in NG108-15 cells. Mechanical nociceptive threshold was determined by the paw pressure test in rats. Key Results In NG108-15 cells and/or rat DRG neurons, dibutyryl cAMP (db-cAMP) or PGE2 increased T currents, an effect blocked by AKAP St-Ht31 inhibitor peptide (AKAPI) or KT5720, a PKA inhibitor. The effect of PGE2 was abolished by RQ-00015986-00, an EP4 receptor antagonist. AKAP150 was co-immunoprecipitated with Cav3.2, regardless of stimulation with db-cAMP, and Cav3.2 was phosphorylated by db-cAMP or PGE2. In rats, intraplantar (i.pl.) administration of db-cAMP or PGE2 caused mechanical hyperalgesia, an effect suppressed by AKAPI, two distinct T-channel blockers, NNC 55-0396 and ethosuximide, or ZnCl2, known to inhibit Cav3.2 among T channels. Oral administration of RQ-00015986-00 suppressed the PGE2-induced mechanical hyperalgesia. Conclusion and Implications Our findings suggest that PGE2 causes AKAP-dependent phosphorylation and sensitization of Cav3.2 through the EP4 receptor/cAMP/PKA pathway, leading to mechanical hyperalgesia in rats.
- Published
- 2013
46. Antihyperalgesic Effect of Buprenorphine Involves Nociceptin/Orphanin FQ Peptide–Receptor Activation in Rats With Spinal Nerve Injury–Induced Neuropathy
- Author
-
Hiroyuki Nishikawa, Kazumasa Okubo, Maho Tsubota-Matsunami, Shota Kojima, Tomoko Takahashi, Motohide Takemura, Fumiko Sekiguchi, and Atsufumi Kawabata
- Subjects
Male ,Agonist ,medicine.drug_class ,Analgesic ,NOP ,Receptors, Opioid, mu ,Pharmacology ,Nociceptin Receptor ,medicine ,Animals ,Rats, Wistar ,Analgesics ,business.industry ,lcsh:RM1-950 ,Buprenorphine ,Rats ,Nociceptin receptor ,Spinal Nerves ,lcsh:Therapeutics. Pharmacology ,Opioid ,Receptors, Opioid ,Neuropathic pain ,Hyperalgesia ,Neuralgia ,Molecular Medicine ,medicine.symptom ,business ,medicine.drug - Abstract
We evaluated the effect of buprenorphine, a mixed agonist for μ-opioid receptors and nociceptin/orphanin FQ peptide (NOP) receptors, in neuropathic rats, using the paw pressure test. Buprenorphine, administered i.p. at 50, but not 20, μg/kg, exhibited naloxone-reversible analgesic activity in naïve rats. In contrast, buprenorphine at 0.5 – 20 μg/kg produced a naloxone-sensitive antihyperalgesic effect in the L5 spinal nerve–injured neuropathic rats. Intrathecal injection of [N-Phe1]nociceptin(1-13)NH2, a NOP-receptor antagonist, reversed the effect of buprenorphine in neuropathic rats, but not in naïve rats. Together, buprenorphine suppresses neuropathic hyperalgesia by activating NOP and opioid receptors, suggesting its therapeutic usefulness in treatment of neuropathic pain. Keywords:: buprenorphine, nociceptin/orphanin FQ peptide receptor, neuropathic pain
- Published
- 2013
47. T-type Calcium Channels: Functional Regulation and Implication in Pain Signaling
- Author
-
Fumiko Sekiguchi and Atsufumi Kawabata
- Subjects
Cell signaling ,medicine.medical_specialty ,Glycosylation ,Glutamic Acid ,Substance P ,Ascorbic Acid ,Calcium Channels, T-Type ,chemistry.chemical_compound ,Internal medicine ,medicine ,Humans ,Protein Isoforms ,Hydrogen Sulfide ,Molecular Targeted Therapy ,Neurons, Afferent ,Pharmacology ,Analgesics ,Voltage-dependent calcium channel ,Chemistry ,lcsh:RM1-950 ,Intracellular Signaling Peptides and Proteins ,Glutamate receptor ,T-type calcium channel ,Visceral pain ,Ascorbic acid ,Pain, Intractable ,Up-Regulation ,Endocrinology ,lcsh:Therapeutics. Pharmacology ,Molecular Medicine ,medicine.symptom ,Protein Kinases ,Neuroscience ,Intracellular ,Signal Transduction - Abstract
Low-voltage-activated T-type Ca2+ channels (T-channels), especially Cav3.2 among the three isoforms (Cav3.1, Cav3.2, and Cav3.3), are now considered to play pivotal roles in processing of pain signals. Cav3.2 T-channels are functionally modulated by extracellular substances such as hydrogen sulfide and ascorbic acid, by intracellular signaling molecules including protein kinases, and by glycosylation. Cav3.2 T-channels are abundantly expressed in both peripheral and central endings of the primary afferent neurons, regulating neuronal excitability and release of excitatory neurotransmitters such as substance P and glutamate, respectively. Functional upregulation of Cav3.2 T-channels is involved in the pathophysiology of inflammatory, neuropathic, and visceral pain. Thus, Cav3.2 T-channels are considered to serve as novel targets for development of drugs for treatment of intractable pain resistant to currently available analgesics. Keywords:: Cav3.2, T-type calcium channel, neuropathy, visceral pain, hydrogen sulfide
- Published
- 2013
