Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Mol Med ; 55(11): 2320-2331, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37945923

RESUMEN

Nucleic acid sensing is involved in viral infections, immune response-related diseases, and therapeutics. Based on the composition of nucleic acids, nucleic acid sensors are defined as DNA or RNA sensors. Pathogen-associated nucleic acids are recognized by membrane-bound and intracellular receptors, known as pattern recognition receptors (PRRs), which induce innate immune-mediated antiviral responses. PRR activation is tightly regulated to eliminate infections and prevent abnormal or excessive immune responses. Nucleic acid sensing is an essential mechanism in tumor immunotherapy and gene therapies that target cancer and infectious diseases through genetically engineered immune cells or therapeutic nucleic acids. Nucleic acid sensing supports immune cells in priming desirable immune responses during tumor treatment. Recent studies have shown that nucleic acid sensing affects the efficiency of gene therapy by inhibiting translation. Suppression of innate immunity induced by nucleic acid sensing through small-molecule inhibitors, virus-derived proteins, and chemical modifications offers a potential therapeutic strategy. Herein, we review the mechanisms and regulation of nucleic acid sensing, specifically covering recent advances. Furthermore, we summarize and discuss recent research progress regarding the different effects of nucleic acid sensing on therapeutic efficacy. This study provides insights for the application of nucleic acid sensing in therapy.


Asunto(s)
Neoplasias , Ácidos Nucleicos , Humanos , Ácidos Nucleicos/uso terapéutico , Ácidos Nucleicos/metabolismo , Transducción de Señal , Inmunidad Innata , Receptores de Reconocimiento de Patrones/metabolismo , Neoplasias/genética , Neoplasias/terapia
2.
Leukemia ; 37(4): 807-819, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36932165

RESUMEN

Clinical effect of donor-derived natural killer cell infusion (DNKI) after HLA-haploidentical hematopoietic cell transplantation (HCT) was evaluated in high-risk myeloid malignancy in phase 2, randomized trial. Seventy-six evaluable patients (aged 21-70 years) were randomized to receive DNKI (N = 40) or not (N = 36) after haploidentical HCT. For the HCT conditioning, busulfan, fludarabine, and anti-thymocyte globulin were administered. DNKI was given twice 13 and 20 days after HCT. Four patients in the DNKI group failed to receive DNKI. In the remaining 36 patients, median DNKI doses were 1.0 × 108/kg and 1.4 × 108/kg on days 13 and 20, respectively. Intention-to-treat analysis showed a lower disease progression for the DNKI group (30-month cumulative incidence, 35% vs 61%, P = 0.040; subdistribution hazard ratio, 0.50). Furthermore, at 3 months after HCT, the DNKI patients showed a 1.8- and 2.6-fold higher median absolute blood count of NK and T cells, respectively. scRNA-sequencing analysis in seven study patients showed that there was a marked increase in memory-like NK cells in DNKI patients which, in turn, expanded the CD8+ effector-memory T cells. In high-risk myeloid malignancy, DNKI after haploidentical HCT reduced disease progression. This enhanced graft-vs-leukemia effect may be related to the DNKI-induced, post-HCT expansion of NK and T cells. Clinical trial number: NCT02477787.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Humanos , Interleucina-15 , Enfermedad Injerto contra Huésped/patología , Células Asesinas Naturales/patología , Progresión de la Enfermedad , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/patología , Acondicionamiento Pretrasplante
3.
Nat Immunol ; 24(3): 463-473, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36624164

RESUMEN

The formation of an immunological synapse (IS) is essential for natural killer (NK) cells to eliminate target cells. Despite an advanced understanding of the characteristics of the IS and its formation processes, the mechanisms that regulate its stability via the cytoskeleton are unclear. Here, we show that Nogo receptor 1 (NgR1) has an important function in modulating NK cell-mediated killing by destabilization of IS formation. NgR1 deficiency or blockade resulted in improved tumor control of NK cells by enhancing NK-to-target cell contact stability and regulating F-actin dynamics during IS formation. Patients with tumors expressing abundant NgR1 ligand had poor prognosis despite high levels of NK cell infiltration. Thus, our study identifies NgR1 as an immune checkpoint in IS formation and indicates a potential approach to improve the cytolytic function of NK cells in cancer immunotherapy.


