Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Nutr Sci Vitaminol (Tokyo) ; 70(2): 150-157, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38684385

RESUMEN

Although the energy stored in lean tissue (LT) and adipose tissue (AT) is well known, the energy required to synthesize these tissues is less clear. While elucidating the energy required for AT synthesis may not be so important, the elucidation of the energy required for LT synthesis is important for individuals who aim to increase their skeletal muscle. Theoretically the energy at the point at which ΔLT/Δbody weight (BW) reaches 100% on a regression curve, which indicates the relationship between ΔLT/ΔBW and the energy used to accumulate body tissue, is considered to be the energy expended to synthesize LT. We therefore investigated the relationship using rats. Rats of different ages, and rats in exercised or sedentary states were used because their ΔLT/ΔBW was expected to be different. ΔLT/ΔBW was higher in the 4-wk-old group than in the 8-wk-old group and higher in the exercise group than in the sedentary group. We found a positive correlation between ΔLT/ΔBW and the energy expended to synthesize tissues that accumulated in the body. This energy was lower in the 8-wk-old group, which had a lower ΔLT/ΔBW in comparison to the 4-wk-old group, but was not affected by exercise. The regression curve revealed that the energy expended to synthesize LT was 2.9 kcal/g, while that expended to synthesize AT was 1.1 kcal/g. Therefore, combined with the energy accumulated to the tissues, the energy required to accumulate LT is approximately 4.0 kcal/g, while that required to accumulate AT is approximately 8.5 kcal/g.


Asunto(s)
Tejido Adiposo , Metabolismo Energético , Músculo Esquelético , Condicionamiento Físico Animal , Animales , Tejido Adiposo/metabolismo , Masculino , Ratas , Músculo Esquelético/metabolismo , Peso Corporal , Composición Corporal , Ratas Wistar
2.
Sci Rep ; 14(1): 1039, 2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200077

RESUMEN

Given possible involvement of the central and peripheral angiotensin system in pain processing, we conducted clinical and preclinical studies to test whether pharmacological inhibition of the angiotensin system would prevent diabetic peripheral neuropathy (DPN) accompanying type 2 diabetes mellitus (T2DM). In the preclinical study, the nociceptive sensitivity was determined in leptin-deficient ob/ob mice, a T2DM model. A clinical retrospective cohort study was conducted, using the medical records of T2DM patients receiving antihypertensives at three hospitals for nearly a decade. In the ob/ob mice, daily treatment with perindopril, an angiotensin-converting enzyme inhibitor (ACEI), or telmisartan, an angiotensin receptor blocker (ARB), but not amlodipine, an L-type calcium channel blocker (CaB), significantly inhibited DPN development without affecting the hyperglycemia. In the clinical study, the enrolled 7464 patients were divided into three groups receiving ACEIs, ARBs and the others (non-ACEI, non-ARB antihypertensives). Bonferroni's test indicated significantly later DPN development in the ARB and ACEI groups than the others group. The multivariate Cox proportional analysis detected significant negative association of the prescription of ACEIs or ARBs and ß-blockers, but not CaBs or diuretics, with DPN development. Thus, our study suggests that pharmacological inhibition of the angiotensin system is beneficial to prevent DPN accompanying T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Neuropatías Diabéticas , Animales , Ratones , Humanos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico , Antihipertensivos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Neuropatías Diabéticas/prevención & control , Estudios Retrospectivos , Antivirales
3.
Biol Pharm Bull ; 46(9): 1343-1346, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37661413

RESUMEN

Cav3.2 channels belong to the T-type calcium channel (T-channel) family, i.e., low voltage-activated calcium channels, and are abundantly expressed in the nociceptors, playing a principal role in the development of pathological pain. The channel activity of Cav3.2 is suppressed by zinc under physiological conditions. We thus tested whether dietary zinc deficiency would cause Cav3.2-dependent nociceptive hypersensitivity in mice. In the mice fed with zinc deficient diet for 2 weeks, plasma zinc levels declined by more than half, and mechanical allodynia developed. The dietary zinc deficiency-induced allodynia was restored by T-channel inhibitors or by Cav3.2 gene silencing. These data demonstrate that zinc deficiency induces Cav3.2-dependent nociceptive hypersensitivity in mice, thereby suggesting that pain experienced by patients with diseases accompanied by zinc deficiency (e.g., chronic kidney disease) might involve the increased Cav3.2 activity.


Asunto(s)
Canales de Calcio Tipo T , Hipersensibilidad , Desnutrición , Animales , Ratones , Nocicepción , Zinc , Hiperalgesia/etiología , Dolor
4.
J Pharmacol Sci ; 152(2): 86-89, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37169483

RESUMEN

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


Asunto(s)
Canales de Calcio Tipo T , Hiperalgesia , Ratas , Ratones , Animales , Hiperalgesia/inducido químicamente , Roedores , Quelantes , Zinc , Canales de Calcio Tipo T/genética , Ganglios Espinales
5.
Redox Biol ; 59: 102579, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36563535

RESUMEN

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


Asunto(s)
Canales de Calcio Tipo T , Cistitis , Sulfuro de Hidrógeno , Pancreatitis , Dolor Visceral , Ratones , Humanos , Animales , Células HEK293 , Canales de Calcio Tipo T/fisiología , Sulfuros/farmacología , Cistitis/inducido químicamente , Sulfuro de Hidrógeno/metabolismo
6.
Biochem Biophys Res Commun ; 639: 142-149, 2023 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-36493557

RESUMEN

Irregular regeneration or inappropriate remodeling of the axons of the primary afferent neurons after peripheral nerve trauma could be associated with the development of neuropathic pain. We analyzed the molecular mechanisms for the neuritogenesis and neurite outgrowth caused by prostaglandin E2 (PGE2) in mouse dorsal root ganglion (DRG) neurons, and evaluated their opioid modulation. PGE2 in combination with IBMX, a phosphodiesterase inhibitor, caused neuritogenesis/neurite outgrowth in DRG cells, an effect abolished by a prostanoid EP4, but not EP2, receptor antagonist, and inhibitors of adenylyl cyclase or protein kinase A (PKA). Blockers of T-type Ca2+ channels (T-channels), that are responsible for window currents involving the sustained low-level Ca2+ entry at voltages near the resting membrane potentials and can be functionally upregulated by PKA, inhibited the neuritogenesis/neurite outgrowth caused by PGE2/IBMX or dibutylyl cyclic AMP, a PKA activator, in DRG neurons, an inhibitory effect mimicked by ZnCl2 and ascorbic acid that block Cav3.2, but not Cav3.1 or Cav3.3, T-channels. Morphine and DAMGO, µ-opioid receptor (MOR) agonists, suppressed the neuritogenesis and/or neurite outgrowth induced by PGE2/IBMX in DRG neurons and also DRG neuron-like ND7/23 cells, an effect reversed by naloxone or ß-funaltrexamine, a selective MOR antagonist. Our data suggest that the EP4 receptor/PKA/Cav3.2 pathway is involved in the PGE2-induced neuritogenesis/neurite outgrowth in DRG neurons, which can be suppressed by MOR stimulation. We propose that MOR agonists including morphine in the early phase after peripheral nerve trauma might delay the axonal regeneration of the primary afferent neurons but prevent the development of neuropathic pain.


Asunto(s)
Analgésicos Opioides , Neuralgia , Animales , Ratones , 1-Metil-3-Isobutilxantina/farmacología , Analgésicos Opioides/farmacología , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Ganglios Espinales/metabolismo , Morfina/farmacología , Neuralgia/metabolismo , Proyección Neuronal , Neuronas/metabolismo , Ratas Sprague-Dawley , Subtipo EP2 de Receptores de Prostaglandina E , Ratas
7.
Eur J Med Chem ; 243: 114716, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36075145

RESUMEN

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


Asunto(s)
Canales de Calcio Tipo T , Dolor Visceral , Ratones , Animales , Pimozida/farmacología , Pimozida/uso terapéutico , Canales de Calcio Tipo T/metabolismo , Dolor Visceral/tratamiento farmacológico , Dopamina , Bloqueadores de los Canales de Calcio/química , Receptores Dopaminérgicos/metabolismo
8.
J Pharmacol Sci ; 148(3): 315-325, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35177211

RESUMEN

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.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Oxaliplatino/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Progresión de la Enfermedad , Femenino , Proteína HMGB1/metabolismo , Humanos , Masculino , Ratones Endogámicos , Persona de Mediana Edad , Enfermedades del Sistema Nervioso Periférico/genética , Estudios Retrospectivos , Índice de Severidad de la Enfermedad
9.
J Pharmacol Sci ; 148(1): 156-161, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34924121