48. Targeting Cav3.2 T-type calcium channels as a therapeutic strategy for chemotherapy-induced neuropathic pain
- Author
-
Atsufumi Kawabata
- Subjects
Pharmacology ,Paclitaxel ,Voltage-dependent calcium channel ,business.industry ,T-type calcium channel ,Peripheral Nervous System Diseases ,Antineoplastic Agents ,Ascorbic Acid ,Dinoprostone ,Calcium Channels, T-Type ,Text mining ,Chemotherapy induced ,Neuropathic pain ,Animals ,Humans ,Neuralgia ,Medicine ,Hydrogen Sulfide ,Molecular Targeted Therapy ,business ,Therapeutic strategy - Published
- 2013
49. Contribution of TRPA1 as a Downstream Signal of Proteinase-Activated Receptor-2 to Pancreatic Pain
- Author
-
Yuka Terada, Fumiko Sekiguchi, Sachiyo Nishimura, Maho Tsubota, Atsufumi Kawabata, Mayuko Fujimura, and Hiroyuki Nishikawa
- Subjects
Male ,Nociception ,medicine.medical_specialty ,TRPV1 ,TRPV Cation Channels ,Mice, Inbred Strains ,Stimulation ,Mice ,Transient Receptor Potential Channels ,Internal medicine ,Animals ,Receptor, PAR-2 ,Medicine ,Anilides ,Receptor ,TRPA1 Cation Channel ,Pharmacology ,Pancreatic duct ,business.industry ,lcsh:RM1-950 ,food and beverages ,medicine.disease ,lcsh:Therapeutics. Pharmacology ,medicine.anatomical_structure ,Endocrinology ,Pancreatitis ,nervous system ,Cinnamates ,Hyperalgesia ,Nociceptor ,Molecular Medicine ,medicine.symptom ,business ,psychological phenomena and processes ,Signal Transduction - Abstract
We examined if TRPA1, like TRPV1, contributes to pancreatic nociceptor excitation following proteinase-activated receptor-2 (PAR2) stimulation and to pancreatitis-related pain in mice. A PAR2-activating peptide, infused into the pancreatic duct, caused spinal Fos expression, which was prevented by AP18, a TRPA1 inhibitor. Repeated administration of cerulein caused referred hyperalgesia accompanying pancreatitis, which was reversed by SB366791, a TRPV1 inhibitor, but not AP18. AP18, administered in combination with a subeffective dose of SB366791, significantly suppressed the referred hyperalgesia. Our findings suggest that TRPA1, like TRPV1, mediates PAR2-triggered pancreatic nociception and that TRPA1 in collaboration with TRPV1 latently contributes to pancreatitis-related pain. Keywords:: transient receptor potential ankyrin-1 (TRPA1), proteinase-activated receptor-2 (PAR2), pancreatic pain
- Published
- 2013
50. Enhanced Hyperthermic Responses to Lipopolysaccharide in Mice Exposed to Repeated Cold Stress
- Author
-
Shiori Tomita, Nanako Sugimoto, Seiji Ichida, Tomoyoshi Miyamoto, Haruka Saeki, Yoshinori Funakami, Takaya Miyazakia, Maho Tsubota, Erika Kawashita, Atsufumi Kawabata, and Ai Nomura
- Subjects
0301 basic medicine ,Hyperthermia ,Lipopolysaccharides ,Male ,medicine.medical_specialty ,Lipopolysaccharide ,Prostaglandin ,Body Temperature ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Diclofenac ,Internal medicine ,medicine ,Animals ,Pharmacology ,Lung ,biology ,General Medicine ,medicine.disease ,Circadian Rhythm ,Cold Temperature ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Hypothalamus ,Cyclooxygenase 2 ,Immunology ,biology.protein ,Systemic administration ,lipids (amino acids, peptides, and proteins) ,Cyclooxygenase ,Corticosterone ,030217 neurology & neurosurgery ,Stress, Psychological ,medicine.drug - Abstract
Lipopolysaccharide (LPS) induces hyperthermia accompanied by various other systemic inflammatory symptoms. The rodents exposed to repeated cold (RC) stress according to a specific schedule are useful as experimental models for autonomic imbalance or fibromyalgia. It is now proven that RC-stressed mice exhibit tolerance to LPS, we examined thermal responses to LPS challenge in RC-stressed mice by monitoring core temperature using the telemetry system. Systemic administration of LPS caused bimodal hyperthermic responses in RC-stressed and unstressed mice. The magnitude of the LPS-induced hyperthermia was greater in RC-stressed mice than in unstressed mice. The RC stress-induced enhancement of hyperthermic responses to LPS was abolished by pretreatment with diclofenac, which is a cyclooxygenase (COX) inhibitor. LPS did not significantly increase COX-2 protein levels in the lung or hypothalamus of RC-stressed or unstressed mice. RC stress did not alter baseline serum corticosterone levels or their increases in response to LPS challenge. These results suggest that RC stress enhances the susceptibility of mice to LPS challenge, leading to greater prostanoid-dependent hyperthermia, which might contribute to tolerance to LPS in RC-stressed mice.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.