Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Biomolecules ; 11(8)2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34439742

RESUMEN

Angiotensin II (Ang II) induces vasoconstriction through myosin light chain (MLC) kinase activation and MLC phosphatase inactivation via phosphorylation of myosin phosphatase targeting subunit 1 (MYPT1) by Rho kinase. However, the detailed mechanism underlying Rho kinase activation by Ang II is still unknown. We investigated the mechanism of Ang II-induced vasoconstriction mediated by Rho kinase in pressure-overloaded rat thoracic aortas. Pressure-overloaded rats were produced by coarctation of the suprarenal abdominal aorta in four-week-old male Wistar rats. The contractile response to Ang II was significantly enhanced in the pressure-overloaded rats. Ang II-induced vasoconstriction was attenuated by inhibitors of Rho kinase, extracellular signal-regulated kinase 1 and 2 (Erk1/2), and epidermal growth factor receptor (EGFR) in both the sham-operated and pressure-overloaded rats. The Ang II-induced vasoconstriction was attenuated by a Janus kinase 2 (JAK2) inhibitor in only the pressure-overloaded rats. The protein levels of MYPT1 and JAK2 increased only in the pressure-overloaded rat thoracic aortas. These results suggested that Ang II-induced contraction is mediated by Rho kinase activation via EGFR, Erk1/2, and JAK2 in pressure-overloaded rat thoracic aortas. Moreover, Ang II-induced contraction was enhanced in pressure-overloaded rats probably because the protein levels of MYPT1 and JAK2 increased in the thoracic aortas.


Asunto(s)
Angiotensina II/fisiología , Aorta Torácica/metabolismo , Vasoconstricción , Quinasas Asociadas a rho/metabolismo , Animales , Aorta Torácica/patología , Masculino , Ratas , Ratas Wistar
3.
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
4.
Biol Pharm Bull ; 42(6): 877-885, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31155587

RESUMEN

Orthovanadate (OVA), a protein tyrosine phosphatase inhibitor, induces contraction in endothelium-denuded mouse thoracic aortas. OVA-induced contraction was significantly (vs. control rings) suppressed by Rho kinase (Y-27632, 10 µM), extracellular signal-regulated kinase 1 and 2 (Erk1/2, FR180204, 10 µM), Erk1/2 kinase (MEK, PD98059, 10 µM), epidermal growth factor receptor (EGFR, AG1478, 10 µM), and Src inhibitors, and was partially suppressed by c-Jun N-terminal kinase (JNK, AS601245, 10 µM) and p38 (SB203580, 10 µM) inhibitors. However, a myosin light chain kinase inhibitor (ML-7, 10 µM) and a metalloproteinase inhibitor (TAPI-0, 10 µM) had no effect on OVA-induced contraction in mouse thoracic aortas. Phosphorylation of myosin phosphatase target subunit 1 (MYPT1) was abolished by inhibitors of Src, EGFR, MEK, Erk1/2, and Rho kinase, but not by inhibitors of JNK and p38. Erk1/2 phosphorylation by OVA was blocked by inhibitors of EGFR, Src, MEK, and Erk1/2, but not by Rho kinase inhibition. Src phosphorylation at Tyr-416 was abrogated by only Src inhibitor. EGFR phosphorylation at Tyr-1173 was suppressed by a Src inhibitor. These findings suggest that OVA induces contraction via activation of Src, EGFR, MEK, Erk1/2, and Rho kinase, leading to inactivation of myosin light chain phosphatase via MYPT1 phosphorylation.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Proteínas Quinasas/fisiología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Vanadatos/farmacología , Animales , Aorta Torácica/fisiología , Masculino , Ratones , Vasoconstricción/efectos de los fármacos
5.
Int J Clin Pharmacol Ther ; 57(6): 315-322, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30900981