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/fisiología , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Neuronas/metabolismo , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos , Enfermedades del Sistema Nervioso Periférico/inmunología , Enfermedades del Sistema Nervioso Periférico/prevención & control , Células RAW 264.7 , Receptor Cross-Talk/inmunología , Receptores Purinérgicos P2X4/metabolismo , Receptores Purinérgicos P2X7/metabolismo
10.
Cells ; 10(10)2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34685531

RESUMEN

Given the role of macrophage-derived high mobility group box 1 (HMGB1) in chemotherapy-induced peripheral neuropathy (CIPN) caused by paclitaxel, we analyzed the role of HMGB1 and macrophages in the CIPN caused by bortezomib, a proteasome-inhibiting chemotherapeutic agent used for the treatment of multiple myeloma. Repeated administration of bortezomib caused CIPN accompanied by early-stage macrophage accumulation in the dorsal root ganglion. This CIPN was prevented by an anti-HMGB1-neutralizing antibody, thrombomodulin alfa capable of accelerating thrombin-dependent degradation of HMGB1, antagonists of the receptor for advanced glycation end-products (RAGE) and C-X-C motif chemokine receptor 4 (CXCR4), known as HMGB1-targeted membrane receptors, or macrophage depletion with liposomal clodronate, as reported in a CIPN model caused by paclitaxel. In macrophage-like RAW264.7 cells, bortezomib as well as MG132, a well-known proteasome inhibitor, caused HMGB1 release, an effect inhibited by caspase inhibitors but not inhibitors of NF-κB and p38 MAP kinase, known to mediate paclitaxel-induced HMGB1 release from macrophages. Bortezomib increased cleaved products of caspase-8 and caused nuclear fragmentation or condensation in macrophages. Repeated treatment with the caspase inhibitor prevented CIPN caused by bortezomib in mice. Our findings suggest that bortezomib causes caspase-dependent release of HMGB1 from macrophages, leading to the development of CIPN via activation of RAGE and CXCR4.


Asunto(s)
Antineoplásicos/efectos adversos , Bortezomib/efectos adversos , Proteína HMGB1/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Animales , Apoptosis , Modelos Animales de Enfermedad , Masculino , Ratones
11.
Cells ; 10(8)2021 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-34440650

RESUMEN

A neuroimmune crosstalk is involved in somatic and visceral pathological pain including inflammatory and neuropathic components. Apart from microglia essential for spinal and supraspinal pain processing, the interaction of bone marrow-derived infiltrating macrophages and/or tissue-resident macrophages with the primary afferent neurons regulates pain signals in the peripheral tissue. Recent studies have uncovered previously unknown characteristics of tissue-resident macrophages, such as their origins and association with regulation of pain signals. Peripheral nerve macrophages and intestinal resident macrophages, in addition to adult monocyte-derived infiltrating macrophages, secrete a variety of mediators, such as tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, high mobility group box 1 and bone morphogenic protein 2 (BMP2), that regulate the excitability of the primary afferents. Neuron-derived mediators including neuropeptides, ATP and macrophage-colony stimulating factor regulate the activity or polarization of diverse macrophages. Thus, macrophages have multitasks in homeostatic conditions and participate in somatic and visceral pathological pain by interacting with neurons.


Asunto(s)
Ganglios Espinales/metabolismo , Macrófagos/metabolismo , Neuroinmunomodulación , Neuronas/metabolismo , Umbral del Dolor , Dolor/metabolismo , Transducción de Señal , Animales , Comunicación Celular , Citocinas/metabolismo , Ganglios Espinales/inmunología , Ganglios Espinales/fisiopatología , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Neuronas/inmunología , Neuropéptidos/metabolismo , Dolor/inmunología , Dolor/fisiopatología , Fenotipo
12.
J Pharmacol Sci ; 146(1): 49-57, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33858655

RESUMEN

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 17ß-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.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Estrógenos/deficiencia , Estrógenos/fisiología , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama , Supervivientes de Cáncer , Femenino , Humanos , Ratones , Ratones Endogámicos , Persona de Mediana Edad , Ovariectomía/efectos adversos , Enfermedades del Sistema Nervioso Periférico/prevención & control , Posmenopausia , Ratas , Estudios Retrospectivos , Factores de Riesgo , Trombomodulina/administración & dosificación
13.
Biol Pharm Bull ; 44(3): 461-464, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33642557

RESUMEN

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.


Asunto(s)
Analgésicos/uso terapéutico , Bepridil/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Colitis/tratamiento farmacológico , Cistitis/tratamiento farmacológico , Antagonistas de los Receptores de Dopamina D2/uso terapéutico , Pimozida/uso terapéutico , Dolor Visceral/tratamiento farmacológico , Animales , Canales de Calcio Tipo T , Colitis/inducido químicamente , Ciclofosfamida , Cistitis/inducido químicamente , Femenino , Masculino , Ratones , Ácido Trinitrobencenosulfónico , Dolor Visceral/inducido químicamente
14.
Br J Pharmacol ; 178(4): 798-812, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32374414

RESUMEN

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.


Asunto(s)
Proteína HMGB1 , Trombomodulina , Humanos , Dolor , Receptor para Productos Finales de Glicación Avanzada , Trombina
16.
Eur J Pharmacol ; 888: 173587, 2020 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-32971090

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo T/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Ganglios Espinales/metabolismo , Proteína HMGB1/biosíntesis , Neuralgia/metabolismo , Proteasas Ubiquitina-Específicas/biosíntesis , Animales , Canales de Calcio Tipo T/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Ganglios Espinales/patología , Expresión Génica , Proteína HMGB1/genética , Vértebras Lumbares , Masculino , Neuralgia/genética , Neuralgia/patología , Ratas , Ratas Wistar , Transducción de Señal/fisiología , Nervios Espinales/lesiones , Nervios Espinales/metabolismo , Nervios Espinales/patología , Proteasas Ubiquitina-Específicas/genética
17.
Eur J Pharmacol ; 887: 173576, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32949597

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Enfermedades del Colon/inducido químicamente , Nociceptores/efectos de los fármacos , Dolor Visceral/inducido químicamente , Animales , Butiratos , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/genética , Enfermedades del Colon/genética , Sulfuro de Hidrógeno/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dimensión del Dolor/efectos de los fármacos , Dolor Visceral/genética
18.
Cells ; 9(8)2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32707767

RESUMEN

Cystitis-related bladder pain involves RAGE activation by HMGB1, and increased Cav3.2 T-type Ca2+ channel activity by H2S, generated by upregulated cystathionine-γ-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-κB 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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales de Calcio Tipo T/metabolismo , Cistitis Intersticial/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Transducción de Señal/genética , Sulfitos/metabolismo , Acroleína/metabolismo , Animales , Canales de Calcio Tipo T/genética , Ciclofosfamida/efectos adversos , Ciclofosfamida/metabolismo , Cistationina gamma-Liasa/metabolismo , Cistitis Intersticial/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Regulación hacia Arriba/genética
19.
J Pharmacol Sci ; 143(1): 60-63, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32151541

RESUMEN

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.


Asunto(s)
Inhibidores de la Calcineurina/efectos adversos , Capsaicina/efectos adversos , Colon , Hiperalgesia/inducido químicamente , Inmunosupresores/efectos adversos , Tacrolimus/efectos adversos , Animales , Hiperalgesia/genética , Síndrome del Colon Irritable/inducido químicamente , Ratones , Canales Catiónicos TRPV/agonistas
20.
J Pharmacol Sci ; 143(2): 112-116, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32222337

RESUMEN

HMGB1, a nuclear protein, once released to the extracellular space, promotes somatic and visceral pain signals. We thus analyzed the role of HMGB1 in an intravesical substance P-induced bladder pain syndrome (BPS) mouse model. Intravesical administration of substance P caused referred hyperalgesia/allodynia in the lower abdomen and hindpaw without producing severe urothelial damage, which was prevented by an anti-HMGB1-neutralizing antibody, thrombomodulin α capable of inactivating HMGB1 and antagonists of RAGE or CXCR4. The HMGB1 inactivation or RAGE blockade also reversed the established bladder pain symptoms. HMGB1 and RAGE are thus considered to serve as therapeutic targets for BPS.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Cistitis Intersticial/etiología , Cistitis Intersticial/genética , Proteína HMGB1/fisiología , Receptores Citoplasmáticos y Nucleares , Sustancia P/efectos adversos , Trombomodulina/uso terapéutico , Animales , Cistitis Intersticial/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Proteína HMGB1/inmunología , Humanos , Masculino , Ratones Endogámicos , Terapia Molecular Dirigida , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptores CXCR4/antagonistas & inhibidores , Sustancia P/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...