Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Hepatol ; 80(2): 268-281, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37939855

RESUMEN

BACKGROUND & AIMS: Cholemic nephropathy (CN) is a severe complication of cholestatic liver diseases for which there is no specific treatment. We revisited its pathophysiology with the aim of identifying novel therapeutic strategies. METHODS: Cholestasis was induced by bile duct ligation (BDL) in mice. Bile flux in kidneys and livers was visualized by intravital imaging, supported by MALDI mass spectrometry imaging and liquid chromatography-tandem mass spectrometry. The effect of AS0369, a systemically bioavailable apical sodium-dependent bile acid transporter (ASBT) inhibitor, was evaluated by intravital imaging, RNA-sequencing, histological, blood, and urine analyses. Translational relevance was assessed in kidney biopsies from patients with CN, mice with a humanized bile acid (BA) spectrum, and via analysis of serum BAs and KIM-1 (kidney injury molecule 1) in patients with liver disease and hyperbilirubinemia. RESULTS: Proximal tubular epithelial cells (TECs) reabsorbed and enriched BAs, leading to oxidative stress and death of proximal TECs, casts in distal tubules and collecting ducts, peritubular capillary leakiness, and glomerular cysts. Renal ASBT inhibition by AS0369 blocked BA uptake into TECs and prevented kidney injury up to 6 weeks after BDL. Similar results were obtained in mice with humanized BA composition. In patients with advanced liver disease, serum BAs were the main determinant of KIM-1 levels. ASBT expression in TECs was preserved in biopsies from patients with CN, further highlighting the translational potential of targeting ASBT to treat CN. CONCLUSIONS: BA enrichment in proximal TECs followed by oxidative stress and cell death is a key early event in CN. Inhibiting renal ASBT and consequently BA enrichment in TECs prevents CN and systemically decreases BA concentrations. IMPACT AND IMPLICATIONS: Cholemic nephropathy (CN) is a severe complication of cholestasis and an unmet clinical need. We demonstrate that CN is triggered by the renal accumulation of bile acids (BAs) that are considerably increased in the systemic blood. Specifically, the proximal tubular epithelial cells of the kidney take up BAs via the apical sodium-dependent bile acid transporter (ASBT). We developed a therapeutic compound that blocks ASBT in the kidneys, prevents BA overload in tubular epithelial cells, and almost completely abolished all disease hallmarks in a CN mouse model. Renal ASBT inhibition represents a potential therapeutic strategy for patients with CN.


Asunto(s)
Proteínas Portadoras , Colestasis , Enfermedades Renales , Hepatopatías , Glicoproteínas de Membrana , Transportadores de Anión Orgánico Sodio-Dependiente , Simportadores , Humanos , Ratones , Animales , Colestasis/complicaciones , Colestasis/metabolismo , Riñón/metabolismo , Simportadores/metabolismo , Ácidos y Sales Biliares/metabolismo , Hígado/metabolismo , Conductos Biliares/metabolismo , Hepatopatías/metabolismo , Sodio
2.
Nanotechnology ; 32(20): 205101, 2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-33561838

RESUMEN

Chemotherapy is a mainstay strategy in the management of cancer. Regrettably, current chemotherapeutic agents are cytotoxic not only to cancer cells but also to healthy cells, resulting in dose-limiting serious side effects. Therefore, many researchers are eager to develop new drug delivery systems that may help to decrease the side effects and the target delivery of chemotherapy to cancer cells. One of the epochal drug delivery systems in this field is based on carbon nanotubes technology. The aim of this work is the dual covalent functionalization of single-walled carbon nanotubes (SWCNTs) with doxorubicin (DOX) connected with acid-labile linkage and mannose (Man) as a targeting agent. The characterization of the developed nano-drug by transmission electron microscopy showed good dispersibility of the functionalized SWCNTs with diameters (6-10) nm. Moreover, the percentage of functionalization was determined by thermogravimetric analysis showing 25% of functionalization in the case of SWNCTs-NHN-DOX (7) and 51% for SWCNTs-Man-NHN-DOX (11). The in vitro release profile of Dox from SWNCTs-NHN-DOX (7) showed 45% of the loaded drug was released over 18 h at pH 7.4 and almost complete release at pH 6.4 at 37 °C. However, the in vitro release profile of Dox from SWCNTs-Man-NHN-DOX (11) showed 75% of the loaded drug was released over 5 h at pH 6.4 at 37 °C. The cytotoxic effect of the compounds was studied on liver cancer cells (HepG2) at different concentrations and different pH conditions and was compared with DOX alone. The cytotoxicity of compounds SWCNTs-NHN-DOX (7) and SWCNTs-Man-NHN-DOX (11) was enhanced at pH 6.5, where the cell viability in both test compounds was significantly reduced by almost 50% compared to the cell viability at pH 7.4 for the same test compound Moreover, the pre-incubation of cells with different concentrations of mannose reduced the cytotoxicity of compound (11) by more than 50%, suggesting that the entry of this complex could be at least in part facilitated by mannose receptors, which imparts this complex a kind of selectivity for cancer cells that overexpress this type of receptors.


