Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
CPT Pharmacometrics Syst Pharmacol ; 12(7): 916-928, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37002678

RESUMEN

Oncology treatments require continuous individual adjustment based on the measurement of multiple clinical parameters. Prediction tools exploiting the patterns present in the clinical data could be used to assist decision making and ease the burden associated to the interpretation of all these parameters. The goal of this study was to predict the evolution of patients with pancreatic cancer at their next visit using information routinely recorded in health records, providing a decision-support system for clinicians. We selected hematological variables as the visit's clinical outcomes, under the assumption that they can be predictive of the evolution of the patient. Multivariate models based on regression trees were generated to predict next-visit values for each of the clinical outcomes selected, based on the longitudinal clinical data as well as on molecular data sets streaming from in silico simulations of individual patient status at each visit. The models predict, with a mean prediction score (balanced accuracy) of 0.79, the evolution trends of eosinophils, leukocytes, monocytes, and platelets. Time span between visits and neutropenia were among the most common factors contributing to the predicted evolution. The inclusion of molecular variables from the systems-biology in silico simulations provided a molecular background for the observed variations in the selected outcome variables, mostly in relation to the regulation of hematopoiesis. In spite of its limitations, this study serves as a proof of concept for the application of next-visit prediction tools in real-world settings, even when available data sets are small.


Asunto(s)
Inteligencia Artificial , Neoplasias Pancreáticas , Humanos , Biología de Sistemas , Simulación por Computador , Neoplasias Pancreáticas/genética
2.
Gigascience ; 112022 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-36155782

RESUMEN

BACKGROUND: In cancer research, high-throughput screening technologies produce large amounts of multiomics data from different populations and cell types. However, analysis of such data encounters difficulties due to disease heterogeneity, further exacerbated by human biological complexity and genomic variability. The specific profile of cancer as a disease (or, more realistically, a set of diseases) urges the development of approaches that maximize the effect while minimizing the dosage of drugs. Now is the time to redefine the approach to drug discovery, bringing an artificial intelligence (AI)-powered informational view that integrates the relevant scientific fields and explores new territories. RESULTS: Here, we show SYNPRED, an interdisciplinary approach that leverages specifically designed ensembles of AI algorithms, as well as links omics and biophysical traits to predict anticancer drug synergy. It uses 5 reference models (Bliss, Highest Single Agent, Loewe, Zero Interaction Potency, and Combination Sensitivity Score), which, coupled with AI algorithms, allowed us to attain the ones with the best predictive performance and pinpoint the most appropriate reference model for synergy prediction, often overlooked in similar studies. By using an independent test set, SYNPRED exhibits state-of-the-art performance metrics either in the classification (accuracy, 0.85; precision, 0.91; recall, 0.90; area under the receiver operating characteristic, 0.80; and F1-score, 0.91) or in the regression models, mainly when using the Combination Sensitivity Score synergy reference model (root mean square error, 11.07; mean squared error, 122.61; Pearson, 0.86; mean absolute error, 7.43; Spearman, 0.87). Moreover, data interpretability was achieved by deploying the most current and robust feature importance approaches. A simple web-based application was constructed, allowing easy access by nonexpert researchers. CONCLUSIONS: The performance of SYNPRED rivals that of the existing methods that tackle the same problem, yielding unbiased results trained with one of the most comprehensive datasets available (NCI ALMANAC). The leveraging of different reference models allowed deeper insights into which of them can be more appropriately used for synergy prediction. The Combination Sensitivity Score clearly stood out with improved performance among the full scope of surveyed approaches and synergy reference models. Furthermore, SYNPRED takes a particular focus on data interpretability, which has been in the spotlight lately when using the most advanced AI techniques.


Asunto(s)
Antineoplásicos , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inteligencia Artificial , Benchmarking , Combinación de Medicamentos , Humanos , Aprendizaje Automático , Neoplasias/tratamiento farmacológico
3.
Methods Mol Biol ; 2190: 267-288, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32804371

RESUMEN

Targeting protein-protein interactions is a challenge and crucial task of the drug discovery process. A good starting point for rational drug design is the identification of hot spots (HS) at protein-protein interfaces, typically conserved residues that contribute most significantly to the binding. In this chapter, we depict point-by-point an in-house pipeline used for HS prediction using only sequence-based features from the well-known SpotOn dataset of soluble proteins (Moreira et al., Sci Rep 7:8007, 2017), through the implementation of a deep neural network. The presented pipeline is divided into three steps: (1) feature extraction, (2) deep learning classification, and (3) model evaluation. We present all the available resources, including code snippets, the main dataset, and the free and open-source modules/packages necessary for full replication of the protocol. The users should be able to develop an HS prediction model with accuracy, precision, recall, and AUROC of 0.96, 0.93, 0.91, and 0.86, respectively.


Asunto(s)
Mapeo de Interacción de Proteínas/métodos , Proteínas/química , Bases de Datos de Proteínas , Aprendizaje Profundo , Redes Neurales de la Computación , Unión Proteica/fisiología
4.
Database (Oxford) ; 20212021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33822911

RESUMEN

Membrane proteins (MPs) are key players in a variety of different cellular processes and constitute the target of around 60% of all Food and Drug Administration-approved drugs. Despite their importance, there is still a massive lack of relevant structural, biochemical and mechanistic information mainly due to their localization within the lipid bilayer. To help fulfil this gap, we developed the MEmbrane protein dimer Novel Structure Analyser database (MENSAdb). This interactive web application summarizes the evolutionary and physicochemical properties of dimeric MPs to expand the available knowledge on the fundamental principles underlying their formation. Currently, MENSAdb contains features of 167 unique MPs (63% homo- and 37% heterodimers) and brings insights into the conservation of residues, accessible solvent area descriptors, average B-factors, intermolecular contacts at 2.5 Å and 4.0 Å distance cut-offs, hydrophobic contacts, hydrogen bonds, salt bridges, π-π stacking, T-stacking and cation-π interactions. The regular update and organization of all these data into a unique platform will allow a broad community of researchers to collect and analyse a large number of features efficiently, thus facilitating their use in the development of prediction models associated with MPs. Database URL: http://www.moreiralab.com/resources/mensadb.


Asunto(s)
Membrana Dobles de Lípidos , Proteínas de la Membrana , Bases de Datos de Proteínas , Enlace de Hidrógeno , Proteínas de la Membrana/genética
5.
Prog Mol Biol Transl Sci ; 169: 105-149, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31952684

RESUMEN

GPCR oligomerization has emerged as a hot topic in the GPCR field in the last years. Receptors that are part of these oligomers can influence each other's function, although it is not yet entirely understood how these interactions work. The existence of such a highly complex network of interactions between GPCRs generates the possibility of alternative targets for new therapeutic approaches. However, challenges still exist in the characterization of these complexes, especially at the interface level. Different experimental approaches, such as FRET or BRET, are usually combined to study GPCR oligomer interactions. Computational methods have been applied as a useful tool for retrieving information from GPCR sequences and the few X-ray-resolved oligomeric structures that are accessible, as well as for predicting new and trustworthy GPCR oligomeric interfaces. Machine-learning (ML) approaches have recently helped with some hindrances of other methods. By joining and evaluating multiple structure-, sequence- and co-evolution-based features on the same algorithm, it is possible to dilute the issues of particular structures and residues that arise from the experimental methodology into all-encompassing algorithms capable of accurately predict GPCR-GPCR interfaces. All these methods used as a single or a combined approach provide useful information about GPCR oligomerization and its role in GPCR function and dynamics. Altogether, we present experimental, computational and machine-learning methods used to study oligomers interfaces, as well as strategies that have been used to target these dynamic complexes.


Asunto(s)
Receptores Acoplados a Proteínas G/química , Algoritmos , Sitio Alostérico , Biología Computacional , Bases de Datos de Proteínas , Evolución Molecular , Transferencia Resonante de Energía de Fluorescencia , Humanos , Aprendizaje Automático , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Solventes , Máquina de Vectores de Soporte
6.
Curr Med Chem ; 27(5): 760-794, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-30182840

RESUMEN

Paediatric Acquired ImmunoDeficiency Syndrome (AIDS) is a life-threatening and infectious disease in which the Human Immunodeficiency Virus (HIV) is mainly transmitted through Mother-To- Child Transmission (MTCT) during pregnancy, labour and delivery, or breastfeeding. This review provides an overview of the distinct therapeutic alternatives to abolish the systemic viral replication in paediatric HIV-1 infection. Numerous classes of antiretroviral agents have emerged as therapeutic tools for downregulation of different steps in the HIV replication process. These classes encompass Non- Nucleoside Analogue Reverse Transcriptase Inhibitors (NNRTIs), Nucleoside/Nucleotide Analogue Reverse Transcriptase Inhibitors (NRTIs/NtRTIs), INtegrase Inhibitors (INIs), Protease Inhibitors (PIs), and Entry Inhibitors (EIs). Co-administration of certain antiretroviral drugs with Pharmacokinetic Enhancers (PEs) may boost the effectiveness of the primary therapeutic agent. The combination of multiple antiretroviral drug regimens (Highly Active AntiRetroviral Therapy - HAART) is currently the standard therapeutic approach for HIV infection. So far, the use of HAART offers the best opportunity for prolonged and maximal viral suppression, and preservation of the immune system upon HIV infection. Still, the frequent administration of high doses of multiple drugs, their inefficient ability to reach the viral reservoirs in adequate doses, the development of drug resistance, and the lack of patient compliance compromise the complete HIV elimination. The development of nanotechnology-based drug delivery systems may enable targeted delivery of antiretroviral agents to inaccessible viral reservoir sites at therapeutic concentrations. In addition, the application of Computer-Aided Drug Design (CADD) approaches has provided valuable tools for the development of anti-HIV drug candidates with favourable pharmacodynamics and pharmacokinetic properties.


Asunto(s)
Antirretrovirales/uso terapéutico , Infecciones por VIH , Niño , Infecciones por VIH/tratamiento farmacológico , Humanos , Inhibidores de la Transcriptasa Inversa
7.
PLoS One ; 15(10): e0240149, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33006999

RESUMEN

From January 2020, COVID-19 is spreading around the world producing serious respiratory symptoms in infected patients that in some cases can be complicated by the severe acute respiratory syndrome, sepsis and septic shock, multiorgan failure, including acute kidney injury and cardiac injury. Cost and time efficient approaches to reduce the burthen of the disease are needed. To find potential COVID-19 treatments among the whole arsenal of existing drugs, we combined system biology and artificial intelligence-based approaches. The drug combination of pirfenidone and melatonin has been identified as a candidate treatment that may contribute to reduce the virus infection. Starting from different drug targets the effect of the drugs converges on human proteins with a known role in SARS-CoV-2 infection cycle. Simultaneously, GUILDify v2.0 web server has been used as an alternative method to corroborate the effect of pirfenidone and melatonin against the infection of SARS-CoV-2. We have also predicted a potential therapeutic effect of the drug combination over the respiratory associated pathology, thus tackling at the same time two important issues in COVID-19. These evidences, together with the fact that from a medical point of view both drugs are considered safe and can be combined with the current standard of care treatments for COVID-19 makes this combination very attractive for treating patients at stage II, non-severe symptomatic patients with the presence of virus and those patients who are at risk of developing severe pulmonary complications.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Reposicionamiento de Medicamentos , Melatonina/uso terapéutico , Neumonía Viral/tratamiento farmacológico , Piridonas/uso terapéutico , COVID-19 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Bases de Datos Farmacéuticas , Furina/metabolismo , Humanos , Melatonina/farmacología , Pandemias , Piridonas/farmacología , Tratamiento Farmacológico de COVID-19
8.
Methods Mol Biol ; 1958: 403-436, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30945231

RESUMEN

Membrane proteins are essential vessels for cell communication both with other cells and noncellular structures. They modulate environment responses and mediate a myriad of biological processes. Dimerization and multimerization processes have been shown to further increase the already high specificity of these processes. Due to their central role in various cell and organism functions, these multimers are often associated with health conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), and diabetes, among others.Understanding the membrane protein dimers' interface takes advantage of the specificity of the structure, for which we must pinpoint the most relevant interfacial residues, since they are extremely likely to be crucial for complex formation. Here, we describe step by step our own in silico protocol to characterize these residues, making use of known experimental structures. We detail the computational pipeline from data acquisition and pre-processing to feature extraction. A molecular dynamics simulation protocol to further study membrane dimer proteins and their interfaces is also illustrated.


Asunto(s)
Biología Computacional/métodos , Proteínas de la Membrana/química , Multimerización de Proteína , Enfermedad de Alzheimer/genética , Comunicación Celular/genética , Simulación por Computador , Diabetes Mellitus/genética , Humanos , Proteínas de la Membrana/genética , Simulación de Dinámica Molecular , Enfermedad de Parkinson/genética , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA