Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomedicines ; 12(5)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38790952

RESUMO

Autism spectrum disorder (ASD) is a heterogeneous group of neurodevelopmental disorders (NDDs) with a high unmet medical need. The diagnosis of ASD is currently based on behavior criteria, which overlooks the diversity of genetic, neurophysiological, and clinical manifestations. Failure to acknowledge such heterogeneity has hindered the development of efficient drug treatments for ASD and other NDDs. DEPI® (Databased Endophenotyping Patient Identification) is a systems biology, multi-omics, and machine learning-driven platform enabling the identification of subgroups of patients with NDDs and the development of patient-tailored treatments. In this study, we provide evidence for the validation of a first clinically and biologically defined subgroup of patients with ASD identified by DEPI, ASD Phenotype 1 (ASD-Phen1). Among 313 screened patients with idiopathic ASD, the prevalence of ASD-Phen1 was observed to be ~24% in 84 patients who qualified to be enrolled in the study. Metabolic and transcriptomic alterations differentiating patients with ASD-Phen1 were consistent with an over-activation of NF-κB and NRF2 transcription factors, as predicted by DEPI. Finally, the suitability of STP1 combination treatment to revert such observed molecular alterations in patients with ASD-Phen1 was determined. Overall, our results support the development of precision medicine-based treatments for patients diagnosed with ASD.

2.
Drug Discov Today ; 28(3): 103486, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36623795

RESUMO

Autism spectrum disorder (ASD) is a heterogenous group of neurodevelopmental disorders (NDDs) with a high unmet medical need. Currently, ASD is diagnosed according to behavior-based criteria that overlook clinical and genomic heterogeneity, thus repeatedly resulting in failed clinical trials. Here, we summarize the scientific evidence pointing to the pressing need to create a precision medicine framework for ASD and other NDDs. We discuss the role of omics and systems biology to characterize more homogeneous disease subtypes with different underlying pathophysiological mechanisms and to determine corresponding tailored treatments. Finally, we provide recent initiatives towards tackling the complexity in NDDs for precision medicine and cost-effective drug discovery.


Assuntos
Transtorno do Espectro Autista , Humanos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/terapia , Medicina de Precisão , Genômica , Genoma
3.
Proc Natl Acad Sci U S A ; 117(51): 32453-32463, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33288711

RESUMO

Pathogenic mutations in the copper transporter ATP7B have been hypothesized to affect its protein interaction landscape contributing to loss of function and, thereby, to hepatic copper toxicosis in Wilson disease. Although targeting mutant interactomes was proposed as a therapeutic strategy, druggable interactors for rescue of ATP7B mutants remain elusive. Using proteomics, we found that the frequent H1069Q substitution promotes ATP7B interaction with HSP70, thus accelerating endoplasmic reticulum (ER) degradation of the mutant protein and consequent copper accumulation in hepatic cells. This prompted us to use an HSP70 inhibitor as bait in a bioinformatics search for structurally similar Food and Drug Administration-approved drugs. Among the hits, domperidone emerged as an effective corrector that recovered trafficking and function of ATP7B-H1069Q by impairing its exposure to the HSP70 proteostatic network. Our findings suggest that HSP70-mediated degradation can be safely targeted with domperidone to rescue ER-retained ATP7B mutants and, hence, to counter the onset of Wilson disease.


Assuntos
ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Domperidona/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Degeneração Hepatolenticular/genética , Benzimidazóis/química , Benzimidazóis/farmacologia , Células Cultivadas , Cobre/metabolismo , Domperidona/química , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Células Hep G2 , Hepatócitos/metabolismo , Degeneração Hepatolenticular/tratamento farmacológico , Degeneração Hepatolenticular/metabolismo , Degeneração Hepatolenticular/patologia , Humanos , Mutação de Sentido Incorreto , Ácidos Nipecóticos/química , Ácidos Nipecóticos/farmacologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Proteômica/métodos
4.
Bioinformatics ; 34(9): 1498-1505, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29236977

RESUMO

Motivation: Drug repositioning has been proposed as an effective shortcut to drug discovery. The availability of large collections of transcriptional responses to drugs enables computational approaches to drug repositioning directly based on measured molecular effects. Results: We introduce a novel computational methodology for rational drug repositioning, which exploits the transcriptional responses following treatment with small molecule. Specifically, given a therapeutic target gene, a prioritization of potential effective drugs is obtained by assessing their impact on the transcription of genes in the pathway(s) including the target. We performed in silico validation and comparison with a state-of-art technique based on similar principles. We next performed experimental validation in two different real-case drug repositioning scenarios: (i) upregulation of the glutamate-pyruvate transaminase (GPT), which has been shown to induce reduction of oxalate levels in a mouse model of primary hyperoxaluria, and (ii) activation of the transcription factor TFEB, a master regulator of lysosomal biogenesis and autophagy, whose modulation may be beneficial in neurodegenerative disorders. Availability and implementation: A web tool for Gene2drug is freely available at http://gene2drug.tigem.it. An R package is under development and can be obtained from https://github.com/franapoli/gep2pep. Contact: dibernardo@tigem.it. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Biologia Computacional/métodos , Simulação por Computador , Reposicionamento de Medicamentos/métodos , Software , Animais , Linhagem Celular , Descoberta de Drogas/métodos , Humanos , Camundongos
5.
NPJ Syst Biol Appl ; 3: 23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28861278

RESUMO

We performed an integrated analysis of drug chemical structures and drug-induced transcriptional responses. We demonstrated that a network representing three-dimensional structural similarities among 5452 compounds can be used to automatically group together drugs with similar scaffolds, physicochemical parameters and mode-of-action. We compared the structural network to a network representing transcriptional similarities among a subset of 1309 drugs for which transcriptional response were available in the Connectivity Map data set. Analysis of structurally similar, but transcriptionally different drugs sharing the same MOA enabled us to detect and remove weak and noisy transcriptional responses, greatly enhancing the reliability of transcription-based approaches to drug discovery and drug repositioning. Cardiac glycosides exhibited the strongest transcriptional responses with a significant induction of pathways related to epigenetic regulation, which suggests an epigenetic mechanism of action for these drugs. Drug classes with the weakest transcriptional responses tended to induce expression of cytochrome P450 enzymes, hinting at drug-induced drug resistance. Analysis of transcriptionally similar, but structurally different drugs with unrelated MOA, led us to the identification of a 'toxic' transcriptional signature indicative of lysosomal stress (lysosomotropism) and lipid accumulation (phospholipidosis) partially masking the target-specific transcriptional effects of these drugs. We found that this transcriptional signature is shared by 258 compounds and it is associated to the activation of the transcription factor TFEB, a master regulator of lysosomal biogenesis and autophagy. Finally, we built a predictive Random Forest model of these 258 compounds based on 128 physicochemical parameters, which should help in the early identification of potentially toxic drug candidates.

6.
Sci Rep ; 6: 36016, 2016 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-27786259

RESUMO

Goblet cell hyperplasia, a feature of asthma and other respiratory diseases, is driven by the Th-2 cytokines IL-4 and IL-13. In human bronchial epithelial cells, we find that IL-4 induces the expression of many genes coding for ion channels and transporters, including TMEM16A, SLC26A4, SLC12A2, and ATP12A. At the functional level, we find that IL-4 enhances calcium- and cAMP-activated chloride/bicarbonate secretion, resulting in high bicarbonate concentration and alkaline pH in the fluid covering the apical surface of epithelia. Importantly, mucin release, elicited by purinergic stimulation, requires the presence of bicarbonate in the basolateral solution and is defective in cells derived from cystic fibrosis patients. In conclusion, our results suggest that Th-2 cytokines induce a profound change in expression and function in multiple ion channels and transporters that results in enhanced bicarbonate transport ability. This change is required as an important mechanism to favor release and clearance of mucus.


Assuntos
Bicarbonatos/metabolismo , Células Caliciformes/metabolismo , Hiperplasia/patologia , Mucinas/metabolismo , Células Cultivadas , Cloretos/metabolismo , Citocinas/metabolismo , Perfilação da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio
7.
Oncotarget ; 7(37): 58743-58758, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27542212

RESUMO

The discovery of inhibitors for oncogenic signalling pathways remains a key focus in modern oncology, based on personalized and targeted therapeutics. Computational drug repurposing via the analysis of FDA-approved drug network is becoming a very effective approach to identify therapeutic opportunities in cancer and other human diseases. Given that gene expression signatures can be associated with specific oncogenic mutations, we tested whether a "reverse" oncogene-specific signature might assist in the computational repositioning of inhibitors of oncogenic pathways. As a proof of principle, we focused on oncogenic PI3K-dependent signalling, a molecular pathway frequently driving cancer progression as well as raising resistance to anticancer-targeted therapies. We show that implementation of "reverse" oncogenic PI3K-dependent transcriptional signatures combined with interrogation of drug networks identified inhibitors of PI3K-dependent signalling among FDA-approved compounds. This led to repositioning of Niclosamide (Niclo) and Pyrvinium Pamoate (PP), two anthelmintic drugs, as inhibitors of oncogenic PI3K-dependent signalling. Niclo inhibited phosphorylation of P70S6K, while PP inhibited phosphorylation of AKT and P70S6K, which are downstream targets of PI3K. Anthelmintics inhibited oncogenic PI3K-dependent gene expression and showed a cytostatic effect in vitro and in mouse mammary gland. Lastly, PP inhibited the growth of breast cancer cells harbouring PI3K mutations. Our data indicate that drug repositioning by network analysis of oncogene-specific transcriptional signatures is an efficient strategy for identifying oncogenic pathway inhibitors among FDA-approved compounds. We propose that PP and Niclo should be further investigated as potential therapeutics for the treatment of tumors or diseases carrying the constitutive activation of the PI3K/P70S6K signalling axis.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Biologia Computacional , Reposicionamento de Medicamentos , Glândulas Mamárias Animais/efeitos dos fármacos , Niclosamida/uso terapêutico , Compostos de Pirvínio/uso terapêutico , Animais , Neoplasias da Mama/patologia , Carcinogênese , Linhagem Celular Tumoral , Aprovação de Drogas , Feminino , Humanos , Glândulas Mamárias Animais/patologia , Camundongos , Niclosamida/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Compostos de Pirvínio/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
8.
J Cyst Fibros ; 15(4): 425-35, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26971626

RESUMO

BACKGROUND: Mistrafficking of CFTR protein caused by F508del, the most frequent mutation in cystic fibrosis (CF), can be corrected by cell incubation at low temperature, an effect that may be mediated by altered expression of proteostasis genes. METHODS: To identify small molecules mimicking low temperature, we compared gene expression profiles of cells kept at 27°C with those previously generated from more than 1300 compounds. The resulting candidates were tested with a functional assay on a bronchial epithelial cell line. RESULTS: We found that anti-inflammatory glucocorticoids, such as mometasone, budesonide, and fluticasone, increased mutant CFTR function. However, this activity was not confirmed in primary bronchial epithelial cells. Actually, glucocorticoids enhanced Na(+) absorption, an effect that could further impair mucociliary clearance in CF airways. CONCLUSIONS: Our results suggest that rescue of F508del-CFTR by low temperature cannot be easily mimicked by small molecules and that compounds with closer transcriptional and functional effects need to be found.


Assuntos
Agonistas dos Canais de Cloreto/farmacologia , Canais de Cloreto/fisiologia , Temperatura Baixa , Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Células Epiteliais/metabolismo , Brônquios/patologia , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Reposicionamento de Medicamentos/métodos , Humanos , Depuração Mucociliar/fisiologia , Proteínas Mutantes/metabolismo , Mutação , Biologia de Sistemas/métodos , Transcriptoma/fisiologia
9.
Bioinformatics ; 32(2): 235-41, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26415724

RESUMO

MOTIVATION: Automated screening approaches are able to rapidly identify a set of small molecules inducing a desired phenotype from large small-molecule libraries. However, the resulting set of candidate molecules is usually very diverse pharmacologically, thus little insight on the shared mechanism of action (MoA) underlying their efficacy can be gained. RESULTS: We introduce a computational method (Drug-Set Enrichment Analysis-DSEA) based on drug-induced gene expression profiles, which is able to identify the molecular pathways that are targeted by most of the drugs in the set. By diluting drug-specific effects unrelated to the phenotype of interest, DSEA is able to highlight phenotype-specific pathways, thus helping to formulate hypotheses on the MoA shared by the drugs in the set. We validated the method by analysing five different drug-sets related to well-known pharmacological classes. We then applied DSEA to identify the MoA shared by drugs known to be partially effective in rescuing mutant cystic fibrosis transmembrane conductance regulator (CFTR) gene function in Cystic Fibrosis. AVAILABILITY AND IMPLEMENTATION: The method is implemented as an online web tool publicly available at http://dsea.tigem.it. CONTACT: dibernardo@tigem.it SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Mutação/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Transcriptoma , Humanos , Fenótipo
10.
Bioinformatics ; 30(12): 1787-8, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24558125

RESUMO

SUMMARY: Elucidation of molecular targets of a compound [mode of action (MoA)] and its off-targets is a crucial step in drug development. We developed an online collaborative resource (MANTRA 2.0) that supports this process by exploiting similarities between drug-induced transcriptional profiles. Drugs are organized in a network of nodes (drugs) and edges (similarities) highlighting 'communities' of drugs sharing a similar MoA. A user can upload gene expression profiles before and after drug treatment in one or multiple cell types. An automated processing pipeline transforms the gene expression profiles into a unique drug 'node' embedded in the drug-network. Visual inspection of the neighbouring drugs and communities helps in revealing its MoA and to suggest new applications of known drugs (drug repurposing). MANTRA 2.0 allows storing and sharing user-generated network nodes, thus making MANTRA 2.0 a collaborative ever-growing resource. AVAILABILITY AND IMPLEMENTATION: The web tool is freely available for academic use at http://mantra.tigem.it.


Assuntos
Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/métodos , Perfilação da Expressão Gênica , Software , Comportamento Cooperativo , Internet , Transcriptoma/efeitos dos fármacos
11.
J Chem Inf Model ; 52(12): 3308-24, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23140085

RESUMO

Virtual fragment screening (VFS) is a promising new method that uses computer models to identify small, fragment-like biologically active molecules as useful starting points for fragment-based drug discovery (FBDD). Training sets of true active and inactive fragment-like molecules to construct and validate target customized VFS methods are however lacking. We have for the first time explored the possibilities and challenges of VFS using molecular fingerprints derived from a unique set of fragment affinity data for the histamine H(3) receptor (H(3)R), a pharmaceutically relevant G protein-coupled receptor (GPCR). Optimized FLAP (Fingerprints of Ligands and Proteins) models containing essential molecular interaction fields that discriminate known H(3)R binders from inactive molecules were successfully used for the identification of new H(3)R ligands. Prospective virtual screening of 156,090 molecules yielded a high hit rate of 62% (18 of the 29 tested) experimentally confirmed novel fragment-like H(3)R ligands that offer new potential starting points for the design of H(3)R targeting drugs. The first construction and application of customized FLAP models for the discovery of fragment-like biologically active molecules demonstrates that VFS is an efficient way to explore protein-fragment interaction space in silico.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Receptores Histamínicos H3/química , Receptores Histamínicos H3/metabolismo , Interface Usuário-Computador , Biologia Computacional , Bases de Dados de Proteínas , Análise Discriminante , Ligantes , Simulação de Acoplamento Molecular , Conformação Proteica
12.
Org Biomol Chem ; 10(45): 8994-9003, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23073236

RESUMO

In the framework of our interest in racemic thiazinooxadiazol-3-ones we determined the absolute configuration and the biological activity as L-type calcium channel blockers of two compounds that differ in the length of the acetal chain, which could affect the pharmacological profile. We observed an interesting inversion of the stereoselectivity, with the activity residing on the R-form for a short chain compound (n = 1) and on the S-form for a long chain one (n = 12). The length of the linear acetal chain appears to be able to invert the stereoselectivity of such a class of compounds, and in silico simulations suggested that this different behaviour might be explained by different hydrophilic and hydrophobic interactions with the binding site.


Assuntos
Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Oxidiazóis/química , Oxidiazóis/farmacologia , Elétrons , Modelos Moleculares , Conformação Molecular , Estereoisomerismo
13.
J Comput Aided Mol Des ; 26(11): 1247-66, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23065321

RESUMO

FLAP fingerprints are applied in the ligand-, structure- and pharmacophore-based mode in a case study on antagonists of all four adenosine receptor (AR) subtypes. Structurally diverse antagonist collections with respect to the different ARs were constructed by including binding data to human species only. FLAP models well discriminate "active" (=highly potent) from "inactive" (=weakly potent) AR antagonists, as indicated by enrichment curves, numbers of false positives, and AUC values. For all FLAP modes, model predictivity slightly decreases as follows: A(2B)R > A(2A)R > A(3)R > A(1)R antagonists. General performance of FLAP modes in this study is: ligand- > structure- > pharmacophore- based mode. We also compared the FLAP performance with other common ligand- and structure-based fingerprints. Concerning the ligand-based mode, FLAP model performance is superior to ECFP4 and ROCS for all AR subtypes. Although focusing on the early first part of the A(2A), A(2B) and A(3) enrichment curves, ECFP4 and ROCS still retain a satisfactory retrieval of actives. FLAP is also superior when comparing the structure-based mode with PLANTS and GOLD. In this study we applied for the first time the novel FLAPPharm tool for pharmacophore generation. Pharmacophore hypotheses, generated with this tool, convincingly match with formerly published data. Finally, we could demonstrate the capability of FLAP models to uncover selectivity aspects although single AR subtype models were not trained for this purpose.


Assuntos
Antagonistas do Receptor A1 de Adenosina/química , Antagonistas do Receptor A2 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/química , Modelos Moleculares , Receptores Purinérgicos P1/química , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Humanos , Ligantes , Simulação de Dinâmica Molecular , Conformação Proteica , Relação Estrutura-Atividade
14.
Eur J Med Chem ; 51: 184-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22424612

RESUMO

The chemokine receptor CXCR7 is an atypical G protein-coupled receptor as it preferentially signals through the ß-arrestin pathway rather than through G proteins. CXCR7 is thought to be of importance in cancer and the development of CXCR7-targeting ligands is of huge importance to further elucidate the pharmacology and the therapeutic potential of CXCR7. In the present study, we synthesized 24 derivatives based on a compound scaffold patented by Chemocentryx and obtained CXCR7 ligands with pK(i) values ranging from 5.3 to 8.1. SAR studies were supported by computational 3D Fingerprint studies, revealing several important affinity descriptors. Two key compounds (29 and 30, VUF11207 and VUF11403) were found to be high-potency ligands that induce recruitment of ß-arrestin2 and subsequent internalization of CXCR7, making them important tool compounds in future CXCR7 research.


Assuntos
Amidas/química , Técnicas de Química Sintética , Modelos Moleculares , Receptores CXCR/agonistas , Estireno/química , Estireno/farmacologia , Células HEK293 , Humanos , Conformação Molecular , Relação Quantitativa Estrutura-Atividade , Estireno/síntese química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...