Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.068
Filtrar
1.
PLoS One ; 17(1): e0262233, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34986201

RESUMEN

The micro- and macro-complications in diabetes mellitus (DM) mainly arise from the damage induced by Amadori and advanced glycation end products, as well as the released free radicals. The primary goal of DM treatment is to reduce the risk of micro- and macro-complications. In this study, we looked at the efficacy of aminoguanidine (AG) to prevent the production of early glycation products in alloxan-diabetic rabbits. Type1 DM was induced in rabbits by a single intravenous injection of alloxan (90 mg/kg body weight). Another group of rabbits was pre-treated with AG (100 mg/kg body weight) prior to alloxan injection; this was followed by weekly treatment with 100 mg/kg of AG for eight weeks. Glucose, insulin, and early glycation products (HbA1C and fructosamine) were measured in control, diabetic and AG treated diabetic rabbits. The effects of hyperglycemia on superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (Gpx), reduced glutathione (rGSH), nitric oxide, lipid peroxides, and protein carbonyl were investigated. Alloxan-diabetic rabbits had lower levels of SOD, CAT, Gpx, and rGSH than control rabbits. Nitric oxide levels were considerably greater. AG administration restored the activities of SOD, CAT, Gpx enzymes up to 70-80% and ameliorated the nitric oxide production. HbA1c and fructosamine levels were considerably lower in AG-treated diabetic rabbits. The observed control of hyperglycemia and amadori adducts in alloxan-diabetic rabbits by AG may be attributed to decrease of stress and restoration of antioxidant defenses.


Asunto(s)
Antioxidantes/administración & dosificación , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Guanidinas/administración & dosificación , Hiperglucemia/tratamiento farmacológico , Aloxano , Animales , Antioxidantes/farmacología , Estudios de Casos y Controles , Catalasa/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Esquema de Medicación , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Guanidinas/farmacología , Hiperglucemia/inducido químicamente , Hiperglucemia/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Conejos , Superóxido Dismutasa/metabolismo
2.
J Pharmacol Sci ; 148(1): 65-72, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34924132

RESUMEN

This study aimed to evaluate the effects of nafamostat, a serin protease inhibitor, in the management of subarachnoid hemorrhage (SAH). SAH was induced by endovascular perforation in male mice. Nafamostat was administered intraperitoneally four times immediately after SAH induction. Cerebral blood flow, neurological behavior tests, SAH grade and protein expression were evaluated at 24 h after SAH induction. In the in vitro model, human brain microvascular endothelial cells (HBMVECs), HBVECs were exposed to thrombin and hypoxia for 24 h; nafamostat was administered and the protein expression was evaluated. Eighty-eight mice were included in the in vivo study. Fifteen mice (17%) were excluded because of death or procedure failure. Nafamostat exerted no significant effect on the SAH grade or cerebral blood flow; however, it improved the neurological behavior and suppressed the thrombin and MMP-9 expression. In addition, nafamostat suppressed the ICAM-1 expression and p38 phosphorylation in the in vitro study. Nafamostat has a protective effect against HBMVEC after exposure to thrombin and hypoxia, suggesting its role in improving the neurological outcomes after SAH. These findings indicate that nafamostat has the potential to be a novel therapeutic drug in the management of SAH.


Asunto(s)
Benzamidinas/administración & dosificación , Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Guanidinas/administración & dosificación , Inhibidores de Serina Proteinasa/administración & dosificación , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico , Animales , Benzamidinas/farmacología , Encéfalo/citología , Lesiones Encefálicas/genética , Células Cultivadas , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Guanidinas/farmacología , Humanos , Infusiones Parenterales , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos , Inhibidores de Serina Proteinasa/farmacología , Hemorragia Subaracnoidea/genética , Trombina/genética , Trombina/metabolismo
3.
PLoS One ; 16(9): e0257019, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34473771

RESUMEN

Nuclear factor kappa B (NF-κB) is a transcriptional factor that can be activated by radiotherapy and chemotherapy. The synthetic protease inhibitor nafamostat mesilate (NM) inhibits NF-κB activity and exerts antitumor actions in various types of cancer. In the present study, we hypothesized that NM might enhance the antitumor action of radiotherapy on gallbladder cancer (GBC) cells by inhibiting radiation-induced NF-κB activity. Thus, we investigated the correlation between radiotherapy and NF-κB activity in GBC cells. We assessed the in vitro effects of radiotherapy with or without NM on NF-κB activity, apoptosis of GBC cells (NOZ and OCUG-1), induction of apoptotic cascade, cell cycle progression, and viability of GBC cells using four treatment groups: 1) radiation (5 Gy) alone; 2) NM (80 µg/mL and 40 µg/mL, respectively) alone; 3) combination (radiation and NM); and 4) vehicle (control). The same experiments were performed in vivo using a xenograft GBC mouse model. In vitro, NM inhibited radiation-induced NF-κB activity. Combination treatment significantly attenuated cell viability and increased cell apoptosis and G2/M phase cell cycle arrest compared with those in the other groups for NOZ and OCUG-1 cells. Moreover, combination treatment upregulated the expression of apoptotic proteins compared with that after the other treatments. In vivo, NM improved the antitumor action of radiation and increased the population of Ki-67-positive cells. Overall, NM enhanced the antitumor action of radiotherapy on GBC cells by suppressing radiation-induced NF-κB activity. Thus, the combination of radiotherapy and NM may be useful for the treatment of locally advanced unresectable GBC.