RESUMEN

OBJECTIVE: This study aimed to compare a novel point-of-care assay that involves a flap endonuclease reaction performed using GTS-7000® to a conventional assay that involves DNA sequencing performed using 3130xl Genetic Analyzers*. MATERIALS AND METHODS: This study enrolled 74 patients who underwent heart transplantation at the National Cerebral and Cardiovascular Center between May 2004 and October 2016. Each patient was genotyped as cytochrome P450 (CYP) 3A5*1/*1, -CYP3A5*1/*3, or CYP3A5*3/*3. Quantitative and qualitative comparison between the two assays was carried out. RESULTS: Four patients were genotyped as CYP3A5*1/*1, 25 as CYP3A5*1/*3, and 45 as CYP3A5*3/*3. Genotyping results of the point-of-care method were completely consistent with those of the conventional method. The total analysis time of the point-of-care method was shorter than that of the conventional method (~ 1.5 vs. 7.5 h). However, the cost of the point-of-care method was higher than that of the conventional method (~ 21 vs. 17 US$). CONCLUSION: Compared with a laboratory-based assay, the point-of-care assay that utilizes GTS-7000® is accurate and rapid despite being slightly more expensive. Further trials using this assay are warranted.


Asunto(s)
Citocromo P-450 CYP3A/genética , Genotipo , Trasplante de Corazón , Inmunosupresores/uso terapéutico , Humanos , Pruebas en el Punto de Atención , Polimorfismo Genético
6.
Eur J Clin Pharmacol ; 75(1): 67-75, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30251062

RESUMEN

PURPOSE: This study aimed to investigate the effects of clotrimazole on the pharmacokinetics of tacrolimus in Japanese patients with heart transplants with different CYP3A5 genotypes. METHODS: Twenty-six patients who underwent heart transplantation between June 2012 and July 2017 were enrolled in this retrospective study. The CYP3A5 (rs776746; CYP3A5*3) genotype was determined after monitoring and analysing tacrolimus blood concentrations. The pharmacokinetic profile of tacrolimus was examined before and after the discontinuation of clotrimazole and in patients with different CYP3A5 genotypes. RESULTS: The CYP3A5*1/*1, *1/*3 and *3/*3 genotypes were detected in 2, 8 and 16 patients, respectively. After clotrimazole was discontinued, the CYP3A5 expresser (CYP3A5*1/*1 or *1/*3) group had a 3.3-fold median increase in apparent oral clearance of tacrolimus (0.27 vs. 0.89 L/h/kg, P = 0.002) compared with the CYP3A5 non-expresser (CYP3A5*3/*3) group with a 2.2-fold median increase (0.18 vs. 0.39 L/h/kg, P < 0.0001). Significant correlations were observed between C0 and area under the concentration-time curve (AUC0-12) of tacrolimus after the discontinuation of clotrimazole in the CYP3A5 expresser and non-expresser groups, respectively (R2 = 0.49 and 0.42, all P < 0.05), but not before the discontinuation of clotrimazole. CONCLUSION: The effects of clotrimazole on tacrolimus pharmacokinetics in the CYP3A5 expresser patients were significantly greater than those in the CYP3A5 non-expresser patients. In addition, clotrimazole disturbed the correlation between C0 and AUC0-12 of tacrolimus. Careful dose adjustment of tacrolimus based on CYP3A5 genotypes may be beneficial for the patients with heart transplants who are concomitantly treated with clotrimazole.


Asunto(s)
Clotrimazol/administración & dosificación , Citocromo P-450 CYP3A/genética , Trasplante de Corazón , Tacrolimus/administración & dosificación , Adulto , Antifúngicos/administración & dosificación , Antifúngicos/farmacología , Área Bajo la Curva , Pueblo Asiatico , Clotrimazol/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Monitoreo de Drogas/métodos , Femenino , Genotipo , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tacrolimus/farmacocinética
7.
Int J Clin Pharmacol Ther ; 56(6): 270-276, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29648533

RESUMEN

OBJECTIVE: Everolimus is an inhibitor of the mammalian target of rapamycin (mTOR) and has been used in combination with calcineurin inhibitors (tacrolimus and cyclosporine) to prevent allograft rejection following organ transplantation. In heart transplant recipients, everolimus should be maintained at a target blood concentration of 3 - 8 ng/mL, in combination with reduced-dose calcineurin inhibitors and therefore, requires strict monitoring. Fluconazole, an azole antifungal agent, affects blood concentration of tacrolimus by inhibiting the cytochromes P450 (CYP) 3A4 and 3A5. Therefore, to avoid overexposure during everolimus-azole cotreatment, the dose of everolimus should be reduced. However, the mechanism of interaction between everolimus and fluconazole remains unclear. CASE REPORT: We report the case of a heart transplant recipient who experienced a 2.8-fold increase in everolimus clearance and a 3.5-fold increase in everolimus dosage, following withdrawal of fluconazole therapy. The clearance and dosage of tacrolimus increased 4.7- and 3.0-fold, respectively. CONCLUSION: The concentrations of everolimus and tacrolimus should be carefully monitored when administered concomitantly with fluconazole to heart transplant recipients. The patient in this case had a CYP3A5*1/*3 genotype, and CYP3A5 constituted the metabolic pathway. Therefore, concomitant use of fluconazole might have a relatively small impact on everolimus and tacrolimus pharmacokinetics in this case.
.


Asunto(s)
Antifúngicos/farmacología , Everolimus/farmacocinética , Fluconazol/farmacología , Trasplante de Corazón , Inmunosupresores/farmacocinética , Tacrolimus/farmacocinética , Inhibidores del Citocromo P-450 CYP3A/farmacología , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
8.
Eur J Drug Metab Pharmacokinet ; 43(6): 665-673, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29691732

RESUMEN

BACKGROUND AND OBJECTIVE: Tacrolimus, a major immunosuppressant used after transplantation, is associated with large interindividual variation involving genetic polymorphisms in metabolic processes. A common variant of the cytochrome P450 (CYP) 3A5 gene, CYP3A5*3, affects blood concentrations of tacrolimus. However, tacrolimus pharmacokinetics at the early stage of transplantation have not been adequately studied in heart transplantation. We retrospectively examined the impact of the CYP3A5 genotype on tacrolimus pharmacokinetics at the early stage of heart transplantation. METHODS: The tacrolimus pharmacokinetic profile was obtained from 65 patients during the first 5 weeks after heart transplantation. Differences in the patients' characteristics and tacrolimus pharmacokinetic parameters between the CYP3A5 expresser (*1/*1 or *1/*3 genotypes) and non-expresser (*3/*3 genotype) groups were assessed by the Chi-square test, Student's t test, or Mann-Whitney U test. RESULTS: The CYP3A5 *1/*1, *1/*3, and *3/*3 genotypes were detected in 5, 22, and 38 patients, respectively. All patients started clotrimazole therapy approximately 1 week after starting tacrolimus. Apparent clearance and dose/weight to reach the target trough concentration (C0) were significantly higher in the expresser group than in the non-expresser group (0.32 vs. 0.19 L/h/kg, p = 0.0003; 0.052 vs. 0.034 mg/kg/day, p = 0.0002); there were no significant differences in the area under the concentration-time curve from 0 to 12 h (AUC0-12) and concentrations at any sampling time point between the two groups. CONCLUSION: Similar concentration-time curves for tacrolimus were obtained in the expresser and non-expresser groups by dose adjustment based on therapeutic drug monitoring. These results demonstrate the importance of the CYP3A5 genotype in tacrolimus dose optimization based on therapeutic drug monitoring after heart transplantation.


Asunto(s)
Alelos , Citocromo P-450 CYP3A/genética , Trasplante de Corazón , Tacrolimus/farmacocinética , Adulto , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Femenino , Expresión Génica/genética , Genotipo , Humanos , Inmunosupresores/sangre , Inmunosupresores/farmacocinética , Masculino , Persona de Mediana Edad , Polimorfismo Genético/genética , Estudios Retrospectivos , Tacrolimus/sangre
9.
Appl Microbiol Biotechnol ; 102(7): 3159-3171, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29468297

RESUMEN

Membrane-bound, pyrroloquinoline quinone (PQQ)-dependent glycerol dehydrogenase (GLDH, or polyol dehydrogenase) of Gluconobacter sp. oxidizes various secondary alcohols to produce the corresponding ketones, such as oxidation of D-sorbitol to L-sorbose in vitamin C production. Substrate specificity of GLDH is considered limited to secondary alcohols in the D-erythro configuration at the next to the last carbon. Here, we suggest that L-ribose, D- and L-lyxoses, and L-tagatose are also substrates of GLDH, but these sugars do not meet the substrate specificity rule of GLDH. The oxygen consumption activity of wild-type Gluconobacter frateurii cell membranes depends on several kinds of sugars as compared with that of the membranes of a GLDH-negative variant. Biotransformation of those sugars with the membranes was examined to determine the reaction products. A time course measuring the pH in the reaction mixture and the increase or decrease in substrates and products on TLC suggested that oxidation products of L-lyxose and L-tagatose were ketones with unknown structures, but those of L-ribose and D-lyxose were acids. The oxidation product of L-ribose was purified and revealed to be L-ribonate by HRMS and NMR analysis. Biotransformation of L-ribose with the membranes and also with the whole cells produced L-ribonate in nearly stoichiometric amounts, indicating that the specific oxidation site in L-ribose is recognized by GLDH. Since purified GLDH produced L-ribonate without any intermediate-like compounds, we propose here a reaction model where the first carbon in the pyranose form of L-ribose is oxidized by GLDH to L-ribonolactone, which is further hydrolyzed spontaneously to produce L-ribonate.


Asunto(s)
Gluconobacter/enzimología , Pentosas/metabolismo , Deshidrogenasas del Alcohol de Azúcar/metabolismo , Gluconobacter/metabolismo , Glicerol , Cofactor PQQ/metabolismo
10.
Int J Clin Pharmacol Ther ; 56(4): 184-188, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29350178

RESUMEN

OBJECTIVE: Tuberculosis is an important concern following organ transplantation. Unfortunately, several antituberculosis drugs interact with immunosuppressants. This report describes our experience with rifabutin (RBT) in the treatment of acute tuberculosis in a cardiac transplant recipient. CASE: A 61-year-old cardiac transplant recipient developed tuberculosis meningitis during treatment of miliary tuberculosis. RBT was given for 27 days concomitantly with cyclosporine (CsA). CsA concentrations at 0 hour (C0) decreased within 3 days of starting RBT. The serum concentration-curve from 0 to 12 hours (AUC0-12h)/dose 7 days after starting RBT therapy decreased by 28%, compared to the values before RBT therapy. The apparent clearance at both 7 and 21 days after starting RBT therapy was 1.4 times higher than before RBT therapy. CONCLUSION: RBT has fewer drug-drug interactions than rifampin and should be preferentially used for the treatment of tuberculosis in transplant patients treated with CsA. Close monitoring of CsA blood concentration during RBT therapy minimized the risk of under- or over-immunosuppression in a cardiac transplant patient.
.


Asunto(s)
Antibióticos Antituberculosos/uso terapéutico , Trasplante de Corazón/efectos adversos , Rifabutina/uso terapéutico , Tuberculosis/tratamiento farmacológico , Ciclosporina/sangre , Ciclosporina/uso terapéutico , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad
11.
J Neuroimmune Pharmacol ; 12(4): 693-707, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28755135

RESUMEN

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.


Asunto(s)
Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Dolor/metabolismo , Pancreatitis/complicaciones , Animales , Quimiocina CXCL12/metabolismo , Masculino , Ratones , Dolor/etiología , Pancreatitis/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología
12.
Pharmacology ; 99(5-6): 281-285, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28253495

RESUMEN

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.


Asunto(s)
Hiperalgesia/inducido químicamente , Dolor/fisiopatología , Canales Catiónicos TRPV/fisiología , Tacrolimus/farmacología , Anilidas/farmacología , Animales , Bencimidazoles/farmacología , Ceruletida/efectos adversos , Cinamatos/farmacología , Ciclopropanos/farmacología , Masculino , Ratones , Naftalenos/farmacología , Dolor/complicaciones , Pancreatitis/inducido químicamente , Pancreatitis/complicaciones , Recurrencia , Tacrolimus/antagonistas & inhibidores
13.
Int J Clin Pharmacol Ther ; 55(1): 1-8, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27781420

RESUMEN

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.
.


Asunto(s)
Cronoterapia de Medicamentos , Monitoreo de Drogas/métodos , Everolimus/administración & dosificación , Everolimus/farmacocinética , Trasplante de Corazón , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacocinética , Adulto , Área Bajo la Curva , Interpretación Estadística de Datos , Everolimus/sangre , Everolimus/uso terapéutico , Femenino , Humanos , Inmunosupresores/sangre , Inmunosupresores/uso terapéutico , Masculino , Estudios Retrospectivos
14.
Biol Pharm Bull ; 38(10): 1649-51, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26424025

RESUMEN

Malaria is one of the most prevalent parasitic diseases and is most widespread in tropical regions. The malarial parasite grows and reproduces in erythrocytes during its life cycle, resulting in programmed erythrocyte death, termed eryptosis. Lipid scrambling, which occurs following the exposure of anionic lipids such as phosphatidylserine (PS) on the outer surface of erythrocytes, is a characteristic physical change that occurs early during eryptosis. Here, we prepared "PS specific peptide (PSP)"-conjugated liposomes (PSP-liposomes) and investigated whether PSP-liposomes hold promise as a novel strategy for actively targeting eryptosis. Eryptosis was induced by exposing red blood cells (RBCs) to ionomycin, a known calcium ionophore. When PSP liposomes were mixed with either RBCs or RBCs undergoing eryptosis (E-RBCs), the amount of PSP-liposome bound to E-RBCs was much higher than the amount bound to RBCs. However, the amount of PSP-liposome bound to E-RBCs was significantly inhibited by the presence of annexin V protein, which binds specifically to PS. These results suggest that PSP-liposomes could be an effective drug nanocarrier for treating E-RBCs and malaria-infected erythrocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Lípidos/farmacología , Liposomas/farmacología , Péptidos/farmacología , Animales , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Lípidos/química , Liposomas/química , Malaria , Masculino , Ratones , Péptidos/química , Plasmodium
15.
Biochem Biophys Res Commun ; 464(4): 1034-1039, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26187668

RESUMEN

We examined the intrinsic cell permeability of a GAGA zinc finger obtained from the Drosophila melanogaster transcription factor and analyzed its mechanism of cellular uptake using confocal microscopy and flow cytometry. HeLa cells were treated with the Cy5-labeld GAGA peptides (containing a fluorescent chromophore) to detect fluorescence signals from the fluorescent labeling peptides by confocal microscopy. The results clearly indicated that GAGA peptides possess intrinsic cell permeability for HeLa cells. Based on the results of the flow cytometry analysis and the theoretical net positive charge of the GAGA peptides, the efficiency of cellular uptake of the GAGA peptides was predicted to depend on the net positive charge of the GAGA peptide as well as the cationic component ratio of Arg residues to Lys residues.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Factores de Transcripción/metabolismo , Dedos de Zinc/fisiología , Secuencia de Aminoácidos , Animales , Arginina/química , Permeabilidad de la Membrana Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Citometría de Flujo , Células HeLa , Humanos , Lisina/química , Microscopía Confocal , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Electricidad Estática , Factores de Transcripción/química , Factores de Transcripción/genética , Dedos de Zinc/genética
16.
Handb Exp Pharmacol ; 230: 217-30, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26162837

RESUMEN

Hydrogen sulfide (H2S) formed by multiple enzymes including cystathionine-γ-lyase (CSE) targets Cav3.2 T-type Ca2+ channels (T-channels) and transient receptor potential ankyrin-1 (TRPA1). Intraplantar and intracolonic administration of H2S donors promotes somatic and visceral pain, respectively, via activation of Cav3.2 and TRPA1 in rats and/or mice. Injection of H2S donors into the plantar tissues, pancreatic duct, colonic lumen, or bladder causes T-channel-dependent excitation of nociceptors, determined as phosphorylation of ERK or expression of Fos in the spinal dorsal horn. Electrophysiological studies demonstrate that exogenous and/or endogenous H2S facilitates membrane currents through T-channels in NG108-15 cells and isolated mouse dorsal root ganglion (DRG) neurons that abundantly express Cav3.2 and also in Cav3.2-transfected HEK293 cells. In mice with cerulein-induced pancreatitis and cyclophosphamide-induced cystitis, visceral pain and/or referred hyperalgesia are inhibited by CSE inhibitors and by pharmacological blockade or genetic silencing of Cav3.2, and CSE protein is upregulated in the pancreas and bladder. In rats with neuropathy induced by L5 spinal nerve cutting or by repeated administration of paclitaxel, an anticancer drug, the neuropathic hyperalgesia is reversed by inhibitors of CSE or T-channels and by silencing of Cav3.2. Upregulation of Cav3.2 protein in DRG is detectable in the former, but not in the latter, neuropathic pain models. Thus, H2S appears to function as a nociceptive messenger by facilitating functions of Cav3.2 and TRPA1, and the enhanced function of the CSE/H2S/Cav3.2 pathway is considered to be involved in the pancreatitis- and cystitis-related pain and in neuropathic pain.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Neuralgia/etiología , Dolor/etiología , Animales , Canales de Calcio/fisiología , Canales de Calcio Tipo T/fisiología , Humanos , Ratones , Proteínas del Tejido Nervioso/fisiología , Ratas , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/fisiología
17.
J Neurosci Res ; 93(2): 361-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25267397

RESUMEN

Hydrogen sulfide (H(2)S), formed by multiple enzymes, including cystathionine-γ-lyase (CSE), targets Ca(v)3.2 T-type Ca(2+) channels (T channels) and transient receptor potential ankyrin-1 (TRPA1), facilitating somatic pain. Pancreatitis-related pain also appears to involve activation of T channels by H(2)S formed by the upregulated CSE. Therefore, this study investigates the roles of the Ca(v)3.2 isoform and/or TRPA1 in pancreatic nociception in the absence and presence of pancreatitis. In anesthetized mice, AP18, a TRPA1 inhibitor, abolished the Fos expression in the spinal dorsal horn caused by injection of a TRPA1 agonist into the pancreatic duct. As did mibefradil, a T-channel inhibitor, in our previous report, AP18 prevented the Fos expression following ductal NaHS, an H(2)S donor. In the mice with cerulein-induced acute pancreatitis, the referred hyperalgesia was suppressed by NNC 55-0396 (NNC), a selective T-channel inhibitor; zinc chloride; or ascorbic acid, known to inhibit Ca(v)3.2 selectively among three T-channel isoforms; and knockdown of Ca(v)3.2. In contrast, AP18 and knockdown of TRPA1 had no significant effect on the cerulein-induced referred hyperalgesia, although they significantly potentiated the antihyperalgesic effect of NNC at a subeffective dose. TRPA1 but not Ca(v)3.2 in the dorsal root ganglia was downregulated at a protein level in mice with cerulein-induced pancreatitis. The data indicate that TRPA1 and Ca(v)3.2 mediate the exogenous H(2)S-induced pancreatic nociception in naïve mice and suggest that, in the mice with pancreatitis, Ca(v)3.2 targeted by H(2)S primarily participates in the pancreatic pain, whereas TRPA1 is downregulated and plays a secondary role in pancreatic nociceptive signaling.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Sulfuro de Hidrógeno/uso terapéutico , Pancreatitis/complicaciones , Canales de Potencial de Receptor Transitorio/metabolismo , Dolor Visceral/tratamiento farmacológico , Dolor Visceral/etiología , Análisis de Varianza , Animales , Bencimidazoles/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Ceruletida/toxicidad , Ciclopropanos/uso terapéutico , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Isotiocianatos/farmacología , Masculino , Ratones , Naftalenos/uso terapéutico , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Pancreatitis/inducido químicamente , Pancreatitis/patología , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
18.
J Pharmacol Sci ; 123(3): 284-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24162021

RESUMEN

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.


Asunto(s)
Hiperalgesia/genética , Pancreatitis/complicaciones , Receptor PAR-2/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Canales Catiónicos TRPV/fisiología , Canales de Potencial de Receptor Transitorio/fisiología , Anilidas/administración & dosificación , Anilidas/farmacología , Animales , Cinamatos/administración & dosificación , Cinamatos/farmacología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Nocicepción/fisiología , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
19.
Gan To Kagaku Ryoho ; 33 Suppl 2: 296-8, 2006 Dec.
Artículo en Japonés | MEDLINE | ID: mdl-17469366

RESUMEN

Home parenteral nutrition (HPN) is a useful measure when terminal cancer patients are hoping to have a transition from the hospital to a home medical care arrangement. Our department has been supporting their family by mixing injections at our clean room. We created a HPN report form to check for an incompatibility of injections, and to prepare an infusion set by our department. By using this report form, the introduction of HPN to the patient's family became smooth. This report form is useful for contacting the accounting section and the visiting nurse station. We report a contribution of the hospital pharmacy for HPN.


Asunto(s)
Servicios de Atención a Domicilio Provisto por Hospital , Nutrición Parenteral en el Domicilio/estadística & datos numéricos , Servicio de Farmacia en Hospital , Registros , Enfermería en Salud Comunitaria , Humanos , Farmacéuticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...