Asunto(s)
Sinapsis Inmunológicas , Neoplasias , Humanos , Receptores de Células Asesinas Naturales , Receptor Nogo 1 , Células Asesinas Naturales , Actinas , Neoplasias/patología
4.
PLoS One ; 17(12): e0277188, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36508415

RESUMEN

Various substances, including collagen (Naticol®) and ascorbic acid, that inhibit and prevent skin aging have been studied. Collagen prevents skin aging, has anti-inflammatory effects, and assists in normal wound healing. Ascorbic acid is a representative antioxidant that plays a role in collagen synthesis. To achieve a synergistic effect of collagen and ascorbic acid on all skin types, we prepared a product named "TEENIALL." In addition, we used a container to separate ascorbic acid and collagen to prevent the oxidation of ascorbic acid. To confirm the effects of TEENIALL, we first confirmed its penetrability in fibroblasts, keratinocytes, melanocyte, and human skin tissues. Thereafter, we confirmed the collagen synthesis ability in normal human fibroblasts. Based on the results of in vitro tests, we conducted a clinical trial (KCT0006916) on female volunteers, aged 40 to 59 years, with skin wrinkles and hyperpigmentation, to evaluate the effects of the product in improving skin wrinkles, skin lifting, and pigmentation areas before using the product, and after 2 and 4 weeks of using the product. The values of nine wrinkle parameters that were evaluated decreased and those for skin sagging, pigmentation, dermal density, and mechanical imprint (pressure) relief were improved. Skin wrinkle and pigmentation were evaluated to ensure that the improvement effect was maintained even after 1 week of discontinuing the product use. The evaluation confirmed that the effects were sustained compared to those after 4 weeks of using the product. Additionally, skin wrinkles, skin lifting, radiance, and moisture content in the skin improved immediately after using the product once. Based on the results of in vitro and ex vivo experiments and the clinical trial, we show that the product containing ascorbic acid and collagen was effective in alleviating skin aging.


Asunto(s)
Ácido Ascórbico , Envejecimiento de la Piel , Femenino , Humanos , Ácido Ascórbico/farmacología , Colágeno/farmacología , Piel , Fibroblastos
5.
Front Immunol ; 13: 1089369, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713381

RESUMEN

Natural killer (NK) cells are immune effector cells with outstanding features for adoptive immunotherapy. Immune effector cells with chimeric antigen receptors (CARs) are promising targeted therapeutic agents for various diseases. Because tumor cells exhibit heterogeneous antigen expression and lose cell surface antigen expression during malignant progression, many CARs fixed against only one antigen have limited efficacy and are associated with tumor relapse. To expand the utility of CAR-NK cells, we designed a split and universal cotinine-CAR (Cot-CAR) system, comprising a Cot-conjugator and NK92 cells (α-Cot-NK92 cells) engineered with a CAR containing an anti-Cot-specific single-chain variable fragment and intracellular signaling domain. The efficacy of the Cot-CAR system was assessed in vitro using a cytolysis assay against various tumor cells, and its single- or multiple- utility potential was demonstrated using an in vivo lung metastasis model by injecting A549-Red-Fluc cells. The α-Cot-NK92 cells could switch targets, logically respond to multiple antigens, and tune cytolytic activation through the alteration of conjugators without re-engineering. Therefore the universal Cot-CAR system is useful for enhancing specificity and diversity of antigens, combating relapse, and controlling cytolytic activity. In conclusion, this universal Cot-CAR system reveals that multiple availability and controllability can be generated with a single, integrated system.


Asunto(s)
Cotinina , Receptores Quiméricos de Antígenos , Humanos , Cotinina/metabolismo , Recurrencia Local de Neoplasia/tratamiento farmacológico , Células Asesinas Naturales , Inmunoterapia Adoptiva , Antígenos/metabolismo
6.
Life Sci Alliance ; 4(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34187874

RESUMEN

High endothelial venules (HEVs) effectively recruit circulating lymphocytes from the blood to lymph nodes. HEVs have endothelial cells (ECs) and perivascular sheaths consisting of fibroblastic reticular cells (FRCs). Yet, post-luminal lymphocyte migration steps are not well elucidated. Herein, we performed intravital imaging to investigate post-luminal T- and B-cell migration in popliteal lymph node, consisting of trans-EC migration, crawling in the perivascular channel (a narrow space between ECs and FRCs) and trans-FRC migration. The post-luminal migration of T cells occurred in a PNAd-dependent manner. Remarkably, we found hot spots for the trans-EC and trans-FRC migration of T- and B cells. Interestingly, T- and B cells preferentially shared trans-FRC migration hot spots but not trans-EC migration hot spots. Furthermore, the trans-FRC T-cell migration was confined to fewer sites than trans-EC T-cell migration, and trans-FRC migration of T- and B cells preferentially occurred at FRCs covered by CD11c+ dendritic cells in HEVs. These results suggest that HEV ECs and FRCs with perivascular DCs delicately regulate T- and B-cell entry into peripheral lymph nodes.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos T/metabolismo , Vénulas/inmunología , Animales , Microscopía Intravital , Ganglios Linfáticos/inmunología , Ratones , Migración Transendotelial y Transepitelial
7.
Mol Cells ; 44(1): 38-49, 2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-33510050

RESUMEN

Airway mucus secretion is an essential innate immune response for host protection. However, overproduction and hypersecretion of mucus, mainly composed of the gel- forming MUC5AC protein, are significant risk factors for patients with asthma and chronic obstructive pulmonary disease (COPD). The transforming growth factor ß (TGFß) signaling pathway negatively regulates MUC5AC expression; however, the underlying molecular mechanism is not fully understood. Here, we showed that TGFß significantly reduces the expression of MUC5AC mRNA and its protein in NCI-H292 cells, a human mucoepidermoid carcinoma cell line. This reduced MUC5AC expression was restored by a TGFß receptor inhibitor (SB431542), but not by the inhibition of NF-κB (BAY11-7082 or Triptolide) or PI3K (LY294002) activities. TGFß-activated Smad3 dose-dependently bound to MUC5AC promoter. Notably, TGFß-activated Smad3 recruited HDAC2 and facilitated nuclear translocation of HDAC2, thereby inducing the deacetylation of NF-κB at K310, which is essential for a reduction in NF-κB transcriptional activity. Both TGFß-induced nuclear translocation of Smad3/HDAC2 and deacetylation of NF-κB at K310 were suppressed by a Smad3 inhibitor (SIS3). These results suggest that the TGFß-activated Smad3/HDAC2 complex is an essential negative regulator for MUC5AC expression and an epigenetic regulator for NF-κB acetylation. Therefore, these results collectively suggest that modulation of the TGFß1/Smad3/HDAC2/NF-κB pathway axis can be a promising way to improve lung function as a treatment strategy for asthma and COPD.


Asunto(s)
Histona Desacetilasa 2/metabolismo , Lisina/metabolismo , Mucina 5AC/genética , FN-kappa B/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Acetilación/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , Mucina 5AC/metabolismo , Regiones Promotoras Genéticas/genética
8.
Biomaterials ; 247: 119960, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32278822

RESUMEN

Adoptive transfer of natural killer (NK) cells is becoming one of the most important parts of cancer immunotherapy. However, recent accomplishments have focused on the improvement of the targeting effects based on the engineering of chimeric antigen receptors (CARs) on cell surfaces. Despite the large quantity of therapeutic cells required for clinical applications, the technology for ex vivo expansion is not well developed. Herein, a three-dimensional (3D) engineered hyaluronic acid-based niche for cell expansion (3D-ENHANCE) is introduced. Compared with the conventional two-dimensional (2D) method, NK-92 cell lines and human EGFR-specific (CAR)-NK cells cultured in 3D-ENHANCE yield favorable mRNA expressions, elevated cytokine release, upregulated proliferative and tumor-lytic abilities, and result in enhanced antitumor efficacy. Furthermore, controllable degradation rates can be realized by tuning the formulation of 3D-ENHANCE so that it can be applied as an implantable cell reservoir at surgical sites. In vivo results with the incompletely resected MDA-MB-231 model confirm that the peri-operative implantation of 3D-ENHANCE prevents the relapse and metastases after surgery. Overall, 3D-ENHANCE presents an effective cytokine-free niche for ex vivo expansion and postsurgical treatment that enhances the low-therapeutic efficacy of human NK cells.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Ácido Hialurónico , Inmunoterapia , Células Asesinas Naturales , Neoplasias/terapia
9.
Cell Signal ; 34: 110-120, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28323005

RESUMEN

The nuclear factor kappa B (NF-κB) pathway is pivotal in controlling survival and apoptosis of cancer cells. Macrophage migration inhibitory factor (MIF), a cytokine that regulates the immune response and tumorigenesis under inflammatory conditions, is upregulated in various tumors. However, the intracellular functions of MIF are unclear. In this study, we found that MIF directly interacted with thioredoxin-interacting protein (TXNIP), a tumor suppressor and known inhibitor of NF-κB activity, and MIF significantly induced NF-κB activation. MIF competed with TXNIP for NF-κB activation, and the intracellular MIF induced NF-κB target genes, including c-IAP2, Bcl-xL, ICAM-1, MMP2 and uPA, by inhibiting the interactions between TXNIP and HDACs or p65. Furthermore, we identified the interaction motifs between MIF and TXNIP via site-directed mutagenesis of their cysteine (Cys) residues. Cys57 and Cys81 of MIF and Cys36 and Cys120 of TXNIP were responsible for the interaction. MIF reversed the TXNIP-induced suppression of cell proliferation and migration. Overall, we suggest that MIF induces NF-κB activity by counter acting the inhibitory effect of TXNIP on the NF-κB pathway via direct interaction with TXNIP. These findings reveal a novel intracellular function of MIF in the progression of cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , FN-kappa B/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos/toxicidad , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Mutagénesis Sitio-Dirigida , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteína bcl-X/metabolismo
10.
Drug Des Devel Ther ; 9: 4559-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26309401

RESUMEN

Valproic acid (VPA) is mainly metabolized via glucuronide, which is hydrolyzed by ß-glucuronidase and undergoes enterohepatic circulation. Amoxicillin/clavulanic acid (AMC) administration leads to decreased levels of ß-glucuronidase-producing bacteria, suggesting that these antibiotics could interrupt enterohepatic circulation and thereby alter the pharmacokinetics of VPA. This study aimed to evaluate the effects of AMC on the pharmacokinetics of VPA. This was an open-label, two-treatment, one-sequence study in 16 healthy volunteers. Two treatments were evaluated; treatment VPA, in which a single dose of VPA 500 mg was administered, and treatment AMC + VPA, in which multiple doses of AMC 500/125 mg were administered three times daily for 7 days and then a single dose of VPA was administered. Blood samples were collected up to 48 hours. Pharmacokinetic parameters were calculated using noncompartmental methods. Fifteen subjects completed the study. Systemic exposures and peak concentrations of VPA were slightly lower with treatment AMC + VPA than with treatment VPA (AUClast, 851.0 h·mg/L vs 889.6 h·mg/L; C max, 52.1 mg/L vs 53.0 mg/L). There were no significant between-treatment effects on pharmacokinetics (95% confidence interval [CI]) of AUClast and C max (95.7 [85.9-106.5] and 98.3 [91.6-105.6], respectively). Multiple doses of AMC had no significant effects on the pharmacokinetics of VPA; thus, no dose adjustment is necessary.


Asunto(s)
Combinación Amoxicilina-Clavulanato de Potasio/administración & dosificación , Antibacterianos/administración & dosificación , Anticonvulsivantes/farmacocinética , Ácido Valproico/farmacocinética , Adulto , Combinación Amoxicilina-Clavulanato de Potasio/efectos adversos , Antibacterianos/efectos adversos , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/sangre , Área Bajo la Curva , Biotransformación , Esquema de Medicación , Interacciones Farmacológicas , Circulación Enterohepática , Semivida , Voluntarios Sanos , Humanos , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Modelos Biológicos , República de Corea , Ácido Valproico/administración & dosificación , Ácido Valproico/efectos adversos , Ácido Valproico/análogos & derivados , Ácido Valproico/sangre , Adulto Joven
11.
Pharmacology ; 96(1-2): 68-75, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26089136

RESUMEN

AIMS: To compare the pharmacokinetics, pharmacodynamics, and safety of sarpogrelate between controlled-release (CR) and immediate-release (IR) formulations after multiple-dose administration. METHODS: This study was a randomized, open-label, 2-period, 2-treatment, crossover study in healthy subjects. All subjects received CR sarpogrelate 300 mg once daily and IR sarpogrelate 100 mg three times daily by random order each for 3 days with a 7-day washout period. Serial blood sampling was performed over 24 h. Pharmacokinetic parameters were determined by noncompartmental methods. Platelet aggregation to collagen, measured by light transmission aggregometry, was reported as maximal platelet aggregation. RESULTS: Thirty-two subjects completed the study. CR sarpogrelate increased rapidly, reaching Cmax in 1.25 h (vs. 1.00 h in IR sarpogrelate) and declined with a t1/2 of 3.59 h (vs. 1.12 h in IR sarpogrelate). The 90% CIs for the geometric mean ratio of AUCτ and Cmax,ss between IR and CR formulations were 1.18 to 1.40 and 0.99 to 1.29, respectively. The degree of inhibition of platelet aggregation was similar between two formulations. CONCLUSIONS: CR sarpogrelate showed slightly higher systemic exposure and similar peak concentration compared with IR sarpogrelate. The profiles of pharmacodynamics and safety were comparable between two formulations.


Asunto(s)
Preparaciones de Acción Retardada , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/farmacocinética , Succinatos/administración & dosificación , Succinatos/farmacocinética , Adulto , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Femenino , Humanos , Masculino , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Succinatos/efectos adversos , Succinatos/farmacología , Adulto Joven
12.
Drug Des Devel Ther ; 9: 2811-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26082611

RESUMEN

A fixed-dose combination of amlodipine and olmesartan is used to treat high blood pressure in patients whose hypertension is not sufficiently controlled with either drug alone. The objective of this study was to evaluate the bioequivalence of two fixed-dose combinations, ie, amlodipine orotate/olmesartan medoxomil 10/40 mg and amlodipine besylate/olmesartan medoxomil 10/40 mg, in healthy subjects. A randomized, open-label, single-dose, two-sequence, two-period, crossover study was conducted in 30 healthy adult volunteers. Blood samples were collected for up to 72 hours post-dose in each period. Safety data included the results of physical examinations, clinical laboratory tests, vital signs, an electrocardiogram, and adverse events. For both amlodipine and olmesartan, the 90% confidence intervals for the geometric mean ratios of AUClast and time to peak plasma concentration fell within the bioequivalence acceptance criteria. The two fixed-dose combinations showed similar safety profiles. Amlodipine orotate/olmesartan medoxomil 10/40 mg was bioequivalent to amlodipine besylate/olmesartan medoxomil 10/40 mg.


Asunto(s)
Combinación Besilato de Amlodipino y Olmesartán Medoxomilo/farmacocinética , Antihipertensivos/farmacocinética , Adulto , Combinación Besilato de Amlodipino y Olmesartán Medoxomilo/administración & dosificación , Combinación Besilato de Amlodipino y Olmesartán Medoxomilo/química , Antihipertensivos/administración & dosificación , Antihipertensivos/química , Área Bajo la Curva , Estudios Cruzados , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sales (Química) , Equivalencia Terapéutica , Adulto Joven
13.
Drug Des Devel Ther ; 9: 737-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25767371

RESUMEN

AIMS: Lobeglitazone has been developed for the treatment of type 2 diabetes mellitus. This study was conducted to evaluate potential drug-drug interactions between lobeglitazone and warfarin, an anticoagulant with a narrow therapeutic index. METHODS: In this open-label, three-treatment, crossover study, 24 healthy male subjects were administered lobeglitazone (0.5 mg) for 1-12 days with warfarin (25 mg) on day 5 in one period. After a washout interval, subjects were administered warfarin (25 mg) alone in the other period. Pharmacokinetics of R- and S-warfarin and lobeglitazone, as well as pharmacodynamics of warfarin, as measured by international normalized ratio (INR) and factor VII activity, were assessed. RESULTS: The geometric mean ratios (GMRs) and 90% confidence intervals (CIs) for area under the curve from time zero to the time of the last quantifiable concentration (AUClast) for warfarin + lobeglitazone: warfarin alone were 1.0076 (90% CI: 0.9771, 1.0391) for R-warfarin and 0.9880 (90% CI: 0.9537, 1.0235) for S-warfarin. The maximum observed plasma concentration (C max) values were 1.0167 (90% CI: 0.9507, 1.0872) for R-warfarin and 1.0028 (90% CI: 0.9518, 1.0992) for S-warfarin, both of which were contained in the interval 0.80-1.25. Lobeglitazone had no effect on the area under the effect-time curve from time 0 to 168 hours (AUEC) of INR and factor VII activity, as demonstrated by the GMRs of 1.0091 (90% CI: 0.9872, 1.0314) and 0.9355 (90% CI: 0.9028, 0.9695), respectively. In addition, the pharmacokinetics of lobeglitazone was also unaffected by warfarin. CONCLUSION: Concomitant administration of lobeglitazone and warfarin was well tolerated. Lobeglitazone had no meaningful effect on the pharmacokinetics or pharmacodynamics of warfarin. These findings indicate that lobeglitazone and warfarin can be coadministered without dosage adjustments for either drug.


Asunto(s)
Pirimidinas/farmacología , Tiazolidinedionas/farmacología , Warfarina/farmacocinética , Adulto , Estudios Cruzados , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad , Pirimidinas/administración & dosificación , Distribución Aleatoria , Tiazolidinedionas/administración & dosificación , Warfarina/administración & dosificación , Warfarina/sangre , Adulto Joven
14.
Drug Des Devel Ther ; 9: 745-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25767372

RESUMEN

PURPOSE: Valsartan, an angiotensin-receptor blocker, and rosuvastatin, a competitive inhibitor of the 3-hydroxy-3-methylglutaryl coenzyme A reductase, are frequently coadministered to treat patients with hypertension and dyslipidemia. The study reported here sought to evaluate the pharmacokinetic and pharmacodynamic interactions between rosuvastatin and valsartan in healthy Korean subjects. SUBJECTS AND METHODS: Thirty healthy male Korean subjects were administered with rosuvastatin (20 mg/day), valsartan (160 mg/day), and both drugs concomitantly for 4 days in a randomized, open-label, multiple-dose, three-treatment, three-period crossover study. Plasma concentrations of rosuvastatin, N-desmethyl rosuvastatin, and valsartan were determined using validated high-performance liquid chromatography with tandem mass spectrometry. Lipid profiles and vital signs (systolic and diastolic blood pressure and pulse rate) were measured for the pharmacodynamic assessment. RESULTS: For rosuvastatin, the geometric mean ratios (90% confidence intervals [CIs]) of coadministration to mono-administration were 0.8809 (0.7873-0.9857) for maximum plasma concentration at steady state and 0.9151 (0.8632-0.9701) for area under the concentration-time curve (AUC) over a dosing interval at steady state. For valsartan, the geometric mean ratios (90% CIs) of those were 0.9300 (0.7946-1.0884) and 1.0072 (0.8893-1.1406), respectively. There were no significant differences in the metabolic ratio of N-desmethyl rosuvastatin AUC to rosuvastatin AUC between coadministration and rosuvastatin alone. No interaction was found in terms of systolic or diastolic blood pressure or lipid profiles. Combined treatment with valsartan and rosuvastatin was generally well tolerated without serious adverse events. CONCLUSION: The pharmacokinetic profiles of rosuvastatin and valsartan in combination were comparable with those of rosuvastatin and valsartan administered individually, suggesting that their individual pharmacokinetics were not affected by their coadministration. No dose adjustment was required and the results are supportive of a study in a larger patient population.


Asunto(s)
Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/farmacocinética , Valsartán/administración & dosificación , Valsartán/farmacocinética , Adulto , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Distribución Aleatoria , Rosuvastatina Cálcica/sangre , Valsartán/sangre , Adulto Joven
15.
Int J Dermatol ; 54(12): 1382-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25556325

RESUMEN

BACKGROUND: The Atopic Dermatitis Antecubital Severity (ADAS) score is a new objective scale for the assessment of the severity of atopic dermatitis (AD). It is calculated by multiplying the intensity of inflammatory signs by the size of an antecubital eczema lesion. AIM: To test the validity, reliability, and sensitivity to changes of the ADAS score compared with those of the Eczema Area and Severity Index (EASI) score. METHODS: Forty patients with AD were enrolled and treated with a moisturizer. At baseline, and in weeks 1 and 2, two independent evaluators measured the ADAS score, the EASI score, and the investigator's global assessment score rated on a six-point scale. RESULTS: The ADAS score showed a higher validity than the EASI score. The superiority of the ADAS to the EASI was prominent in mild AD. Inter-evaluator reliability was excellent in both the ADAS score and the EASI. The sensitivity to changes was higher in the ADAS score than in the EASI score. CONCLUSIONS: The ADAS score may be used as a simple scoring system with good validity, reliability, and sensitivity to changes, especially in patients with mild-to-moderate AD.


Asunto(s)
Dermatitis Atópica/patología , Índice de Severidad de la Enfermedad , Adolescente , Adulto , Niño , Preescolar , Dermatitis Atópica/tratamiento farmacológico , Femenino , Antebrazo , Humanos , Masculino , Variaciones Dependientes del Observador , Reproducibilidad de los Resultados , Crema para la Piel/uso terapéutico , Adulto Joven
16.
J Cosmet Dermatol ; 12(4): 287-95, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24305427

RESUMEN

BACKGROUND: Epidermal and fibroblast growth factor (EGF and FGF1) proteins play an important role in the regeneration and proliferation of skin cells. EGF and FGF1 have considerable potential as possible therapeutic or cosmetic agents for the treatment of skin damage including wrinkles. OBJECTIVES: Using protein transduction domains (PTD), we investigated whether PTD-EGF and FGF1 transduced into skin cells and tissue. Transduced proteins showed protective effects in a UV-induced skin damage model as well as against skin wrinkles. METHODS: Transduced PTD-EGF and FGF1 proteins were detected by immunofluorescence and immunohistochemistry. The effects of PTD-EGF and FGF1 were examined by WST assay, Western blotting, immunohistochemistry, and skin wrinkle parameters. RESULTS: The PTD-EGF and FGF1 increased cell proliferation and collagen type 1 alpha 1 protein accumulation in skin tissue. Also, PTD-EGF and FGF1 inhibited UV-induced skin damage. Furthermore, topical application of PTD-EGF and FGF1 contained ampoules which were considered to improve the wrinkle parameters of human skin. CONCLUSION: These results show that PTD-EGF and FGF1 can be a potential therapeutic or cosmetic agent for skin damaged and injury including wrinkles and aging.


Asunto(s)
Fármacos Dermatológicos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Factor 1 de Crecimiento de Fibroblastos/farmacología , Envejecimiento de la Piel/efectos de la radiación , Piel/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
17.
FEBS Lett ; 582(16): 2371-6, 2008 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-18539148

RESUMEN

Nuclear factor kappaB (NF-kappaB) is associated with the transcriptional activation of genes encoding chemokines, adhesion molecules, cytokines, and anti-apoptotic proteins, which are key components in immune responses and viral infection. Many viruses modulate NF-kappaB through numerous viral gene products to allow productive infections and immune escape. Here we report that herpes simplex virus-1 infected cell protein 27 (HSV-1 ICP27), an immediate early protein of HSV-1, represses NF-kappaB activity through binding to inhibitor of kappaB (IkappaBalpha), blocking phosphorylation and ubiquitination of IkappaBalpha, and stabilizing IkappaBalpha. These data may explain how NF-kappaB activity is regulated by ICP27 to escape immune responses during the very early period of HSV-1 infection.


Asunto(s)
Proteínas I-kappa B/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , FN-kappa B/antagonistas & inhibidores , Animales , Línea Celular , Chlorocebus aethiops , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Fosforilación , Ubiquitinación , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...