Asunto(s)
Benzamidinas/administración & dosificación , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/radioterapia , Guanidinas/administración & dosificación , FN-kappa B/antagonistas & inhibidores , Inhibidores de Proteasas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Terapia Combinada/métodos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
PLoS One ; 16(8): e0248604, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34351936

RESUMEN

Indoor residual spraying (IRS) is one of the main malaria vector control strategies in Mozambique alongside the distribution of insecticide treated nets. As part of the national insecticide resistance management strategy, Mozambique introduced SumiShield™ 50WG, a third generation IRS product, in 2018. Its residual efficacy was assessed in southern Mozambique during the 2018-2019 malaria season. Using a susceptible Anopheles arabiensis strain, residual efficacy was assessed on two different wall surfaces, cement and mud-plastered walls, using standard WHO (World Health Organization) cone bioassay tests at three different heights. Female mosquitoes of two age groups (2-5 and 13-26 day old) were exposed for 30 minutes, after which mortality was observed 24h, 48h, 72h, and 96h and 120h post-exposure to assess (delayed) mortality. Lethal times (LT) 90, LT50 and LT10 were estimated using Bayesian models. Mortality 24h post exposure was consistently below 80%, the current WHO threshold value for effective IRS, in both young and old mosquitoes, regardless of wall surface type. Considering delayed mortality, residual efficacies (mosquito mortality equal or greater than 80%) ranged from 1.5 to ≥12.5 months, with the duration depending on mortality time post exposure, wall type and mosquito age. Looking at mortality 72h after exposure, residual efficacy was between 6.5 and 9.5 months, depending on wall type and mosquito age. The LT50 and LT10 (i.e. 90% of the mosquitoes survive exposure to the insecticides) values were consistently higher for older mosquitoes (except for LT10 values for 48h and 72h post-exposure mortality) and ranged from 0.9 to 5.8 months and 0.2 to 7.8 months for LT50 and LT10, respectively. The present study highlights the need for assessing mosquito mortality beyond the currently recommended 24h post exposure. Failure to do so may lead to underestimation of the residual efficacy of IRS products, as delayed mortality will lead to a further reduction in mosquito vector populations and potentially negatively impact disease transmission. Monitoring residual efficacy on relevant wall surfaces, including old mosquitoes that are ultimately responsible for malaria transmission, and assessing delayed mortalities are critical to provide accurate and actionable data to guide vector control programmes.


Asunto(s)
Anopheles , Guanidinas/administración & dosificación , Insecticidas/administración & dosificación , Control de Mosquitos/métodos , Neonicotinoides/administración & dosificación , Tiazoles/administración & dosificación , Aerosoles , Factores de Edad , Animales , Femenino , Vivienda , Mozambique
5.
Parasit Vectors ; 14(1): 416, 2021 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-34419129

RESUMEN

BACKGROUND: We evaluated the efficiency of an ex vivo feeding technique using a silicone membrane-based feeding chamber to (i) assess the anti-feeding and acaricidal efficacy of a spot-on combination of dinotefuran, pyriproxyfen and permethrin (DPP, Vectra® 3D) against adult Ixodes scapularis and Ixodes ricinus ticks, and to (ii) explore its effect on blocking the acquisition of Borrelia burgdorferi sensu stricto. METHODS: Eight purpose-bred dogs were randomly allocated to two equal-size groups based on body weight assessed on day 2. DPP was administered topically, as spot-on, to four dogs on day 0. Hair from the eight dogs was collected individually by brushing the whole body on days 2, 7, 14, 21, 28 and 35. On each day of hair collection, 0.05 g of sampled hair was applied on the membrane corresponding to each feeding unit (FU). Seventy-two FU were each seeded with 30 adults of I. scapularis (n = 24 FU) or I. ricinus ticks (n = 48 FU). Bovine blood spiked with B. burgdorferi sensu stricto (strain B31) was added into each unit and changed every 12 h for 4 days. Tick mortality was assessed 1 h after seeding. One additional hour of incubation was added for live/moribund specimens and reassessed for viability. All remaining live/moribund ticks were left in the feeders and tick engorgement status was recorded at 96 h after seeding, and the uptake of B. burgdorferi s.s. was examined in the collected ticks by applying quantitative real-time PCR. RESULTS: Exposure to DPP-treated hair was 100% effective in blocking B. burgdorferi s.s. acquisition. The anti-feeding efficacy remained stable (100%) against both Ixodes species throughout the study. The acaricidal efficacy of DPP evaluated at 1 and 2 h after exposure was 100% throughout the study for I. ricinus, except the 1-h assessment on day 28 (95.9%) and day 35 (95.3%). The 1-h assessment of acaricidal efficacy was 100% at all time points for I. scapularis. CONCLUSIONS: The ex vivo feeding system developed here demonstrated a protective effect of DPP against the acquisition of B. burgdorferi without exposing the animals to the vectors or to the pathogen.