Asunto(s)
Antineoplásicos , Portadores de Fármacos , Nanotubos de Carbono/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Manosa/química , Manosa/farmacología
3.
Arch Toxicol ; 93(6): 1609-1637, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31250071

RESUMEN

Drug-induced liver injury (DILI) cannot be accurately predicted by animal models. In addition, currently available in vitro methods do not allow for the estimation of hepatotoxic doses or the determination of an acceptable daily intake (ADI). To overcome this limitation, an in vitro/in silico method was established that predicts the risk of human DILI in relation to oral doses and blood concentrations. This method can be used to estimate DILI risk if the maximal blood concentration (Cmax) of the test compound is known. Moreover, an ADI can be estimated even for compounds without information on blood concentrations. To systematically optimize the in vitro system, two novel test performance metrics were introduced, the toxicity separation index (TSI) which quantifies how well a test differentiates between hepatotoxic and non-hepatotoxic compounds, and the toxicity estimation index (TEI) which measures how well hepatotoxic blood concentrations in vivo can be estimated. In vitro test performance was optimized for a training set of 28 compounds, based on TSI and TEI, demonstrating that (1) concentrations where cytotoxicity first becomes evident in vitro (EC10) yielded better metrics than higher toxicity thresholds (EC50); (2) compound incubation for 48 h was better than 24 h, with no further improvement of TSI after 7 days incubation; (3) metrics were moderately improved by adding gene expression to the test battery; (4) evaluation of pharmacokinetic parameters demonstrated that total blood compound concentrations and the 95%-population-based percentile of Cmax were best suited to estimate human toxicity. With a support vector machine-based classifier, using EC10 and Cmax as variables, the cross-validated sensitivity, specificity and accuracy for hepatotoxicity prediction were 100, 88 and 93%, respectively. Concentrations in the culture medium allowed extrapolation to blood concentrations in vivo that are associated with a specific probability of hepatotoxicity and the corresponding oral doses were obtained by reverse modeling. Application of this in vitro/in silico method to the rat hepatotoxicant pulegone resulted in an ADI that was similar to values previously established based on animal experiments. In conclusion, the proposed method links oral doses and blood concentrations of test compounds to the probability of hepatotoxicity.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Administración Oral , Algoritmos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Técnicas In Vitro , Dosis Máxima Tolerada , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/sangre , Farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Máquina de Vectores de Soporte
4.
Nanotechnology ; 29(24): 245101, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29583132

RESUMEN

Single walled carbon nanotubes (SWCNT) are currently under intensive investigation by many labs all over the world for being promising candidates for cancer chemotherapy delivery. On the other hand, combretastatin A4 (CA4) is an anticancer drug that induces cell apoptosis by inhibiting tubulin polymerization. However, it has the disadvantage of low water solubility and the non-selective targeting. Therefore, we aim to create nano-drug from the functionalization of SWCNT covalently with CA4 through click reaction in the presence of tetraethylene glycol linker in order to improve its dispersibility. Scanning electron microscopy and transmission electron microscopy showed good dispersibility of the functionalized SWCNT with diameters of 5-15 nm. Moreover, thermogravometric analysis showed that the efficiency of SWCNT functionalization was around 45%. The in vitro release profile of CA4 at physiological conditions showed that approximately 90% of the loaded drug was released over 50 h. After that MTS test was used to determine the suitable concentration range for the in vitro investigation of the SWCNT-CA4. After that the cytotoxic activity of the SWCNT-CA4 was evaluated by flow cytometry using annexin V/propidium iodide (PI) test. In comparison with free CA4, SWCNT-CA4 treatment demonstrated a significant increase in necrotic cells (around 50%) at the expense of the proportion of the apoptotic cells. Moreover, cell cycle PI test demonstrated that free CA4 and SWCNT-CA4 caused G2/M arrest. However with CA4 treatment higher proportion of cells were in the S-phase while with SWCNT-CA4 treatment greater proportion of cells appeared to be in the G1-phase. Taken together, the provided data suggest that the novel SWCNT-CA4 has a significant anticancer activity that might be superior to that of free CA4.


Asunto(s)
Nanotubos de Carbono/química , Neoplasias/tratamiento farmacológico , Estilbenos/uso terapéutico , Apoptosis , Ciclo Celular , Supervivencia Celular , Química Clic , Liberación de Fármacos , Células HeLa , Humanos , Nanotubos de Carbono/ultraestructura , Neoplasias/patología , Estilbenos/síntesis química , Estilbenos/química , Termogravimetría
5.
Chin J Traumatol ; 20(2): 108-113, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28209447

RESUMEN

PURPOSE: Ephedra alata (E. alata) is perennial tough shrub plant that grows in Palestine and other regions. It is used often in folk's medicine for the treatment of various diseases. In this project, E. alata extract was tested for its ability to improve wound and burn healing. METHODS: An aqueous extract of E. alata was prepared and underwent several phytochemical analyses for the presence of the major classes of phytochemical compounds. After that, a polyethylene glycol-based ointment containing the extract of E. alata was prepared and its wound and burn healing activities were tested in-vivo using an animal model for deep wound and full thickness skin burn. The effect was compared against a placebo ointment. Skin biopsies were evaluated by a blinded clinical histopathologist, in addition to digital analysis. RESULTS: Phytochemical analysis demonstrated the presence of the major classes of phytochemical compounds in the prepared extract including flavonoids, alkaloids, phytosteroids, phenolic compounds, volatile oils and tannins. As compared to placebo ointment, E. alata ointment significantly improved the healing of the wound ulcers, whereas it showed no advantage on the quality of the healing of burn ulcers. CONCLUSION: E. alata extract is rich in phytochemical compounds and can improve wound healing when applied topically.


Asunto(s)
Ephedra , Extractos Vegetales/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Animales , Quemaduras/tratamiento farmacológico , Modelos Animales de Enfermedad , Ephedra/química , Masculino , Mesocricetus , Pomadas , Extractos Vegetales/análisis
6.
Int J Clin Pharmacol Ther ; 54(9): 723-31, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27443664

RESUMEN

BACKGROUND: Treatment of neuropathic pain has always been challenging, not only from the pharmaco-therapeutic/toxicological point of view, but also due to the unpredictable pharmacokinetic (PK) variations among different generic formulations of the same drug, which require further dose optimization. OBJECTIVES: This progressive work aims to evaluate the bioequivalence (BE) of a generic product of 150 mg pregabalin capsule (antineuropathic drug) vs. the reference brand drug Lyrica®. METHOD: An LC-MS/MS bioanalytical method was developed and validated according to the International Conference on Harmonization (ICH) guidelines in order to be used for the analysis of pregabalin in plasma. BE of capsules was tested by comparison against the reference brand capsules in accordance with the requirements of the declarations of Helsinki, the current Good Clinical Practice (GCP) Guidelines and the ICH. The resulting data were compared against corresponding pregabalin data published on other human races. RESULTS: The relationship between concentration and peak area ratio was found to be linear within the range 0.096 - 6.068 µg/mL for pregabalin. The correlation coefficient (r) was equal to 0.9983. Statistical comparison of the main PK parameters showed no significant difference between test and reference. The mean Cmax values for test and reference were 4.290 and 4.164 µg/mL, and the mean AUC0-last values were 24.275 h×µg/mL and 23.674 h×µg/mL, respectively. The 90% CIs of geometric mean ratios (test/reference) for pregabalin were 100.34 - 104.78%, 100.34 - 104.70%, and 95.65 - 110.96% for AUC0-last, AUC0-∞, and Cmax, respectively, thus fall within the international specified BE limit (80 - 125%). Both products were well tolerated by all the volunteers and there were no significant differences on physical examination or in vital signs and laboratory tests between groups. All volunteers completed the study and were discharged in good health. CONCLUSION: The tested generic capsules appear to be bioequivalent to the reference brand and are expected to have a similar efficacy and safety profile.


Asunto(s)
Analgésicos/farmacocinética , Árabes , Medicamentos Genéricos/farmacocinética , Pregabalina/farmacocinética , Adolescente , Adulto , Analgésicos/administración & dosificación , Analgésicos/efectos adversos , Área Bajo la Curva , Cápsulas , Cromatografía Liquida/métodos , Estudios Cruzados , Medicamentos Genéricos/administración & dosificación , Medicamentos Genéricos/efectos adversos , Humanos , Persona de Mediana Edad , Pregabalina/administración & dosificación , Pregabalina/efectos adversos , Espectrometría de Masas en Tándem/métodos , Equivalencia Terapéutica , Adulto Joven
7.
J Mol Cell Cardiol ; 88: 39-54, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26392029

RESUMEN

Cardiac remodeling, a hallmark of heart disease, is associated with intense auto- and paracrine signaling leading to cardiac fibrosis. We hypothesized that the specific mediator of Gq/11-dependent RhoA activation p63RhoGEF, which is expressed in cardiac fibroblasts, plays a role in the underlying processes. We could show that p63RhoGEF is up-regulated in mouse hearts subjected to transverse aortic constriction (TAC). In an engineered heart muscle model (EHM), p63RhoGEF expression in cardiac fibroblasts increased resting and twitch tensions, and the dominant negative p63ΔN decreased both. In an engineered connective tissue model (ECT), p63RhoGEF increased tissue stiffness and its knockdown as well as p63ΔN reduced stiffness. In 2D cultures of neonatal rat cardiac fibroblasts, p63RhoGEF regulated the angiotensin II (Ang II)-dependent RhoA activation, the activation of the serum response factor, and the expression and secretion of the connective tissue growth factor (CTGF). All these processes were inhibited by the knockdown of p63RhoGEF or by p63ΔN likely based on their negative influence on the actin cytoskeleton. Moreover, we show that p63RhoGEF also regulates CTGF in engineered tissues and correlates with it in the TAC model. Finally, confocal studies revealed a closely related localization of p63RhoGEF and CTGF in the trans-Golgi network.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/metabolismo , Miocardio/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factor de Respuesta Sérica/genética , Proteína de Unión al GTP rhoA/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Animales Recién Nacidos , Aorta/cirugía , Comunicación Autocrina/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Constricción , Femenino , Fibroblastos/patología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Cardiovasculares , Miocardio/patología , Comunicación Paracrina/genética , Ratas , Ratas Wistar , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Remodelación Ventricular , Proteína de Unión al GTP rhoA/metabolismo , Red trans-Golgi/metabolismo , Red trans-Golgi/ultraestructura
8.
ACS Biomater Sci Eng ; 10(6): 3833-3841, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38747490

RESUMEN

This study explores the realm of personalized medicine by investigating the utilization of 3D-printed dosage forms, specifically focusing on patient-specific enteric capsules designed for the modified release of ketoprofen, serving as a model drug. The research investigates two distinct scenarios: the modification of drug release from 3D-printed capsules crafted from hydroxypropyl methylcellulose phthalate:polyethylene glycol (HPMCP:PEG) and poly(vinyl alcohol) (PVA), tailored for pH sensitivity and delayed release modes, respectively. Additionally, a novel ketoprofen-loaded self-nanoemulsifying drug delivery system (SNEDDS) based on pomegranate seed oil (PSO) was developed, characterized, and employed as a fill material for the capsules. Through the preparation and characterization of the HPMCP:PEG based filament via the hot-melt extrusion method, the study thoroughly investigated its thermal and mechanical properties. Notably, the in vitro drug release analysis unveiled the intricate interplay between ketoprofen release, polymer type, and capsule thickness. Furthermore, the incorporation of ketoprofen into the SNEDDS exhibited an enhancement in its in vitro cylooxygenase-2 (COX-2) inhibitory activity. These findings collectively underscore the potential of 3D printing in shaping tailored drug delivery systems, thereby contributing significantly to the advancement of personalized medicine.


Asunto(s)
Cápsulas , Liberación de Fármacos , Emulsiones , Cetoprofeno , Medicina de Precisión , Impresión Tridimensional , Cetoprofeno/química , Medicina de Precisión/métodos , Humanos , Emulsiones/química , Polietilenglicoles/química , Sistemas de Liberación de Medicamentos/métodos , Preparaciones de Acción Retardada , Metilcelulosa/química , Metilcelulosa/análogos & derivados , Alcohol Polivinílico/química
9.
RSC Adv ; 13(10): 7000-7008, 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36874935

RESUMEN

Several approaches, including plasmid transfection and viral vectors, were used to deliver genes into cells for therapeutic and experimental purposes. However, due to the limited efficacy and questionable safety issues, researchers are looking for better new approaches. Over the past decade, graphene has attracted tremendous attention in versatile medical applications, including gene delivery, which could be safer than the traditional viral vectors. This work aims to covalently functionalize pristine graphene sheets with a polyamine to allow the loading of plasmid DNA (pDNA) and enhance its delivery into cells. Graphene sheets were successfully covalently functionalized with a derivative of tetraethylene glycol connected to polyamine groups to improve their water dispersibility and capacity to interact with the pDNA. The improved dispersibility of the graphene sheets was demonstrated visually and by transmission electron microscopy. Also, it was shown by thermogravimetric analysis that the degree of functionalization was about 58%. Moreover, the surface charge of the functionalized graphene was +29 mV as confirmed by zeta potential analysis. The complexion of f-graphene with pDNA was achieved at a relatively low mass ratio (10 : 1). The incubation of HeLa cells with f-graphene loaded with pDNA that encodes enhanced green fluorescence protein (eGFP) resulted in the detection of fluorescence signal in the cells within one hour. f-Graphene showed no toxic effect in vitro. Density functional theory (DFT) and quantum theory of atoms in molecules (QTAIM) calculations revealed strong binding with ΔH 298 = 74.9 kJ mol-1. QTAIM between the f-graphene and a simplified model of pDNA. Taken together, the developed functionalized graphene could be used for the development of a new non-viral gene delivery system.

10.
Sci Rep ; 12(1): 12062, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35835926

RESUMEN

Tissue engineering is one of the hot topics in recent research that needs special requirements. It depends on the development of scaffolds that allow tissue formation with certain characteristics, carbon nanotubes (CNTs)-collagen composite attracted the attention of the researchers with this respect. However, CNTs suffer from low water dispersibility, which hampered their utilization. Therefore, we aim to functionalize CNTs non-covalently with pyrene moiety using an appropriate hydrophilic linker derivatized from polyethylene glycol (PEG) terminated with hydroxyl or carboxyl group to disperse them in water. The functionalization of the CNTs is successfully confirmed by TEM, absorption spectroscopy, TGA, and zeta potential analysis. 3T3 cells-based engineered connective tissues (ECTs) are generated with different concentrations of the functionalized CNTs (f-CNTs). These tissues show a significant enhancement in electrical conductivity at a concentration of 0.025%, however, the cell viability is reduced by about 10 to 20%. All ECTs containing f-CNTs show a significant reduction in tissue fibrosis and matrix porosity relative to the control tissues. Taken together, the developed constructs show great potential for further in vivo studies as engineered tissue.


Asunto(s)
Nanotubos de Carbono , Animales , Ratones , Nanotubos de Carbono/química , Polietilenglicoles/química , Porosidad , Ingeniería de Tejidos/métodos , Agua
11.
Chem Biol Interact ; 351: 109728, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34717914

RESUMEN

An in vitro/in silico method that determines the risk of human drug induced liver injury in relation to oral doses and blood concentrations of drugs was recently introduced. This method utilizes information on the maximal blood concentration (Cmax) for a specific dose of a test compound, which can be estimated using physiologically-based pharmacokinetic modelling, and a cytotoxicity test in cultured human hepatocytes. In the present study, we analyzed if the addition of an assay that measures the inhibition of bile acid export carriers, like BSEP and/or MRP2, to the existing method improves the differentiation of hepatotoxic and non-hepatotoxic compounds. Therefore, an export assay for 5-chloromethylfluorescein diacetate (CMFDA) was established. We tested 36 compounds in a concentration-dependent manner for which the risk of hepatotoxicity for specific oral doses and the capacity to inhibit hepatocyte export carriers are known. Compared to the CTB cytotoxicity test, substantially lower EC10 values were obtained using the CMFDA assay for several known BSEP and/or MRP2 inhibitors. To quantify if the addition of the CMFDA assay to our test system improves the overall separation of hepatotoxic from non-hepatotoxic compounds, the toxicity separation index (TSI) was calculated. We obtained a better TSI using the lower alert concentration from either the CMFDA or the CTB test (TSI: 0.886) compared to considering the CTB test alone (TSI: 0.775). In conclusion, the data show that integration of the CMFDA assay with an in vitro test battery improves the differentiation of hepatotoxic and non-hepatotoxic compounds in a set of compounds that includes bile acid export carrier inhibitors.


Asunto(s)
Citotoxinas/toxicidad , Hepatocitos/efectos de los fármacos , Pruebas de Toxicidad/métodos , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/antagonistas & inhibidores , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas , Fluoresceínas/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos/metabolismo
12.
Int J Pharm Compd ; 25(6): 523-527, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34807848

RESUMEN

Ivermectin is a gamma amino butyric acid (GABA)-gated-Cl-channels modulator, which has been used orally in the treatment of numerous parasitic infections. The study target was to set up a stable, efficient 0.2% w/v ivermectin solution, which would be achieved from pure ivermectin powder as a source of the active pharmaceutical ingredient. Several trial solutions were prepared. The most fitting solution, with respect to its organoleptic properties, was chosen for additional investigation, including the solution's physical stability. Two storage conditions, room temperature (25°C, 60% relative humidity) and accelerated stability chambers (40°C, 75% relative humidity) were subjected to guarantee the physical stability of the solution through the 3-month study period. Furthermore, other quality tests were assessed, (e.g., pH, assay, organoleptic properties, microbial contamination) for the same latter period. Quantification of ivermectin was validated utilizing a high-performance liquid chromatography analytical method. The adopted solution showed accepted organoleptic properties. The pH of the solutions was approximately 5 and remained unchanged during the stability study. The mean percent of remained ivermectin solution was close to 97% ± 0.2 at room temperature. Ivermectin solution was additionally tested for microbial contamination, and it was free from any microbial contamination (E. coli bacteria: Negative/mL, yeast and molds count: <10 cfu/mL and aerobic microbial count results in <10 cfu/mL). The adopted formula showed the best physical stability within at least the three months of storage at both room and accelerated conditions. Ivermectin solution was successfully prepared from its pure powder. This formula provides a stable, efficient oral solution for those who suffer from swallowing difficulties or patients in the intensive care unit who cannot receive the medication in a solid dosage form. Using the suggested formula, community and hospital pharmacists could prepare an effective, high quality ivermectin oral solution using pure ivermectin powder.


Asunto(s)
Escherichia coli , Ivermectina , Cromatografía Líquida de Alta Presión , Estabilidad de Medicamentos , Humanos , Resultado del Tratamiento
13.
Int J Nanomedicine ; 16: 989-1000, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33633447

RESUMEN

BACKGROUND: Under certain conditions, the physiological repair of connective tissues might fail to restore the original structure and function. Optimized engineered connective tissues (ECTs) with biophysical properties adapted to the target tissue could be used as a substitution therapy. This study aimed to investigate the effect of ECT enforcement by a complex of multiwall carbon nanotubes with chitosan (C-MWCNT) to meet in vivo demands. MATERIALS AND METHODS: ECTs were constructed from human foreskin fibroblasts (HFF-1) in collagen type I and enriched with the three different percentages 0.025, 0.05 and 0.1% of C-MWCNT. Characterization of the physical properties was performed by biomechanical studies using unidirectional strain. RESULTS: Supplementation with 0.025% C-MWCNT moderately increased the tissue stiffness, reflected by Young's modulus, compared to tissues without C-MWCNT. Supplementation of ECTs with 0.1% C-MWCNT reduced tissue contraction and increased the elasticity and the extensibility, reflected by the yield point and ultimate strain, respectively. Consequently, the ECTs with 0.1% C-MWCNT showed a higher resilience and toughness as control tissues. Fluorescence tissue imaging demonstrated the longitudinal alignment of all cells independent of the condition. CONCLUSION: Supplementation with C-MWCNT can enhance the biophysical properties of ECTs, which could be advantageous for applications in connective tissue repair.


Asunto(s)
Quitosano/farmacología , Tejido Conectivo/fisiología , Nanotubos de Carbono/química , Ingeniería de Tejidos , Animales , Fenómenos Biomecánicos , Bovinos , Línea Celular , Quitosano/química , Módulo de Elasticidad , Fibroblastos/efectos de los fármacos , Humanos
14.
ACS Biomater Sci Eng ; 6(1): 112-121, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33463246

RESUMEN

The ongoing spread of multi-drug-resistant bacteria over the past few decades necessitates collateral efforts to develop new classes of antibacterial agents with different mechanisms of action. The utilization of graphene nanosheets has recently gained attention with this respect. Herein, we have synthesized and tested the antibacterial activity of an array of graphene materials covalently functionalized with hydroxyl-, amine-, or carboxyl-containing groups. Fourier transform infrared spectroscopy and transmission electron microscopy confirmed successful functionalization of the few-layer graphene (FLG). The percentage of weight loss was measured by thermogravimetric analysis, which was found to be 22%, 23%, and 37% for FLG-TEG-OH, FLG-NH2, and FLG-DEG-COOH, respectively. In comparison with pristine graphene sheets, the functionalized few-layer graphene (f-FLG) materials gained an adequate dispersibility in water as confirmed by ζ potential analysis. Moreover, there was a significant improvement in the antibacterial activity against Staphylococcus aureus and Escherichia coli, where all f-FLG compounds were able to suppress bacterial growth, with a complete suppression achieved by FLG-DEG-COOH. The minimum inhibitory concentration (MIC) was 250 µg mL-1 for both FLG-TEG-OH and FLG-NH2, while it was 125 µg mL-1 for FLG-DEG-COOH. The glutathione oxidation test demonstrated an oxidative stress activity by all f-FLG compounds. However, FLG-DEG-COOH demonstrated the highest reduction in glutathione activity. FLG-DEG-COOH and FLG-TEG-OH showed adequate biocompatibility and hemocompatibility. The chemical functionalization of graphene might be a step toward the foundation of an effective class of antimicrobial agents.


Asunto(s)
Antibacterianos , Grafito , Antibacterianos/farmacología , Escherichia coli , Grafito/farmacología , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus
15.
RSC Adv ; 9(2): 1055-1061, 2019 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-35517625

RESUMEN

Cancer is a major worldwide health problem, for which chemotherapy is a common treatment option. However drug toxicity and the development of resistance to chemotherapy are two main challenges associated with the traditional anticancer drugs. Combined pharmacological therapy based on different mechanisms might be an effective strategy in cancer treatment, and could exhibit a synergistic therapeutic efficacy. Herein, we aim to combine combretastatin A4 (CA4) and camptothecin (Cpt) chemically into a codrug through two hydrophilic linkers utilizing click chemistry to improve their water solubility and anticancer activity. The synthesized amphiphilic structure could self-assemble into a micelle structure as confirmed by atomic force microscopy (AFM) and dynamic light scattering (DLS), which showed a high stability and improved water solubility at pH 7.4, with a low critical micelle concentration (CMC) value of 0.9 mM. Moreover, in vitro hydrolysis was observed upon incubation of the hybrid compound with an esterase enzyme, which suggested a complete disassembly into the starting active drugs. Finally, cytotoxicity studies on HeLa cancer cells showed that the codrug demonstrated an enhanced (five fold) cytotoxicity as compared with the free drugs. In addition the combination index (CI) was <1, which suggests a synergistic activity for the codrug. Moreover, the tested concentrations of the codrug were not significantly cytotoxic to a noncancerous fibroblast cell line. The imaging of HeLa cells treated with FITC-loaded micelles showed a rapid internalization. In conclusion, the codrug of CA4 and Cpt might be a potential novel anticancer drug as it demonstrated a synergistic cytotoxic activity that might spare noncancerous cells.

16.
Fundam Clin Pharmacol ; 32(1): 14-25, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28833476

RESUMEN

Cardiac remodeling (cardiac hypertrophy and fibrosis) is a hallmark of heart failure (HF). It can be induced by the abnormal elevation of several endogenous factors including angiotensin II (Ang II), which is generated from its precursor angiotensin I (Ang I) by the action of angiotensin-converting enzyme. The inhibition of this enzyme or the blockade of the Ang II receptors demonstrated a high clinical value against the progression of HF. Ang I and Ang II may also be converted into angiotensin 1-7 (Ang 1-7) and angiotensin 1-9 (Ang 1-9), respectively, by the action of angiotensin-converting enzyme 2. Both derivatives demonstrated a promising anticardiac remodeling activity especially against the detrimental effects of Ang II. This manuscript thoroughly reviews the available in vitro and in vivo data on Ang 1-7 and Ang 1-9 in the context of the treatment of HF and discusses the associated molecular mechanisms and the trials to clinically utilize Ang 1-7 mimetics for the treatment of that disease.


Asunto(s)
Angiotensina II/metabolismo , Angiotensina I/uso terapéutico , Fármacos Cardiovasculares/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Peptidil-Dipeptidasa A/metabolismo , Angiotensina I/efectos adversos , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Fármacos Cardiovasculares/efectos adversos , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Humanos , Imitación Molecular , Terapia Molecular Dirigida , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
17.
Int J Nanomedicine ; 13: 7195-7206, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30510412

RESUMEN

BACKGROUND: Impaired wound healing is commonly associated with many health problems, including diabetes, bedsores and extensive burns. In such cases, healing often takes a long time, which subjects patients to various complications. This study aims to investigate whether single-wall or multi-wall carbon nanotubes complexed with chitosan hydrogel can improve wound healing. MATERIALS AND METHODS: Initially, the effects of the complexes on the viability and functionality of fibroblasts were investigated in engineered connective tissues. Then, their activity on wound healing was investigated in a mouse model with induced full-thickness wounds, in which the wounds were treated daily with these complexes. Finally, the effect of the complexes on collagen deposition by fibroblasts was investigated in vitro. RESULTS: The engineered connective tissue studies showed that fibroblasts were viable in the presence of the complexes and were still able to effectively organize and contract the extracellular matrix. In vivo data showed that both types of complexes improved the re-epithelialization of the healing wounds; however, they also increased the percentage of wounds with higher fibrosis. In particular, the chitosan-multi-wall carbon nanotube complex significantly enhanced the extensiveness of this fibrosis, which is in line with in vitro data showing a concentration-dependent enhancement of collage deposition by these complexes. These observations were associated with an increase in inflammatory signs in the wound bed. CONCLUSION: Single-wall and multi-wall carbon nanotubes complexed with chitosan improved the re-epithelialization of wounds, but an increase in fibrosis was detected.


Asunto(s)
Quitosano/farmacología , Nanotubos de Carbono/química , Cicatrización de Heridas/efectos de los fármacos , Animales , Colágeno/metabolismo , Tejido Conectivo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Inflamación/patología , Masculino , Ratones , Nanotubos de Carbono/ultraestructura , Ingeniería de Tejidos
18.
Am J Health Syst Pharm ; 74(19): 1579-1583, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28947528

RESUMEN

PURPOSE: The stability of an extemporaneously prepared rosuvastatin suspension stored over 30 days under various storage conditions was evaluated. METHODS: Rosuvastatin suspension was extemporaneously prepared using commercial rosuvastatin tablets as the source of active pharmaceutical ingredient. The organoleptic properties, dissolution profile, and stability of the formulation were investigated. For the stability studies, samples of the suspension were stored under 2 storage conditions, room temperature (25 °C and 60% relative humidity) and accelerated stability chambers (40 °C and 75% relative humidity). Viscosity, pH, organoleptic properties, and microbial contamination were evaluated according to the approved specifications. High-performance liquid chromatography was used for the analysis and quantification of rosuvastatin in selected samples. Microbiological investigations were also conducted. RESULTS: The prepared suspension showed acceptable organoleptic properties. It showed complete release of rosuvastatin within 15 minutes. The pH of the suspension was 9.8, which remained unchanged during the stability studies. The microbiological investigations demonstrated that the preparation was free of any microbial contamination. In addition, the suspension showed stability within at least the period of use of a 100-mL rosuvastatin bottle. CONCLUSION: Extemporaneously prepared rosuvastatin 20-mg/mL suspension was stable for 30 days when stored at room temperature.


Asunto(s)
Composición de Medicamentos/métodos , Almacenaje de Medicamentos/métodos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/síntesis química , Rosuvastatina Cálcica/síntesis química , Administración Oral , Cromatografía Líquida de Alta Presión/métodos , Cromatografía Líquida de Alta Presión/normas , Composición de Medicamentos/normas , Liberación de Fármacos , Estabilidad de Medicamentos , Almacenaje de Medicamentos/normas , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/análisis , Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/análisis , Solubilidad , Suspensiones
19.
Expert Rev Respir Med ; 10(9): 943-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27485393

RESUMEN

INTRODUCTION: Montelukast is recommended for the treatment of asthma, exercise -induced bronchospasm and allergic rhinitis. Several trials demonstrated potential therapeutic effects in other respiratory conditions, and different animal-model-based studies explored potential pharmacological actions in non-respiratory conditions. AREAS COVERED: Clinical investigations on the pharmacotherapeutic effects of montelukast, in addition to in-vivo studies on animal models of non-respiratory diseases. The data discussed in this review were mainly obtained from clinical randomized trials, real-life studies, and studies based on animal models as approve of concept. As a condition, all of the discussed articles were published in journals cited by Pubmed. Expert commentary: The current clinical data are in favor of montelukast use in the management of chronic asthma as an add-on or alternative therapy to the inhaled corticosteroids. Further clinical trials are required to confirm the effectiveness and feasibility of montelukast for the treatment of conditions other than the current clinical indications.


Asunto(s)
Acetatos/uso terapéutico , Asma/tratamiento farmacológico , Antagonistas de Leucotrieno/uso terapéutico , Quinolinas/uso terapéutico , Corticoesteroides/uso terapéutico , Antiasmáticos/uso terapéutico , Asma Inducida por Ejercicio/tratamiento farmacológico , Ciclopropanos , Humanos , Sulfuros
20.
PLoS One ; 10(10): e0137519, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26448568

RESUMEN

INTRODUCTION: RhoA has been shown to be beneficial in cardiac disease models when overexpressed in cardiomyocytes, whereas its role in cardiac fibroblasts (CF) is still poorly understood. During cardiac remodeling CF undergo a transition towards a myofibroblast phenotype thereby showing an increased proliferation and migration rate. Both processes involve the remodeling of the cytoskeleton. Since RhoA is known to be a major regulator of the cytoskeleton, we analyzed its role in CF and its effect on myofibroblast characteristics in 2 D and 3D models. RESULTS: Downregulation of RhoA was shown to strongly affect the actin cytoskeleton. It decreased the myofibroblast marker α-sm-actin, but increased certain fibrosis-associated factors like TGF-ß and collagens. Also, the detailed analysis of CTGF expression demonstrated that the outcome of RhoA signaling strongly depends on the involved stimulus. Furthermore, we show that proliferation of myofibroblasts rely on RhoA and tubulin acetylation. In assays accessing three different types of migration, we demonstrate that RhoA/ROCK/Dia1 are important for 2D migration and the repression of RhoA and Dia1 signaling accelerates 3D migration. Finally, we show that a downregulation of RhoA in CF impacts the viscoelastic and contractile properties of engineered tissues. CONCLUSION: RhoA positively and negatively influences myofibroblast characteristics by differential signaling cascades and depending on environmental conditions. These include gene expression, migration and proliferation. Reduction of RhoA leads to an increased viscoelasticity and a decrease in contractile force in engineered cardiac tissue.


Asunto(s)
Miofibroblastos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas/fisiología , Reacción en Cadena de la Polimerasa , Ratas , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA