Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cells ; 12(24)2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38132113

RESUMEN

Atopic dermatitis (AD) is the most common chronic inflammatory skin disease and presents a major public health problem worldwide. It is characterized by a recurrent and/or chronic course of inflammatory skin lesions with intense pruritus. Its pathophysiologic features include barrier dysfunction, aberrant immune cell infiltration, and alterations in the microbiome that are associated with genetic and environmental factors. There is a complex crosstalk between these components, which is primarily mediated by cytokines. Epidermal barrier dysfunction is the hallmark of AD and is caused by the disruption of proteins and lipids responsible for establishing the skin barrier. To better define the role of cytokines in stratum corneum lipid abnormalities related to AD, we conducted a systematic review of biomedical literature in PubMed from its inception to 5 September 2023. Consistent with the dominant TH2 skewness seen in AD, type 2 cytokines were featured prominently as possessing a central role in epidermal lipid alterations in AD skin. The cytokines associated with TH1 and TH17 were also identified to affect barrier lipids. Considering the broad cytokine dysregulation observed in AD pathophysiology, understanding the role of each of these in lipid abnormalities and barrier dysfunction will help in developing therapeutics to best achieve barrier homeostasis in AD patients.


Asunto(s)
Dermatitis Atópica , Humanos , Dermatitis Atópica/patología , Citocinas/metabolismo , Epidermis/metabolismo , Piel/patología , Lípidos
2.
Antioxidants (Basel) ; 11(6)2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35740103

RESUMEN

Constitutive pigmentation determines the response to sun exposure and the risk for melanoma, an oxidative stress-driven tumor. Using primary cultures of human melanocytes, we compared the effects of constitutive pigmentation on their antioxidant response to solar UV. The quantitation of eumelanin and pheomelanin showed that the eumelanin content and eumelanin to pheomelanin ratio correlated inversely with the basal levels of reactive oxygen species (ROS). Irradiation with 7 J/cm2 solar UV increased ROS generation without compromising melanocyte viability. Among the antioxidant enzymes tested, the basal levels of heme oxygenase-1 (HO-1) and the glutamate cysteine ligase catalytic subunit and modifier subunit (GCLC and GCLM) correlated directly with the eumelanin and total melanin contents. The levels of HO-1 and GCLM decreased at 6 h but increased at 24 h post-solar UV. Consistent with the GCLC and GCLM levels, the basal glutathione (GSH) content was significantly lower in light than in dark melanocytes. The expression of HMOX1, GCLC, GCLM, and CAT did not correlate with the melanin content and was reduced 3 h after solar UV irradiation, particularly in lightly pigmented melanocytes. Solar UV increased p53 and lipid peroxidation, which correlated inversely with the eumelanin and total melanin contents. These intrinsic differences between light and dark melanocytes should determine their antioxidant response and melanoma risk.

3.
J Antimicrob Chemother ; 77(6): 1725-1732, 2022 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-35383374

RESUMEN

BACKGROUND: Cefotaxime is frequently used in critically ill children, however pharmacokinetic (PK) studies to support adequate dosing in this patient population are limited. OBJECTIVES: To characterize cefotaxime PK in critically ill children and evaluate exposures achieved by current and alternative dosing regimens. METHODS: Children (0-18 years) admitted to the paediatric ICU, receiving intravenous cefotaxime (100-150 mg/kg/day, interval 6-8 h) were included (Clinicaltrials.gov NCT03248349). Total plasma cefotaxime concentrations were measured on multiple study days. Population-PK analysis was performed using nonlinear mixed effects modelling (NONMEM™). Dose evaluations were performed using typical patients across the paediatric age range and target attainment was determined for MICs of 0.5, 2 and 4 mg/L. RESULTS: 479 cefotaxime plasma concentrations from 52 children (median age 1.6, range 0.03-17.7 years) were used to describe cefotaxime PK. We describe a two-compartment structural model with interindividual variability, including bodyweight as covariate for volume of distribution and clearance. Model predicted exposure for 150 mg/kg/day (current dose) showed trough concentrations <0.5 mg/L in patients >4 years of age. The maximum cefotaxime doses (200 mg/kg/day, interval 6 h) proved adequate for MICs ≤0.5 mg/L across the whole age range. Similar daily doses with increased frequency (interval 4 h) covered MICs up to 2 mg/L, while a loading dose followed by continuous infusion regimens are needed to adequately treat MICs of 4 mg/L. CONCLUSIONS: Higher cefotaxime doses are required for adequate exposure for most pathogens in critically ill children. A higher dose frequency or continuous infusion is advisable to improve target attainment for intermediately susceptible pathogens.


Asunto(s)
Cefotaxima , Enfermedad Crítica , Administración Intravenosa , Adolescente , Antibacterianos/uso terapéutico , Niño , Preescolar , Enfermedad Crítica/terapia , Humanos , Lactante , Recién Nacido , Pruebas de Sensibilidad Microbiana
4.
Pharm Res ; 38(10): 1711-1720, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34664207

RESUMEN

AIM: In critically ill mechanically ventilated children, midazolam is used first line for sedation, however its exact sedative effects have been difficult to quantify. In this analysis, we use parametric time-to-event (PTTE) analysis to quantify the effects of midazolam in critically ill children. METHODS: In the PTTE analysis, data was analyzed from a published study in mechanically ventilated children in which blinded midazolam or placebo infusions were administered during a sedation interruption phase until, based on COMFORT-B and NISS scores, patients became undersedated and unblinded midazolam was restarted. Using NONMEM® v.7.4.3., restart of unblinded midazolam was analysed as event. Patients in the trial were divided into internal and external validation cohorts prior to analysis. RESULTS: Data contained 138 events from 79 individuals (37 blinded midazolam; 42 blinded placebo). In the PTTE model, the baseline hazard was best described by a constant function. Midazolam reduced the hazard for restart of unblinded midazolam due to undersedation by 51%. In the blinded midazolam group, time to midazolam restart was 26 h versus 58 h in patients with low versus high disease severity upon admission (PRISM II < 10 versus > 21), respectively. For blinded placebo, these times were 14 h and 33 h, respectively. The model performed well in an external validation with 42 individuals. CONCLUSION: The PTTE analysis effectively quantified the effect of midazolam in prolonging sedation and also the influence of disease severity on sedation in mechanically ventilated critically ill children, and provides a valuable tool to quantify the effect of sedatives. Clinical trial number and registry URL: Netherlands Trial Register, Trial NL1913 (NTR2030), date registered 28 September 2009 https://www.trialregister.nl/trial/1913 .


Asunto(s)
Hipnóticos y Sedantes/metabolismo , Midazolam/metabolismo , Adolescente , Niño , Preescolar , Enfermedad Crítica , Femenino , Humanos , Hipnóticos y Sedantes/farmacocinética , Lactante , Recién Nacido , Infusiones Intravenosas , Masculino , Midazolam/farmacocinética , Modelos Estadísticos , Países Bajos , Respiración Artificial , Índice de Severidad de la Enfermedad , Factores de Tiempo
5.
Clin Pharmacokinet ; 60(10): 1361-1372, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34036552

RESUMEN

BACKGROUND AND OBJECTIVE: Ceftriaxone is a cornerstone antibiotic for critically ill children with severe infections. Despite its widespread use, information on the pharmacokinetics of ceftriaxone is lacking in this population. We aimed to determine ceftriaxone pharmacokinetics in critically ill children and to propose ceftriaxone dosing guidelines resulting in adequate target attainment using population pharmacokinetic modeling and simulation. METHODS: Critically ill children (aged 0-18 years) treated with intravenous ceftriaxone (100 mg/kg once daily, infused in 30 minutes) and a central or arterial line in place were eligible. Opportunistic blood sampling for total and unbound ceftriaxone concentrations was used. Population pharmacokinetic analysis was performed using non-linear mixed-effects modeling on NONMEM™ Version 7.4.3. Simulations were performed to select optimal doses using probability of target attainment for two pharmacokinetic targets of the minimum inhibitory concentration (MIC) reflecting the susceptibility of pathogens (f T > MIC 100% and fT > 4 × MIC 100%). RESULTS: Two hundred and five samples for total and 43 time-matched samples for unbound plasma ceftriaxone concentrations were collected from 45 patients, median age 2.5 (range 0.1-16.7) years. A two-compartment model with bodyweight as the co-variate for volume of distribution and clearance, and creatinine-based estimated glomerular filtration rate as an additional covariate for clearance, best described ceftriaxone pharmacokinetics. For a typical patient (2.5 years, 14 kg) with an estimated glomerular filtration rate of 80 mL/min/1.73 m2, the current 100-mg/kg once-daily dose results in a probability of target attainment of 96.8% and 60.8% for a MIC of 0.5 mg/L and 4 × MIC (2 mg/L), respectively, when using fT > MIC 100% as a target. For a 50-mg/kg twice-daily regimen, the probability of target attainment was 99.9% and 93.4%, respectively. CONCLUSIONS: The current dosing regimen of ceftriaxone provides adequate exposure for susceptible pathogens in most critically ill children. In patients with an estimated glomerular filtration rate of > 80 mL/min/1.73 m2 or in areas with a high prevalence of less-susceptible pathogens (MIC ≥ 0.5 mg/L), a twice-daily dosing regimen of 50 mg/kg can be considered to improve target attainment. CLINICAL TRIAL REGISTRATION: POPSICLE study (ClinicalTrials.gov, NCT03248349, registered 14 August, 2017), PERFORM study (ClinicalTrials.gov, NCT03502993, registered 19 April, 2018).


Asunto(s)
Ceftriaxona , Enfermedad Crítica , Adolescente , Antibacterianos/uso terapéutico , Ceftriaxona/farmacocinética , Niño , Preescolar , Creatinina , Humanos , Lactante , Pruebas de Sensibilidad Microbiana , Estudios Prospectivos
6.
Pigment Cell Melanoma Res ; 34(4): 762-776, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33973367

RESUMEN

Human epidermal melanocytes play a central role in sensing the environment and protecting the skin from the drastic effects of solar ultraviolet radiation and other environmental toxins or inflammatory agents. Melanocytes survive in the epidermis for decades, which subjects them to chronic environmental insults. Melanocytes have a poor self-renewal capacity; therefore, it is critical to ensure their survival with genomic stability. The function and survival of melanocytes is regulated by an elaborate network of paracrine factors synthesized mainly by epidermal keratinocytes and dermal fibroblasts. A symbiotic relationship exists between epidermal melanocytes and keratinocytes on the one hand, and between melanocytes and dermal fibroblasts on the other hand. Melanocytes protect epidermal keratinocytes and dermal fibroblasts from the damaging effects of solar radiation, and the latter cells synthesize biochemical mediators that maintain the homeostasis, and regulate the stress response of melanocytes. Disruption of the paracrine network results in pigmentary disorders, due to abnormal regulation of melanin synthesis, and compromise of melanocyte survival or genomic stability. This review provides an update of the current knowledge of keratinocyte- and fibroblast-derived paracrine factors and their contribution to melanocyte physiology, and how their abnormal production is involved in the pathogenesis of common pigmentary disorders.


Asunto(s)
Fibroblastos/metabolismo , Homeostasis , Queratinocitos/metabolismo , Melanocitos/metabolismo , Trastornos de la Pigmentación/patología , Rayos Ultravioleta , Animales , Fibroblastos/efectos de la radiación , Homeostasis/efectos de la radiación , Humanos , Queratinocitos/efectos de la radiación , Melanocitos/efectos de la radiación
7.
CPT Pharmacometrics Syst Pharmacol ; 10(4): 350-361, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33792207

RESUMEN

Pharmacometric modeling can capture tumor growth inhibition (TGI) dynamics and variability. These approaches do not usually consider covariates in high-dimensional settings, whereas high-dimensional molecular profiling technologies ("omics") are being increasingly considered for prediction of anticancer drug treatment response. Machine learning (ML) approaches have been applied to identify high-dimensional omics predictors for treatment outcome. Here, we aimed to combine TGI modeling and ML approaches for two distinct aims: omics-based prediction of tumor growth profiles and identification of pathways associated with treatment response and resistance. We propose a two-step approach combining ML using least absolute shrinkage and selection operator (LASSO) regression with pharmacometric modeling. We demonstrate our workflow using a previously published dataset consisting of 4706 tumor growth profiles of patient-derived xenograft (PDX) models treated with a variety of mono- and combination regimens. Pharmacometric TGI models were fit to the tumor growth profiles. The obtained empirical Bayes estimates-derived TGI parameter values were regressed using the LASSO on high-dimensional genomic copy number variation data, which contained over 20,000 variables. The predictive model was able to decrease median prediction error by 4% as compared with a model without any genomic information. A total of 74 pathways were identified as related to treatment response or resistance development by LASSO, of which part was verified by literature. In conclusion, we demonstrate how the combined use of ML and pharmacometric modeling can be used to gain pharmacological understanding in genomic factors driving variation in treatment response.


Asunto(s)
Antineoplásicos/metabolismo , Neoplasias/tratamiento farmacológico , Farmacogenética/instrumentación , Carga Tumoral/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Teorema de Bayes , Variación Biológica Poblacional/genética , Variaciones en el Número de Copia de ADN/genética , Desarrollo de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Genómica , Humanos , Aprendizaje Automático , Ratones , Modelos Animales , Neoplasias/patología , Valor Predictivo de las Pruebas , Resultado del Tratamiento , Carga Tumoral/genética
8.
J Invest Dermatol ; 141(7): 1819-1829, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33609553

RESUMEN

Activation of the human melanocortin 1 receptor (hMC1R) expressed on melanocytes by α-melanocortin plays a central role in regulating human pigmentation and reducing the genotoxicity of UV by activating DNA repair and antioxidant defenses. For the development of a hMC1R-targeted photoprotection strategy, we designed tetra- and tripeptide agonists with modifications that provide the necessary lipophilicity and hMC1R selectivity to be effective drugs. These peptides proved to be superior to most of the existing analogs of the physiological tridecapeptide α-melanocortin because of their small size and high hMC1R selectivity. Testing on primary cultures of human melanocytes showed that these peptides are highly potent with prolonged stimulation of melanogenesis, enhanced repair of UV-induced DNA photoproducts, and reduced apoptosis. One of the tripeptides, designated as LK-514 (5), with a molecular weight of 660 Da, has unprecedented (>100,000) hMC1R selectivity when compared with the other melanocortin receptors hMC3R, hMC4R, and hMC5R, and increases pigmentation (sunless tanning) in a cultured, three-dimensional skin model. These new analogs should be efficacious in preventing skin cancer, including melanoma, and treatment of skin disorders, such as vitiligo and polymorphic light eruptions.


Asunto(s)
Daño del ADN/efectos de los fármacos , Fármacos Dermatológicos/farmacología , Receptor de Melanocortina Tipo 1/agonistas , Pigmentación de la Piel/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Células Cultivadas , Daño del ADN/efectos de la radiación , Fármacos Dermatológicos/uso terapéutico , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanocitos/efectos de la radiación , Melanoma/etiología , Melanoma/prevención & control , Trastornos por Fotosensibilidad/tratamiento farmacológico , Trastornos por Fotosensibilidad/genética , Cultivo Primario de Células , Receptor de Melanocortina Tipo 1/metabolismo , Piel/efectos de los fármacos , Piel/efectos de la radiación , Enfermedades Cutáneas Genéticas/tratamiento farmacológico , Enfermedades Cutáneas Genéticas/genética , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/prevención & control , Pigmentación de la Piel/efectos de la radiación , Vitíligo/tratamiento farmacológico , Vitíligo/genética , alfa-MSH/metabolismo
9.
Pigment Cell Melanoma Res ; 33(6): 778-787, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32198977

RESUMEN

Vitiligo is the most common acquired pigmentary disorder, which afflicts 0.5%-1% of the world population, and is characterized by depigmented skin patches resulting from melanocyte loss. Vitiligo has a complex etiology and varies in its manifestations, progression, and response to treatment. It presents as an autoimmune disease, evidenced by circulating melanocyte-specific antibodies, and association with other autoimmune diseases. However, autoimmunity may be secondary to the high oxidative stress in vitiligo skin and to intrinsic defects in melanocytes and their microenvironment, which contribute to aberrant stress response, neo-antigenicity, and susceptibility of melanocytes to immune attack and apoptosis. There is also a genetic predisposition to vitiligo, which sensitizes melanocytes to environmental agents, such as phenolic compounds. Currently, there are different treatment modalities for re-pigmenting vitiligo skin. However, when repigmentation is achieved, the major challenge is maintaining the pigmentation, which is lost in 40% of cases. In this review, we present an overview of the clinical aspects of vitiligo, its pathophysiology, the intrinsic defects in melanocytes and their microenvironment, and treatment strategies. Based on lessons from the biology of human melanocytes, we present our perspective of how repigmentation of vitiligo skin can be achieved and sustained.


Asunto(s)
Vitíligo/fisiopatología , Vitíligo/terapia , Autoinmunidad , Microambiente Celular , Humanos , Melanocitos/patología , Estrés Oxidativo , Vitíligo/inmunología
10.
Clin Pharmacokinet ; 58(8): 1029-1043, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30868471

RESUMEN

Targeted therapies, based on identification of common oncogenic mutations such as BRAF V600E/K and monoclonal antibody immunotherapies, have transformed the treatment of melanoma. Dual mitogen-activated protein kinase (MAPK) pathway inhibition of BRAF V600E/K and MEK 1/2 kinases with BRAF-MEK inhibitors using dabrafenib-trametinib, vemurafenib-cobimetinib and encorafenib-binimetinib is now the standard of care for BRAF V600E/K tumours. Monoclonal antibodies, such as pembrolizumab and nivolumab, against programmed cell death protein (PD-1) on T cells, as well as ipilimumab against cytotoxic T lymphocyte antigen-4 (CTLA-4), enable restoration of suppressed T-cell antitumour response, and have also shown improved clinical benefit compared with traditional chemotherapy. Exploration of different combination therapies, sequence of treatment, and dosing strategies is ongoing, and the understanding of the pharmacokinetics (PK) and pharmacodynamics (PD) of these new agents is fundamental in devising the optimal regimen. Preclinical and clinical studies, as well as population PK modelling, provide essential data in terms of PK parameters, metabolism, interpatient variability, drug interactions and PD effects at the target. This review gathers the current evidence and understanding of the clinical PK and PD of drugs used in the modern treatment of melanoma, and the factors determining drug disposition, exposure and clinical response, and also highlighting areas of further research.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Carcinogénesis/efectos de los fármacos , Melanoma/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Inhibidores de Proteínas Quinasas/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores/metabolismo , Carcinogénesis/genética , Terapia Combinada/métodos , Femenino , Humanos , Masculino , Melanoma/genética , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mutación , Farmacogenética/métodos , Inhibidores de Proteínas Quinasas/uso terapéutico , Microambiente Tumoral/genética
11.
Biomed Chromatogr ; 31(7)2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27925250

RESUMEN

Dimethylacetamide (DMA) is a solvent used in the preparation of intravenous busulfan, an alkylating agent used in blood or marrow transplantation. DMA may contribute to hepatic toxicity, so it is important to monitor its clearance. The aim of this study was to develop an HPLC-UV assay for measurement of DMA in human plasma. After precipitation of plasma proteins with acetonitrile followed by dilution (1:4) with water, the extract was injected onto the HPLC and detected at 195 nm. Separation was performed using a Cogent-HPS 5 µm C18 column (250 × 4.6 mm) preceded by a Brownlee 7 µm RP18 , pre-column (1.5 cm × 3.2 mm). The mobile phase was 25 mm sodium phosphate buffer (pH 3), containing 2.5% (v/v) acetonitrile and 0.0005% (v/v) sodium-octyl-sulfonate. Using a flow rate of 1 mL/min, the retention times of DMA and the internal standard (IS), 2-chloroacetamide, were 9.5 and 3.5 min, respectively. Peak area ratio (DMA:IS) was a linear function of concentration from 1 to 1000 µg/mL. There was excellent intraday precision (<5% for 5-700 µg/mL DMA), accuracy (<3% deviation from the true concentration) and recovery (74-98%). The limits of detection and quantification were 1 and 5 µg/mL, respectively. In eight children who received intravenous busulfan, DMA concentrations ranged from 110 to 438 µg/mL.


Asunto(s)
Acetamidas/sangre , Alquilantes/sangre , Busulfano/administración & dosificación , Cromatografía Líquida de Alta Presión/métodos , Espectrofotometría Ultravioleta/métodos , Humanos , Infusiones Intravenosas , Estándares de Referencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...