Asunto(s)
Enfermedades de los Perros/prevención & control , Guanidinas/administración & dosificación , Insecticidas/administración & dosificación , Ixodes/efectos de los fármacos , Enfermedad de Lyme/prevención & control , Neonicotinoides/administración & dosificación , Nitrocompuestos/administración & dosificación , Permetrina/administración & dosificación , Piridinas/administración & dosificación , Administración Tópica , Animales , Borrelia burgdorferi/fisiología , Enfermedades de los Perros/microbiología , Perros , Combinación de Medicamentos , Conducta Alimentaria , Femenino , Ixodes/clasificación , Ixodes/microbiología , Enfermedad de Lyme/transmisión , Masculino
6.
Clin Transl Sci ; 14(5): 1967-1976, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33982445

RESUMEN

Camostat mesylate, an oral serine protease inhibitor, is used to treat chronic pancreatitis and reflux esophagitis. Recently, camostat mesylate and its active metabolite 4-(4-guanidinobenzoyloxy)phenylacetic acid (GBPA) were reported to inhibit the infection of cells by severe acute respiratory syndrome coronavirus 2 by inhibiting type II transmembrane serine protease. We conducted a phase I study to investigate high-dose camostat mesylate as a treatment for coronavirus disease 2019. Camostat mesylate was orally administered to healthy adults at 600 mg 4 times daily under either of the following conditions: fasted state, after a meal, 30 min before a meal, or 1 h before a meal, and the pharmacokinetics and safety profiles were evaluated. In addition, the time of plasma GBPA concentration exceeding the effective concentration was estimated as the time above half-maximal effective concentration (EC50 ) by using pharmacokinetic/pharmacodynamic modeling and simulation. Camostat mesylate was safe and tolerated at all dosages. Compared with the fasted state, the exposure of GBPA after a meal and 30 min before a meal was significantly lower; however, no significant difference was observed at 1 h before a meal. The time above EC50 was 11.5 h when camostat mesylate 600 mg was administered 4 times daily in the fasted state or 1 h before a meal. Based on the results of this phase I study, we are currently conducting a phase III study.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Ésteres/efectos adversos , Guanidinas/efectos adversos , Inhibidores de Serina Proteinasa/administración & dosificación , Administración Oral , Adolescente , Adulto , Simulación por Computador , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Ésteres/administración & dosificación , Ésteres/farmacocinética , Interacciones Alimento-Droga , Guanidinas/administración & dosificación , Guanidinas/farmacocinética , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/efectos adversos , Adulto Joven
7.
Oncogene ; 40(22): 3799-3814, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33958721

RESUMEN

Despite the development of new targeted and immune therapies, the prognosis of metastatic melanoma remains bleak. Therefore, it is critical to better understand the mechanisms controlling advanced melanoma to develop more effective treatment regimens. Hedgehog/GLI (HH/GLI) signaling inhibitors targeting the central pathway transducer Smoothened (SMO) have shown to be clinical efficacious in skin cancer; however, several mechanisms of non-canonical HH/GLI pathway activation limit their efficacy. Here, we identify a novel SOX2-BRD4 transcriptional complex driving the expression of GLI1, the final effector of the HH/GLI pathway, providing a novel mechanism of non-canonical SMO-independent activation of HH/GLI signaling in melanoma. Consistently, we find a positive correlation between the expression of GLI1 and SOX2 in human melanoma samples and cell lines. Further, we show that combined targeting of canonical HH/GLI pathway with the SMO inhibitor MRT-92 and of the SOX2-BRD4 complex using a potent Proteolysis Targeted Chimeras (PROTACs)-derived BRD4 degrader (MZ1), yields a synergistic anti-proliferative effect in melanoma cells independently of their BRAF, NRAS, and NF1 mutational status, with complete abrogation of GLI1 expression. Combination of MRT-92 and MZ1 strongly potentiates the antitumor effect of either drug as single agents in an orthotopic melanoma model. Together, our data provide evidence of a novel mechanism of non-canonical activation of GLI1 by the SOX2-BRD4 transcriptional complex, and describe the efficacy of a new combinatorial treatment for a subset of melanomas with an active SOX2-BRD4-GLI1 axis.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Dipéptidos/farmacología , Guanidinas/farmacología , Proteínas Hedgehog/antagonistas & inhibidores , Compuestos Heterocíclicos con 3 Anillos/farmacología , Melanoma/tratamiento farmacológico , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dipéptidos/administración & dosificación , Sinergismo Farmacológico , Femenino , Guanidinas/administración & dosificación , Proteínas Hedgehog/metabolismo , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/antagonistas & inhibidores , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Pancreas ; 50(3): 313-316, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33835961

RESUMEN

OBJECTIVES: The aim of this study was to clarify the effectiveness of combination chemotherapy targeting gemcitabine (GEM)-induced nuclear factor kappa B as adjuvant therapy for pancreatic cancer. METHODS: Patients who were planned after curative surgery (residual tumor classification R0 or R1) for pancreatic cancer to receive six cycles of adjuvant chemotherapy of regional arterial infusion of nafamostat mesilate with GEM between June 2011 and April 2017 were enrolled in this single-center, institutional review board-approved phase II trial (UMIN000006163). The Kaplan-Meier method was used to estimate disease-free survival and overall survival. RESULTS: In 32 patients [male/female: 18/14; age: median, 65.5 years (range, 48-77 years); pathological stage (Union for International Cancer Control 8th): IA/IB/IIA/IIB/III, 2/2/9/18/1, respectively] who met the eligibility criteria, the median overall survival and disease-free survival were 36.4 months (95% confidence interval, 31.7-48.3) and 16.4 months (95% confidence interval, 14.3-22.0), respectively. Grade 4 treatment-related hematological toxicities were seen in 5 patients (15.6%) (all neutropenia). One patient developed grade 3 nonhematological toxicities (rash). CONCLUSIONS: Adjuvant chemotherapy with regional arterial infusion of nafamostat mesilate and GEM is safe and has potential as an option in adjuvant setting after curative surgery for pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia Adyuvante/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/cirugía , Anciano , Benzamidinas/administración & dosificación , Terapia Combinada/efectos adversos , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Exantema/etiología , Femenino , Guanidinas/administración & dosificación , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neutropenia/etiología , Periodo Posoperatorio , Resultado del Tratamiento , Gemcitabina
9.
Expert Rev Anti Infect Ther ; 19(8): 1039-1046, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33641583

RESUMEN

BACKGROUND: The study was to compare the efficacy between IV peramivir and oral oseltamivir treatments in patients with influenza. METHODS: The PubMed, EMBASE, Scopus, ClinicalTrials.gov, and Cochrane Library databases were searched for studies published before January 2020. RESULTS: The meta-analysis was conducted to calculate the pooled effect size by using a random-effects model. Seven randomized controlled trials (RCTs) including 1,138 patients were reviewed. The incidence of total complications revealed no significant difference between 600 mg IV peramivir (P600) and 75 mg oral oseltamivir (O75) treatments (2.8% vs. 4.1%; risk ratio [RR] = 0.70; 95% confidence interval [CI]: 0.36-1.38). The incidence of pneumonia was not significantly different between the P600 and O75 treatment groups (2.2% vs. 2.7%; RR = 0.74; 95% CI: 0.37-1.51). Regarding the time to the alleviation of symptoms, no difference was found in P600 and O75 treatment (MD = -3.00; 95% CI: -11.07 to 5.06). The rate of fever clearance in 24 h and the time to fever resolution were not statistically different between the IV peramivir and oral oseltamivir treatments (at different dosages) groups. CONCLUSIONS: The treatment of influenza with IV peramivir or oral oseltamivir had similar clinical efficacy.


Asunto(s)
Ácidos Carbocíclicos/administración & dosificación , Guanidinas/administración & dosificación , Gripe Humana/tratamiento farmacológico , Oseltamivir/administración & dosificación , Administración Intravenosa , Administración Oral , Antivirales/administración & dosificación , Humanos , Gripe Humana/virología , Neumonía/epidemiología , Neumonía/virología , Ensayos Clínicos Controlados Aleatorios como Asunto
10.
Toxicol Lett ; 342: 95-103, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33609686

RESUMEN

Neonicotinoids, which act as agonists of the nicotinic acetylcholine receptors of insects, are widely used pesticides worldwide. Although epidemiological studies revealed that the detection amounts of neonicotinoids in urine are higher in the elderly population than other age-groups, there is no available information regarding the risks of neonicotinoids to older mammals. This study was aimed to investigate aging-related differences in the behavioral effects of the neonicotinoid pesticide clothianidin (CLO). We acutely administered a sub-NOAEL level (5 mg/kg) of CLO to adult (12-week-old) and aging (90-week-old) mice and conducted four behavioral tests focusing on the emotional behavior. In addition, we measured the concentrations of CLO and its metabolites in blood, brain and urine. There were age-related changes in most parameters in all behavioral tests, and CLO significantly decreased the locomotor activity in the open field test and elevated plus-maze test in the aging group, but not in the adult group. The concentrations of most CLO and its metabolites were significantly higher in the blood and brain and were slightly lower in the urine in the aging group compared to the adult group. These findings should contribute to our understanding of age-related differences in the adverse effects of neonicotinoids in mammals.


Asunto(s)
Conducta Animal/efectos de los fármacos , Guanidinas/toxicidad , Insecticidas/toxicidad , Neonicotinoides/toxicidad , Tiazoles/toxicidad , Envejecimiento , Animales , Relación Dosis-Respuesta a Droga , Guanidinas/administración & dosificación , Insecticidas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Neonicotinoides/administración & dosificación , Tiazoles/administración & dosificación
11.
Biol Pharm Bull ; 44(2): 259-265, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33518678

RESUMEN

Nafamostat mesilate (NFM) is used as an anticoagulant during hemodialysis in patients who have had complications due to hemorrhages. The formation of precipitates, which could lead to the interruption of hemodialysis has been reported when NFM is infused into blood during hemodialysis. We report herein on an examination of possible factors that could cause this. The effects of electrolytes such as phosphates, citrates or succinates on the formation of precipitates were examined by mixing NFM with aqueous solutions or plasma that contained these electrolytes. The formation of precipitates was observed in all electrolyte solutions when higher concentrations of NFM were mixed at around physiological pH. In the case of plasma, precipitates were observed when solutions containing higher concentrations of NFM were mixed with plasma that contained phosphate and citrate. In addition, the formation of precipitates under dynamic conditions where NFM was infused into flowing electrolyte solutions was also evaluated. The data suggested that such precipitates might be formed and disrupt the blood flow and/or an NFM infusion when NFM is infused into blood flowing in the hemodialysis circuit. The findings presented herein suggest the serum levels of anionic electrolytes (e.g., phosphate), the type of excipients present in pharmaceutical products (e.g., succinic acid or citric acid), the concentration of NFM used for the infusion or the rates of NFM infusion and blood flow are all factors that could affect precipitate formation during NFM infusions for hemodialysis.


Asunto(s)
Anticoagulantes/administración & dosificación , Benzamidinas/administración & dosificación , Soluciones para Diálisis/química , Guanidinas/administración & dosificación , Diálisis Renal/efectos adversos , Aniones/sangre , Aniones/química , Anticoagulantes/química , Benzamidinas/química , Electrólitos/sangre , Electrólitos/química , Guanidinas/química , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Humanos , Plasma/química , Solubilidad
12.
Arch Toxicol ; 95(3): 1039-1053, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33426623

RESUMEN

Co-occurrence of pesticide residues in food commodities raises a potential safety issue as their mixture effects on human health are largely unknown. In a previous study, we reported the toxicological effects (pathology and histopathology) of imazalil (IMZ), thiacloprid (THI), and clothianidin (CTD) alone and in binary mixtures in a 28-day oral gavage study in female Wistar rats. Five dose levels (up to 350 mg/kg body weight/day) ranging from a typical toxicological reference value to a clear effect dose were applied. In the present study, we undertook a transcriptomics analysis of rat livers by means of total RNA sequencing (RNA-Seq). Bioinformatic data analysis involving Ingenuity Pathway Analysis (IPA) was used to gain mechanistic information on hepatotoxicity-related pathways affected after treatment with the pesticides, alone and in mixtures. Our data show that 2986 genes were differentially regulated by CTD while IMZ and THI had effects on 194 and 225 genes, respectively. All three individual compounds shared a common subset of genes whose network is associated with xenobiotic metabolism and nuclear receptor activation. Similar networks were retrieved for the mixtures. Alterations in the expression of individual genes were in line with the assumption of dose addition. Our results bring new insight into the hepatotoxicity mechanisms of IMZ, THI, and CTD and their mixtures.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Guanidinas/toxicidad , Imidazoles/toxicidad , Neonicotinoides/toxicidad , Tiazinas/toxicidad , Tiazoles/toxicidad , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Guanidinas/administración & dosificación , Imidazoles/administración & dosificación , Neonicotinoides/administración & dosificación , Plaguicidas/toxicidad , Ratas , Ratas Wistar , Análisis de Secuencia de ARN , Tiazinas/administración & dosificación , Tiazoles/administración & dosificación
13.
Endocr J ; 68(4): 477-484, 2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33361650

RESUMEN

We provide the details of the successful management of a patient with active Cushing's disease complicated with coronavirus disease 2019 (COVID-19) pneumonia. The patient was a 27-year-old Japanese female healthcare worker who was scheduled to undergo pituitary surgery for Cushing's disease. She had been in close contact with an undiagnosed patient infected with COVID-19 and then developed COVID-19 pneumonia. Despite a lack of known risk factors associated with severe COVID-19 infection, the patient's dyspnea worsened and her respiratory condition deteriorated, as indicated by the need for 7 L/min oxygen supply by mask to maintain her oxygen saturation at >90%. Medical treatment was initiated to control hypercortisolism by the 'block and replace' regimen using steroidogenesis inhibitors and hydrocortisone. The COVID-19 pneumonia improved with multi-modal treatment including antiviral therapy. One month later, after a negative severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) test result and with appropriate protection against virus transmission to medical staff in the operating room and daily medical care nurses, trans-sphenoidal surgery was performed by our highly experienced pituitary surgeon. One month after the surgery, the patient's basal ACTH and cortisol levels and urinary free cortisol were all under the detection limit. Surgical remission was expected. Since hypercortisolism due to active Cushing's disease may worsen a COVID-19 infection, multi-disciplinary management that includes appropriate and prompt treatment strategies is mandatory in such cases.


Asunto(s)
Amidas/administración & dosificación , Benzamidinas/administración & dosificación , COVID-19/terapia , Guanidinas/administración & dosificación , Metirapona/administración & dosificación , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/terapia , Pregnenodionas/administración & dosificación , Pirazinas/administración & dosificación , Adenoma Hipofisario Secretor de ACTH/complicaciones , Adenoma Hipofisario Secretor de ACTH/tratamiento farmacológico , Adenoma/complicaciones , Adenoma/tratamiento farmacológico , Adulto , COVID-19/complicaciones , COVID-19/patología , Terapia Combinada , Dihidrotestosterona/administración & dosificación , Dihidrotestosterona/análogos & derivados , Progresión de la Enfermedad , Femenino , Personal de Salud , Heparina/administración & dosificación , Humanos , Japón , Procedimientos Neuroquirúrgicos , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/sangre , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/complicaciones , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/patología , SARS-CoV-2/fisiología , Resultado del Tratamiento , Combinación Trimetoprim y Sulfametoxazol/administración & dosificación
14.
Drug Chem Toxicol ; 44(2): 215-221, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30691306

RESUMEN

Lipopolysaccharide (LPS) as the major component of the outer membrane of Gram-negative bacteria activates macrophages to produce a high level of pro-inflammatory cytokines which is considered as a cause of liver dysfunction. Overproduction of nitric oxide (NO) has been suggested to have a role in hepatic injury. The aim of the present study was to explore the protective effects of aminoguanidine (AG) as inducible nitric oxide synthase (iNOS) inhibitor against LPS -induced liver dysfunction in rat. The animals were divided into five groups: (1) control (2) LPS (3) LPS-AG50, (4) LPS-AG100 and (5) LPS-AG150. LPS (1 mg/kg) was injected for 5 weeks and AG (50, 100 and 150 mg/kg) was administered 30 min before LPS. Drugs were injected intraperitoneally. LPS induced liver dysfunction presented by increasing the serum level of alkaline phosphatase (ALK-P), alanine aminotransferase (ALT), aspartate aminotransferase (AST). Pretreatment with AG restored harmful effects of LPS on liver function. In addition, LPS resulted in hepatotoxicity, accompanied by enhancing the level of interleukin (IL)-6, malondialdehyde (MDA) and nitric oxide (NO) metabolites and decreasing the content of total thiol groups and superoxide dismutase (SOD) and catalase (CAT) activity. Injection of AG before LPS attenuated LPS-induced hepatotoxicity through decreasing the level of IL-6, MDA and NO metabolites and increasing total thiols and SOD and CAT activity. Considering the protective effect of AG which was seen in the present study, it seems that increased levels of NO due to activation of iNOS has a role in LPS-induced hepatic injury.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Hepatopatías/prevención & control , Óxido Nítrico/metabolismo , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Guanidinas/administración & dosificación , Lipopolisacáridos/toxicidad , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Ratas , Ratas Wistar
15.
Int J Infect Dis ; 102: 529-531, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33157292

RESUMEN

Nafamostat, a serine proteinase inhibitor with various actions including antithrombin, antiplasmin, and antitrypsin effects, has been used in clinical practice to treat disseminated intravascular coagulation (DIC) and pancreatitis. This case report describes the clinical course of a patient with COVID-19 pneumonia whose severe hypoxemia, probably caused by DIC and pulmonary embolism, showed remarkable improvement with combination heparin and nafamostat therapy. In addition, beneficial mechanisms of nafamostat against COVID-19 and the necessity of attention to hyperkalemia as an adverse effect are discussed.


Asunto(s)
Anticoagulantes/administración & dosificación , COVID-19/complicaciones , Coagulación Intravascular Diseminada/tratamiento farmacológico , Guanidinas/administración & dosificación , Anciano , Benzamidinas , Coagulación Sanguínea/efectos de los fármacos , COVID-19/sangre , COVID-19/virología , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/etiología , Coagulación Intravascular Diseminada/virología , Humanos , Masculino , SARS-CoV-2/fisiología
16.
Basic Clin Pharmacol Toxicol ; 128(2): 204-212, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33176395

RESUMEN

The coronavirus responsible for COVID-19, SARS-CoV-2, utilizes a viral membrane spike protein for host cell entry. For the virus to engage in host membrane fusion, SARS-CoV-2 utilizes the human transmembrane surface protease, TMPRSS2, to cleave and activate the spike protein. Camostat mesylate, an orally available well-known serine protease inhibitor, is a potent inhibitor of TMPRSS2 and has been hypothesized as a potential antiviral drug against COVID-19. In vitro human cell and animal studies have shown that camostat mesylate inhibits virus-cell membrane fusion and hence viral replication. In mice, camostat mesylate treatment during acute infection with influenza, also dependent on TMPRSS2, leads to a reduced viral load. The decreased viral load may be associated with an improved patient outcome. Because camostat mesylate is administered as an oral drug, it may be used in outpatients as well as inpatients at all disease stages of SARS-CoV-2 infection if it is shown to be an effective antiviral agent. Clinical trials are currently ongoing to test whether this well-known drug could be repurposed and utilized to combat the current pandemic. In the following, we will review current knowledge on camostat mesylate mode of action, potential benefits as an antiviral agent and ongoing clinical trials.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ésteres/uso terapéutico , Guanidinas/uso terapéutico , Inhibidores de Serina Proteinasa/uso terapéutico , Animales , Antivirales/administración & dosificación , Antivirales/efectos adversos , Reposicionamiento de Medicamentos , Ésteres/administración & dosificación , Ésteres/efectos adversos , Guanidinas/administración & dosificación , Guanidinas/efectos adversos , Humanos , Ratones , Seguridad del Paciente , Serina Endopeptidasas/efectos de los fármacos , Inhibidores de Serina Proteinasa/administración & dosificación , Inhibidores de Serina Proteinasa/efectos adversos
17.
Cancer Res ; 80(22): 5024-5034, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32998997

RESUMEN

The aggressive primary brain tumor glioblastoma (GBM) is characterized by aberrant metabolism that fuels its malignant phenotype. Diverse genetic subtypes of malignant glioma are sensitive to selective inhibition of the NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT). However, the potential impact of NAD+ depletion on the brain tumor microenvironment has not been elaborated. In addition, systemic toxicity of NAMPT inhibition remains a significant concern. Here we show that microparticle-mediated intratumoral delivery of NAMPT inhibitor GMX1778 induces specific immunologic changes in the tumor microenvironment of murine GBM, characterized by upregulation of immune checkpoint PD-L1, recruitment of CD3+, CD4+, and CD8+ T cells, and reduction of M2-polarized immunosuppressive macrophages. NAD+ depletion and autophagy induced by NAMPT inhibitors mediated the upregulation of PD-L1 transcripts and cell surface protein levels in GBM cells. NAMPT inhibitor modulation of the tumor immune microenvironment was therefore combined with PD-1 checkpoint blockade in vivo, significantly increasing the survival of GBM-bearing animals. Thus, the therapeutic impacts of NAMPT inhibition extended beyond neoplastic cells, shaping surrounding immune effectors. Microparticle delivery and release of NAMPT inhibitor at the tumor site offers a safe and robust means to alter an immune tumor microenvironment that could potentiate checkpoint immunotherapy for glioblastoma. SIGNIFICANCE: Microparticle-mediated local inhibition of NAMPT modulates the tumor immune microenvironment and acts cooperatively with anti-PD-1 checkpoint blockade, offering a combination immunotherapy strategy for the treatment of GBM.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias Encefálicas/terapia , Cianuros/administración & dosificación , Citocinas/antagonistas & inhibidores , Glioblastoma/terapia , Guanidinas/administración & dosificación , NAD/efectos de los fármacos , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Acrilamidas/administración & dosificación , Animales , Autofagia , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Movimiento Celular , Cianuros/efectos adversos , Preparaciones de Acción Retardada , Portadores de Fármacos/síntesis química , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Guanidinas/efectos adversos , Humanos , Inyecciones Intralesiones , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , NAD/análisis , NAD/deficiencia , Piperidinas/administración & dosificación , Polímeros/síntesis química , ARN Mensajero/metabolismo , Transducción de Señal , Microambiente Tumoral/inmunología , Regulación hacia Arriba/efectos de los fármacos
18.
PLoS Negl Trop Dis ; 14(10): e0008775, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33091049

RESUMEN

Mycetoma is a chronic infectious disease that can be caused by fungi or bacteria, Madurella mycetomatis and Nocardia brasiliensis are frequent etiologic agents of this disease. Mycetoma produced by bacteria is known as actinomycetoma. In mycetoma produced by fungi (eumycetoma) and actinomycetoma, diagnosis of the disease is based on clinical findings: severe inflammation, with deformities of affected tissues, abscesses, fistulae, sinuses and discharge of purulent material that contains micro colonies of the etiologic agent. Microscopic examination of infected tissue is similar regardless of the offending microbe; hallmark of infected tissue is severe inflammation with abundant neutrophils around micro colonies and granuloma formation with macrophages, lymphocytes, dendritic and foamy cells. Even though medical treatment is available for mycetoma patients, amputation, or surgical intervention is frequently needed. The pathogenesis of actinomycetoma is little known, most information was obtained from experimental animal models infected with bacteria. In other experimental mice infections with different microbes, it was demonstrated that nitric oxide is responsible for the intracellular killing of Mycobacterium tuberculosis by activated macrophages. Nitric oxide is a free radical with potent stimulatory and suppressive effects in innate and adaptive immunity. The unstable nitric oxide molecule is produced by action of nitric oxide synthases on L-arginine. There are three nitric oxide synthases expressed in different cells and tissues, two are constitutively expressed one in neurons, and the other in endothelial cells and one that is inducible in macrophages. Aminoguanidine is a competitive inhibitor of inducible nitric oxide synthase. Its administration in experimental animals may favor or harm them. We used aminoguanidine in mice infected with Nocardia brasiliensis, and demonstrated that all treated animals were protected from actinomycetoma development. Anti N. brasiliensis antibodies and T cell proliferation were not affected, but inflammation was reduced.


Asunto(s)
Guanidinas/administración & dosificación , Micetoma/tratamiento farmacológico , Micetoma/enzimología , Óxido Nítrico Sintasa de Tipo II/inmunología , Animales , Progresión de la Enfermedad , Femenino , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Micetoma/inmunología , Micetoma/microbiología , Neutrófilos/inmunología , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Nocardia/efectos de los fármacos , Nocardia/fisiología
19.
J Infect Chemother ; 26(12): 1319-1323, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32893123

RESUMEN

The number of people infected with severe acute respiratory syndrome coronavirus 2 is increasing globally, and some patients have a fatal clinical course. In light of this situation, the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) a pandemic on March 11, 2020. While clinical studies and basic research on a treatment for COVID-19 are ongoing around the world, no treatment has yet been proven to be effective. Several clinical studies have demonstrated the efficacy of chloroquine phosphate and nafamostat mesylate with COVID-19. Here, we report the case of a Japanese patient with COVID-19 with severe respiratory failure who improved following the administration of hydroxychloroquine and continuous hemodiafiltlation with nafamostat mesylate. Hence, hydroxychloroquine with nafamostat mesylate might be a treatment option for severe COVID-19.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Guanidinas/administración & dosificación , Hemodiafiltración/métodos , Hidroxicloroquina/administración & dosificación , Neumonía Viral/tratamiento farmacológico , Anciano , Antiinflamatorios no Esteroideos/administración & dosificación , Antivirales/administración & dosificación , Benzamidinas , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/terapia , Combinación de Medicamentos , Humanos , Japón , Lopinavir/administración & dosificación , Masculino , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/terapia , Insuficiencia Respiratoria/complicaciones , Ritonavir/administración & dosificación , SARS-CoV-2 , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19
20.
Biomed Pharmacother ; 129: 110464, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32768954

RESUMEN

Peramivir, a neuraminidase inhibitor, was approved globally and is indicated for the treatment of uncomplicated influenza in adults and children. However, the only approved intravenous formulation of peramivir limits its clinical application due to the need for the specialized dosing techniques and increases the risk of contracting influenza virus infection among healthcare professionals when dosing within a short distance to the patient. The purpose of this study was to investigate the pharmacokinetic profile of peramivir in plasma and the lung of rats and to compare the profiles following administration through trans-nasal aerosol inhalation (0.0888, 0.1776, and 0.3552 mg/kg) and intravenous injection (30 mg/kg). The plasma concentration reached the Cmax within 1.0 h (upon inhalation) and decreased at a t1/2 of 6.71 and 10.9 h after inhalation and injection, respectively. The absolute bioavailability of peramivir after inhalation was 78.2 %. Overall, the pharmacokinetic exposure of peramivir in the lungs was higher than that in the plasma after aerosol inhalation. After inhalation, the Cmax of peramivir in the lung was achieved within 1.0 h, and the elimination of the drug was slower than in the case of intravenous injection with t1/2 values 1.81 h for injection and 5.72, 53.5, and 32.1 h for low, middle, and high doses administered through inhalation. The Cmax and AUC0-t values for peramivir in the lungs increased linearly with the increased inhalation dose. The results elucidate the pharmacokinetic process of peramivir after trans-nasal aerosol inhalation to rats and provide useful information for further rational application of this drug formulation.


Asunto(s)
Ácidos Carbocíclicos/administración & dosificación , Ácidos Carbocíclicos/farmacocinética , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Guanidinas/administración & dosificación , Guanidinas/farmacocinética , Pulmón/metabolismo , Ácidos Carbocíclicos/sangre , Administración por Inhalación , Aerosoles , Animales , Disponibilidad Biológica , Inhibidores Enzimáticos/sangre , Guanidinas/sangre , Semivida , Inyecciones Intravenosas , Masculino , Tasa de Depuración Metabólica , Neuraminidasa/antagonistas & inhibidores , Ratas Sprague-Dawley , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA