Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Pharmacol Res ; 185: 106490, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36216131

RESUMEN

Acquired drug resistance and epithelial-mesenchymal transition (EMT) mediated metastasis are two highly interacting determinants for non-small-cell lung cancer (NSCLC) prognosis. This study investigated the common mechanisms of drug resistance and EMT from the perspective of metabolic reprogramming, which may offer new ideas to improve anticancer therapy. Acquired resistant cells were found to grow faster and have a greater migratory and invasive capacity than their parent cells. Metabolomics analysis revealed that acquired resistant cells highly relied on glutamine utilization and mainly fluxed into oxidative phosphorylation energy production. Further mechanistic studies screened out glutamate dehydrogenase 1 (GLUD1) as the determinant of glutamine addiction in acquired resistant NSCLC cells, and provided evidence that GLUD1-mediated α-KG production and the accompanying reactive oxygen species (ROS) accumulation primarily triggered migration and invasion by inducing Snail. Pharmacological and genetic interference with GLUD1 in vitro significantly reversed drug resistance and decreased cell migration and invasion capability. Lastly, the successful application of R162, a selective GLUD1 inhibitor, to overcome both acquired resistance and EMT-induced metastasis in vivo, identified GLUD1 as a promising and druggable therapeutic target for malignant progression of NSCLC. Collectively, our study offers a potential strategy for NSCLC therapy, especially for drug-resistant patients with highly expressed GLUD1.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/uso terapéutico , Glutamina/metabolismo , Glutamina/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Factores de Transcripción de la Familia Snail/metabolismo
2.
Neurochem Res ; 45(10): 2258-2277, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32794152

RESUMEN

Stroke is the fifth leading cause of death worldwide and is a main cause of disability in adults. Neither currently marketed drugs nor commonly used treatments can promote nerve repair and neurogenesis after stroke, and the repair of neurons damaged by ischemia has become a research focus. This article reviews several possible mechanisms of stroke and neurogenesis and introduces novel neurogenic agents (fibroblast growth factors, brain-derived neurotrophic factor, purine nucleosides, resveratrol, S-nitrosoglutathione, osteopontin, etc.) as well as other treatments that have shown neuroprotective or neurogenesis-promoting effects.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/etiología , Humanos , Neuronas/metabolismo , Transducción de Señal/fisiología , Accidente Cerebrovascular/etiología
3.
J Exp Clin Cancer Res ; 41(1): 163, 2022 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-35501907

RESUMEN

BACKGROUND: Inevitably developed resistance of the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) limited its clinical benefit on non-small cell lung cancer (NSCLC). Upfront combination therapy is promising to prevent this resistance. Compelling clinical evidence indicated the failure of third-generation EGFR TKIs combined with either immunotherapy or antiangiogenic agents. In comparison, combined treatment of third-generation EGFR TKIs and chemotherapy might be a favorable choice. Herein, we systematically analyzed and compared the effects of pemetrexed and a novel third-generation EGFR TKI aumolertinib combined in different sequences, subsequently revealed the potential mechanisms and proved the optimal combination schedule with clinical retrospective study. METHODS: Three combination schedules involving pemetrexed and aumolertinib in different sequences were developed. Their inhibition effects on cell proliferation and metastasis were firstly compared upon three human NSCLC cell lines in vitro, by cell counting kit-8, colony formation, wound healing and transwell assays respectively. Further evaluation in vivo was proceeded upon H1975 and HCC827 xenograft model. Gene and protein expression were detected by Q-PCR and western blot. Drug concentration was determined by LC-MS/MS. VEGF secretion was determined by ELISA. Tumor vessel was visualized by immunofluorescence. Lastly, a clinical retrospective study was raised with 65 patients' data. RESULTS: The combination of pemetrexed and aumolertinib exhibited a sequence-dependent and EGFR mutant-dependent synergistic effect in vitro and in vivo. Only treatment with aumolertinib following pemetrexed (P-A) exhibited synergistic effect with stronger anti-tumor growth and anti-metastasis ability than monotherapy and also other combination sequences. This synergism could exclusively be observed in H1975 and HCC827 but not A549. Pathway analysis showed that P-A significantly enhanced the suppression of EGFR pathway. In addition, our results intriguingly found an obvious reduction of VEGF secretion and the accompanying normalization of the intratumor vessel, consequently increasing intratumoral accumulation of pemetrexed in P-A group. Finally, the clinical retrospective study verified the synergistic effect of P-A combination by significantly superior tumor response than aumolertinib monotherapy. CONCLUSION: Aumolertinib-pemetrexed combined therapy is promising for EGFR mutant NSCLC but only in right administration sequence. P-A could become an advantageous combination strategy in clinical with synergistic inhibition of tumor growth and metastasis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Pemetrexed , Acrilamidas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Cromatografía Liquida , Receptores ErbB/metabolismo , Humanos , Indoles/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Pemetrexed/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Estudios Retrospectivos , Espectrometría de Masas en Tándem , Factor A de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Brain Res Bull ; 165: 146-160, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33065175

RESUMEN

Stroke is one of the leading causes of death worldwide with limited therapies. After ischemic stroke occurs, a robust sterile inflammatory response happens and lasts for days and determines neurological prognosis. Pyroptosis is an inflammatory programmed cell death characterized by cleavage of pore-forming proteins gasdermins as a result of activating caspases and inflammasomes. It has morphological characteristics of rapid plasma-membrane rupture and release of proinflammatory intracellular contents as well as cytokines. Recent researches implicate pyroptosis involvement in the pathogenesis of ischemic stroke and inhibition of pyroptosis attenuates ischemic brain injury. In this review, we discussed molecular mechanisms of pyroptosis, evidences for pyroptosis involvement in different kinds of the central nervous system cells, as well as potential inhibitors for intervention of pyroptosis. Based on the review, we hypothesize the feasibility of therapeutic strategies targeting pyroptosis in the context of ischemic stroke.


Asunto(s)
Encéfalo/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Piroptosis/fisiología , Animales , Encéfalo/patología , Caspasas/metabolismo , Accidente Cerebrovascular Isquémico/patología
5.
Curr Drug Targets ; 21(3): 288-301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31490748

RESUMEN

BACKGROUND: Some of the current challenges and complications of cancer therapy are chemotherapy- induced peripheral neuropathy (CIPN) and the neuropathic pain that are associated with this condition. Many major chemotherapeutic agents can cause neurotoxicity, significantly modulate the immune system and are always accompanied by various adverse effects. Recent evidence suggests that cross-talk occurs between the nervous system and the immune system during treatment with chemotherapeutic agents; thus, an emerging concept is that neuroinflammation is one of the major mechanisms underlying CIPN, as demonstrated by the upregulation of chemokines. Chemokines were originally identified as regulators of peripheral immune cell trafficking, and chemokines are also expressed on neurons and glial cells in the central nervous system. OBJECTIVE: In this review, we collected evidence demonstrating that chemokines are potential mediators and contributors to pain signalling in CIPN. The expression of chemokines and their receptors, such as CX3CL1/CX3CR1, CCL2/CCR2, CXCL1/CXCR2, CXCL12/CXCR4 and CCL3/CCR5, is altered in the pathological conditions of CIPN, and chemokine receptor antagonists attenuate neuropathic pain behaviour. CONCLUSION: By understanding the mechanisms of chemokine-mediated communication, we may reveal chemokine targets that can be used as novel therapeutic strategies for the treatment of CIPN.


Asunto(s)
Antineoplásicos/efectos adversos , Quimiocinas/metabolismo , Quimiocinas/fisiología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Animales , Sistema Nervioso Central/metabolismo , Humanos , Ratones , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/inmunología , Síndromes de Neurotoxicidad/etiología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/inmunología , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/fisiología , Transducción de Señal/efectos de los fármacos
6.
Neurotherapeutics ; 17(1): 340-355, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31617070

RESUMEN

Antimicrotubulin chemotherapeutic agents, including plant-derived vincaalkaloids such as vincristine, can cause peripheral neuropathic pain. Exogenously activated heme oxygenase 1 (HO-1) is a potential therapy for chemotherapy-induced neuroinflammation. In this study, we investigated a role for Nrf2/HO-1/CO in mediating vincristine-induced neuroinflammation by inhibiting connexin 43 (Cx43) production in the spinal cord following the intrathecal application of the HO-1 inducer protoporphyrin IX cobalt chloride (CoPP) or inhibitor protoporphyrin IX zinc (ZnPP), and we analyzed the underlying mechanisms by which levo-corydalmine (l-CDL, a tetrahydroprotoberberine) attenuates vincristine-induced pain. Treatment with levo-corydalmine or oxycodone hydrochloride (a semisynthetic opioid analgesic, used as a positive control) attenuated vincristine-induced persistent pain hypersensitivity and degeneration of the sciatic nerve. In addition, the increased prevalence of atypical mitochondria induced by vincristine was ameliorated by l-CDL in both A-fibers and C-fibers. Next, we evaluated whether nuclear factor E2-related factor 2 (Nrf2), an upstream activator of HO-1, directly bound to the HO-1 promoter sequence and degraded heme to produce carbon monoxide (CO) following stimulation with vincristine. Notably, l-CDL dose-dependently increased HO-1/CO expression by activating Nrf2 to inhibit Cx43 expression in both the spinal cord and in cultured astrocytes stimulated with TNF-α, corresponding to decreased Cx43-mediated hemichannel. Furthermore, l-CDL had no effect on Cx43 following the silencing of the HO-1 gene. Taken together, our findings reveal a novel mechanism by which Nrf2/HO-1/CO mediates Cx43 expression in vincristine-induced neuropathic pain. In addition, the present findings suggest that l-CDL likely protects against nerve damage and attenuates vincristine-induced neuroinflammation by upregulating Nrf2/HO-1/CO to inhibit Cx43 expression.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Berberina/análogos & derivados , Conexina 43/metabolismo , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Transducción de Señal/efectos de los fármacos , Vincristina/toxicidad , Animales , Astrocitos/efectos de los fármacos , Berberina/administración & dosificación , Encefalitis/inducido químicamente , Encefalitis/complicaciones , Hemo-Oxigenasa 1/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos ICR , Factor 2 Relacionado con NF-E2/metabolismo , Nervio Ciático/efectos de los fármacos , Nervio Ciático/ultraestructura , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Regulación hacia Arriba
7.
Neuroscience ; 418: 189-204, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31487541

RESUMEN

JLX001, a novel compound with similar structure with cyclovirobuxine D (CVB-D), has been proved to exert therapeutical effects on permanent focal cerebral ischemia. However, the protective effects of JLX001 on cerebral ischemia/reperfusion (I/R) injury and its anti-apoptotic effects have not been reported. We investigated the efficacy of JLX001 in two pharmacodynamic tests (pre-treatment test and post-treatment) with rats subjected to middle cerebral artery occlusion/reperfusion (MCAO/R). The pharmacodynamic tests demonstrated that JLX001 ameliorated I/R injury by reducing infarct sizes and brain edema. The results of Morris water maze, neurological scores, cylinder test and posture reflex test implied that JLX001 improved the learning, memory and motor ability after MCAO/R in the long term. Anti-apoptotic effects of JLX001 and its regulation of cytosolic c-Jun N-terminal Kinases (JNKs) signal pathway were confirmed in vivo by co-immunofluorescence staining and western immunoblotting. Furthermore, primary cortical neuron cultures were prepared and exposed to oxygen glucose deprivation/reoxygenation (OGD/R) for in vitro studies. Cytotoxicity test and mitochondrial membrane potential (MMP) test showed that JLX001 enhanced cell survival rate and maintained MMP. Flow cytometry and TdT-mediated dUTP-X nick end labeling (TUNEL) staining demonstrated the anti-apoptotic effects of JLX001 in vitro. Likewise, JLX001 regulated JNK signal pathway in vivo, which was also confirmed by western immunoblotting. Collectively, this study presents the first evidence that JLX001 exerted protective effects against I/R injury by reducing neuronal apoptosis via down-regulating JNK signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Triterpenos/farmacología , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Masculino , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Brain Res Bull ; 153: 162-170, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31472184

RESUMEN

(3ß,5α,16α,20S)-4,4,14-trimethyl-3,20-bis(methylamino)-9,19-cyclopregnan-16-ol-dihydrochloride (JLX-001), a structural analogue of cyclovirobuxine D (CVB-D), is a novel compound from synthesis. This study aims to confirm the therapeutic effects of JLX001 on ischemic stroke (IS) and research its induction of autophagy function via 5'-AMP-activated protein kinase (AMPK)-Human Serine/threonine-protein kinase (ULK1) signaling pathway activation. The therapeutic effects of JLX001 were evaluated by infarct sizes, brain edema, neurological scores and proportion of apoptotic neurons in Sprague-Dawley (SD) rats with middle cerebral artery occlusion/reperfusion (MCAO/R). The number of autophagosomes was obtained by transmission electron microscopy. The expression of LC3-II was measured by immunofluorescence. p-AMPK and activated ULK1 were detected by western blots. Results showed that JLX001 treatment markedly alleviated cerebral infarcts, edema, neurological scores and proportion of apoptotic neurons in MCAO/R rats. The number of autophagosomes was increased, accompanying with the increased expressions of LC3-II, p-AMPK and ULK1. In summary, JLX001 attenuates cerebral ischemia injury and the underlying mechanisms may relate to inducing autophagy via AMPK-ULK1 signaling pathway activation.


Asunto(s)
Autofagia/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Triterpenos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/fisiología , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Edema Encefálico , Infarto de la Arteria Cerebral Media , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Serina-Treonina Quinasas TOR/metabolismo , Triterpenos/uso terapéutico
9.
RSC Adv ; 9(31): 17591, 2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-35532405

RESUMEN

[This corrects the article DOI: 10.1039/C7RA08879E.].

10.
Neuropharmacology ; 135: 34-47, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29518397

RESUMEN

Vincristine is a commonly used chemotherapeutic drug that can produce painful peripheral neuropathy. The chemokine (C-X-C motif) ligand 1 (CXCL1) and its receptor chemokine (C-X-C motif) receptor 2 (CXCR2) may mediate the resolution of this inflammation. In this study, we investigated whether and how CXCL1 contributes to vincristine-induced pain and the underlying mechanisms of levo-corydalmine (l-CDL, a tetrahydroprotoberberine). Oxycodone hydrochloride (a semisynthetic opioid analgesic) was used as positive control in vivo experiments. The results revealed that both l-CDL and oxycodone attenuated vincristine-induced persistent pain hypersensitivity and proinflammatory factors release in mice. CXCL1 and CXCR2 were increased from 6 to 14 days after vincristine administration in the spinal cord. In addition, vincristine injection induced the phosphorylation of NFκB by activating p65/RelA. To confirm these results, we demonstrated that l-CDL controlled astrocytic-released CXCL1 by inhibiting p65/RelA activation, thus acting on the CXCR2 receptor in the spinal cord. In cultured astrocytes, TNF-α elicited marked release of the chemokine CXCL1; moreover, the release was blocked by NFκB p65 small interfering RNA, NFκB inhibitor, and was dose-dependently decreased by l-CDL. However, l-CDL had no effect on CXCL1 in response to NFκB p65-silenced. In primary neurons, l-CDL indirectly reduced an increase in CXCR2 by astrocyte-conditioned medium but did not act directly on the CXCR2 site. Taken together, our data first demonstrate that an NFκB-dependent CXCL1/CXCR2 signaling pathway is involved in vincristine-induced neuropathic pain. In addition, the present findings suggest that l-CDL likely attenuates this inflammation through down-regulation of this signaling pathway.


Asunto(s)
Berberina/análogos & derivados , Quimiocina CXCL1/metabolismo , FN-kappa B/metabolismo , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Receptores de Interleucina-8B/metabolismo , Transducción de Señal/efectos de los fármacos , Vincristina/farmacología , Animales , Astrocitos/metabolismo , Berberina/uso terapéutico , Células Cultivadas , Relación Dosis-Respuesta a Droga , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Neuronas/metabolismo , Oxicodona/uso terapéutico , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño , Factor de Transcripción ReIA/antagonistas & inhibidores
11.
J Mol Neurosci ; 66(3): 342-355, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30276612

RESUMEN

Ischemic stroke is one of the leading health issues and the major cause of permanent disability in adults worldwide. Energy depletion and hypoxia occurring after ischemic stroke result in cell death, which activates resident glia cells and promotes the peripheral immune cells breaching into brain performing various functions even contradictory effects. The infiltration of immune cells may mediate neuron apoptosis and escalate ischemic damage, while it enhances neuron repair, differentiation, and neuroregeneration. The central nervous system (CNS) is immune-privileged site as it is separated from the peripheral immune system by the blood-brain barrier (BBB). Pathologically, the diapedesis of peripheral immune cells to CNS is controlled by BBB and regulated by immune cells/endothelial interactions. As immune responses play a key role in modulating the progression of ischemic injury development, understanding the characteristics and the contribution on regulating inflammatory responses of glia cells and peripheral immune cells may provide novel approaches for potential therapies. This review summarizes the multistep process of periphery immune cell extravasation into brain parenchyma during immunosurveillance and chronic inflammation after ischemic stroke onset. Furthermore, the review highlights promising target intervention, which may promote the development of future therapeutics for ischemic stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Linfocitos/inmunología , Neuroglía/inmunología , Accidente Cerebrovascular/inmunología , Animales , Barrera Hematoencefálica/metabolismo , Movimiento Celular , Humanos
12.
Biomed Pharmacother ; 106: 805-812, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29990874

RESUMEN

(3ß,5α,16α,20S)-4,4,14-trimethyl-3,20-bis(methylamino)-9,19-cyclopregnan-16-ol-dihydrochloride (JLX001), a derivative of cyclovirobuxine D (CVB-D), is a novel compound from synthesis. This study aims to confirm the therapeutic effect of JLX001 on cerebral ischemia and researchits antiplatelet and antithrombosis activities via thromboxane (TXA2)/phospholipase C-ß-3(PLCß3)/protein kinase C (PKC) pathway suppression. The therapeutic effects of JLX001 was evaluated by infarct sizes, brain edema and neurological scores in Sprague-Dawley (SD) rats with middle cerebral artery occlusion (MCAO). Brain TXA2 and prostacyclin (PGI2) were measured by enzyme-linked immunosorbentassay (ELISA). P-PLCß3and activated PKC were detected by immunohistochemical method. Adenosine diphosphate (ADP) or 9, 11-dieoxy-11α, 9α-epoxymethanoeprostaglandin F2α (U46619) was used as platelet agonist in the in vivo and in vitro platelet aggregation experiments. Clotting time and bleeding time were determined. Besides, two whole-animal experiments including arteriovenous shunt thrombosis and pulmonary thromboembolism model were conducted. Results showed that JLX001 treatment markedly alleviated cerebral infarcts, edema, and neurological scores in permanent middle cerebral artery occlusion (pMCAO) rats. Brain TXA2 level, p-PLCß3and activated PKC were decreased, while PGI2level had no significant change. Besides, JLX001 inhibited platelet aggregation induced by ADP or U46619 and exhibited anti-coagulation effects with a minor bleeding risk. In the two whole-animal experiments, JLX001 inhibited thrombus formation. In summary, JLX001 attenuates cerebral ischemia injury and the underlying mechanisms relate to inhibiting platelet activation and thrombus formation via TXA2/PLCß3/PKC pathway suppression.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Infarto de la Arteria Cerebral Media/prevención & control , Trombosis Intracraneal/prevención & control , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Triterpenos/farmacología , Animales , Aspirina/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Edema Encefálico/sangre , Edema Encefálico/patología , Edema Encefálico/prevención & control , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Epoprostenol/metabolismo , Femenino , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/patología , Trombosis Intracraneal/sangre , Trombosis Intracraneal/enzimología , Trombosis Intracraneal/patología , Masculino , Ratones Endogámicos ICR , Fosfolipasa C beta/metabolismo , Inhibidores de Agregación Plaquetaria/uso terapéutico , Proteína Quinasa C/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tromboxano A2/metabolismo , Triterpenos/uso terapéutico
13.
Life Sci ; 190: 68-77, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28964813

RESUMEN

Neuropathic pain is maladaptive pain caused by injury or dysfunction in peripheral and central nervous system, and remains a worldwide thorny problem leading to decreases in physical and mental quality of people's life. Currently, drug therapy is the main treatment regimen for resolving pain, while effective drugs are still unmet in medical need, and commonly used drugs such as anticonvulsants and antidepressants often make patients experience adverse drug reactions like dizziness, somnolence, severe headache, and high blood pressure. Thus, in this review we overview the anatomical physiology, underlying mechanisms of neuropathic pain to provide a better understanding in the initiation, development, maintenance, and modulation of this pervasive disease, and inspire research in the unclear mechanisms as well as potential targets. Furthermore, we summarized the existing drug therapies and new compounds that have shown antalgic effects in laboratory studies to be helpful for rational regimens in clinical treatment and promotion in novel drug discovery.


Asunto(s)
Analgésicos/uso terapéutico , Diseño de Fármacos , Neuralgia/tratamiento farmacológico , Analgésicos/efectos adversos , Analgésicos/farmacología , Animales , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Antidepresivos/efectos adversos , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Descubrimiento de Drogas/métodos , Humanos , Neuralgia/fisiopatología , Calidad de Vida
14.
Eur J Pharmacol ; 810: 112-119, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28629736

RESUMEN

1-(5-(1H-indol-5-yl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl)-N-methylmethanamine (KFP-H008),a novel and potent potassium-competitive acid blocker for the treatment of acid secretion related diseases, has not been reported previously. In this study, we demonstrated that KFP-H008 inhibits basal acid secretion, 2-deoxy-D-glucose- (2DG-) stimulated gastric acid secretion in rats. KFP-H008 blocked histamine-stimulated acid secretion in rats and heidenhain pouch dogs and reversed acid output in isolated gastric perfusion under histamine stimulation. In all the animal experiments, KFP-H008 exerted a more effective, potent and longer-lasting inhibitory action in comparison with lansoprazole, a proton pump inhibitor (PPI) commonly used in clinic. KFP-H008 inhibited H+-K+-ATPase activity both at pH 6.5 and pH 7.5, and was unaffected by pH. The inhibitory action was reversible and was achieved in a K+-competitive manner. Furthermore, KFP-H008 did not affect Na+-K+-ATPase activity, thus exhibiting high selectivity, which is different from PPIs. In all, KFP-H008, a novel potassium-competitive acid blocker, may provide new option for the patients with acid-related diseases and provide longer-lasting inhibitory action than drugs commonly used in clinical treatment.


Asunto(s)
Ácido Gástrico/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Indoles/farmacología , Inhibidores de la Bomba de Protones/farmacología , Piridinas/farmacología , Pirroles/farmacología , Animales , Histamina/farmacología , Masculino , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA