Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928221

RESUMEN

Methionine oxidation to the sulfoxide form (MSox) is a poorly understood post-translational modification of proteins associated with non-specific chemical oxidation from reactive oxygen species (ROS), whose chemistries are linked to various disease pathologies, including neurodegeneration. Emerging evidence shows MSox site occupancy is, in some cases, under enzymatic regulatory control, mediating cellular signaling, including phosphorylation and/or calcium signaling, and raising questions as to the speciation and functional nature of MSox across the proteome. The 5XFAD lineage of the C57BL/6 mouse has well-defined Alzheimer's and aging states. Using this model, we analyzed age-, sex-, and disease-dependent MSox speciation in the mouse hippocampus. In addition, we explored the chemical stability and statistical variance of oxidized peptide signals to understand the needed power for MSox-based proteome studies. Our results identify mitochondrial and glycolytic pathway targets with increases in MSox with age as well as neuroinflammatory targets accumulating MSox with AD in proteome studies of the mouse hippocampus. Further, this paper establishes a foundation for reproducible and rigorous experimental MSox-omics appropriate for novel target identification in biological discovery and for biomarker analysis in ROS and other oxidation-linked diseases.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer , Glucólisis , Hipocampo , Metionina , Ratones Endogámicos C57BL , Mitocondrias , Proteómica , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Hipocampo/metabolismo , Ratones , Mitocondrias/metabolismo , Proteómica/métodos , Metionina/metabolismo , Metionina/análogos & derivados , Envejecimiento/metabolismo , Masculino , Femenino , Oxidación-Reducción , Proteoma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Modelos Animales de Enfermedad
2.
Bioinformatics ; 38(15): 3785-3793, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35731218

RESUMEN

MOTIVATION: Protein phosphorylation is a ubiquitous regulatory mechanism that plays a central role in cellular signaling. According to recent estimates, up to 70% of human proteins can be phosphorylated. Therefore, the characterization of phosphorylation dynamics is critical for understanding a broad range of biological and biochemical processes. Technologies based on mass spectrometry are rapidly advancing to meet the needs for high-throughput screening of phosphorylation. These technologies enable untargeted quantification of thousands of phosphorylation sites in a given sample. Many labs are already utilizing these technologies to comprehensively characterize signaling landscapes by examining perturbations with drugs and knockdown approaches, or by assessing diverse phenotypes in cancers, neuro-degerenational diseases, infectious diseases and normal development. RESULTS: We comprehensively investigate the concept of 'co-phosphorylation' (Co-P), defined as the correlated phosphorylation of a pair of phosphosites across various biological states. We integrate nine publicly available phosphoproteomics datasets for various diseases (including breast cancer, ovarian cancer and Alzheimer's disease) and utilize functional data related to sequence, evolutionary histories, kinase annotations and pathway annotations to investigate the functional relevance of Co-P. Our results across a broad range of studies consistently show that functionally associated sites tend to exhibit significant positive or negative Co-P. Specifically, we show that Co-P can be used to predict with high precision the sites that are on the same pathway or that are targeted by the same kinase. Overall, these results establish Co-P as a useful resource for analyzing phosphoproteins in a network context, which can help extend our knowledge on cellular signaling and its dysregulation. AVAILABILITY AND IMPLEMENTATION: github.com/msayati/Cophosphorylation. This research used the publicly available datasets published by other researchers as cited in the manuscript. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Fosfoproteínas , Proteómica , Humanos , Fosforilación , Proteómica/métodos , Fosfoproteínas/química , Espectrometría de Masas/métodos , Fosfotransferasas/metabolismo
3.
Medicina (Kaunas) ; 59(1)2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36676744

RESUMEN

Background and Objectives: There is no biomarker to predict lithium response. This study used CellPrint™ enhanced flow cytometry to study 28 proteins representing a spectrum of cellular pathways in monocytes and CD4+ lymphocytes before and after lithium treatment in patients with bipolar disorder (BD). Materials and Methods: Symptomatic patients with BD type I or II received lithium (serum level ≥ 0.6 mEq/L) for 16 weeks. Patients were assessed with standard rating scales and divided into two groups, responders (≥50% improvement from baseline) and non-responders. Twenty-eight intracellular proteins in CD4+ lymphocytes and monocytes were analyzed with CellPrint™, an enhanced flow cytometry procedure. Data were analyzed for differences in protein expression levels. Results: The intent-to-treat sample included 13 lithium-responders (12 blood samples before treatment and 9 after treatment) and 11 lithium-non-responders (11 blood samples before treatment and 4 after treatment). No significant differences in expression between the groups was observed prior to lithium treatment. After treatment, the majority of analytes increased expression in responders and decreased expression in non-responders. Significant increases were seen for PDEB4 and NR3C1 in responders. A significant decrease was seen for NR3C1 in non-responders. Conclusions: Lithium induced divergent directionality of protein expression depending on the whether the patient was a responder or non-responder, elucidating molecular characteristics of lithium responsiveness. A subsequent study with a larger sample size is warranted.


Asunto(s)
Trastorno Bipolar , Litio , Humanos , Litio/farmacología , Litio/uso terapéutico , Trastorno Bipolar/tratamiento farmacológico , Compuestos de Litio , Citometría de Flujo , Línea Celular
4.
Bioinformatics ; 37(2): 221-228, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-32730576

RESUMEN

MOTIVATION: Protein phosphorylation is a ubiquitous mechanism of post-translational modification that plays a central role in cellular signaling. Phosphorylation is particularly important in the context of cancer, as downregulation of tumor suppressors and upregulation of oncogenes by the dysregulation of associated kinase and phosphatase networks are shown to have key roles in tumor growth and progression. Despite recent advances that enable large-scale monitoring of protein phosphorylation, these data are not fully incorporated into such computational tasks as phenotyping and subtyping of cancers. RESULTS: We develop a network-based algorithm, CoPPNet, to enable unsupervised subtyping of cancers using phosphorylation data. For this purpose, we integrate prior knowledge on evolutionary, structural and functional association of phosphosites, kinase-substrate associations and protein-protein interactions with the correlation of phosphorylation of phosphosites across different tumor samples (a.k.a co-phosphorylation) to construct a context-specific-weighted network of phosphosites. We then mine these networks to identify subnetworks with correlated phosphorylation patterns. We apply the proposed framework to two mass-spectrometry-based phosphorylation datasets for breast cancer (BC), and observe that (i) the phosphorylation pattern of the identified subnetworks are highly correlated with clinically identified subtypes, and (ii) the identified subnetworks are highly reproducible across datasets that are derived from different studies. Our results show that integration of quantitative phosphorylation data with network frameworks can provide mechanistic insights into the differences between the signaling mechanisms that drive BC subtypes. Furthermore, the reproducibility of the identified subnetworks suggests that phosphorylation can provide robust classification of disease response and markers. AVAILABILITY AND IMPLEMENTATION: CoPPNet is available at http://compbio.case.edu/coppnet/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/genética , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Reproducibilidad de los Resultados , Transducción de Señal
5.
PLoS Comput Biol ; 15(2): e1006678, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30811403

RESUMEN

We present CoPhosK to predict kinase-substrate associations for phosphopeptide substrates detected by mass spectrometry (MS). The tool utilizes a Naïve Bayes framework with priors of known kinase-substrate associations (KSAs) to generate its predictions. Through the mining of MS data for the collective dynamic signatures of the kinases' substrates revealed by correlation analysis of phosphopeptide intensity data, the tool infers KSAs in the data for the considerable body of substrates lacking such annotations. We benchmarked the tool against existing approaches for predicting KSAs that rely on static information (e.g. sequences, structures and interactions) using publically available MS data, including breast, colon, and ovarian cancer models. The benchmarking reveals that co-phosphorylation analysis can significantly improve prediction performance when static information is available (about 35% of sites) while providing reliable predictions for the remainder, thus tripling the KSAs available from the experimental MS data providing to a comprehensive and reliable characterization of the landscape of kinase-substrate interactions well beyond current limitations.


Asunto(s)
Biología Computacional/métodos , Proteínas Quinasas/fisiología , Especificidad por Sustrato/fisiología , Teorema de Bayes , Sitios de Unión , Bases de Datos de Proteínas , Humanos , Espectrometría de Masas , Fosforilación/fisiología , Fosfotransferasas/fisiología , Unión Proteica , Mapeo de Interacción de Proteínas , Proteoma , Análisis de Secuencia de Proteína , Programas Informáticos
6.
Proteomics ; 17(22)2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28961369

RESUMEN

Activation of protein phosphatase 2A (PP2A) is a promising anticancer therapeutic strategy, as this tumor suppressor has the ability to coordinately downregulate multiple pathways involved in the regulation of cellular growth and proliferation. In order to understand the systems-level perturbations mediated by PP2A activation, we carried out mass spectrometry-based phosphoproteomic analysis of two KRAS mutated non-small cell lung cancer (NSCLC) cell lines (A549 and H358) treated with a novel small molecule activator of PP2A (SMAP). Overall, this permitted quantification of differential signaling across over 1600 phosphoproteins and 3000 phosphosites. Kinase activity assessment and pathway enrichment implicate collective downregulation of RAS and cell cycle kinases in the case of both cell lines upon PP2A activation. However, the effects on RAS-related signaling are attenuated for A549 compared to H358, while the effects on cell cycle-related kinases are noticeably more prominent in A549. Network-based analyses and validation experiments confirm these detailed differences in signaling. These studies reveal the power of phosphoproteomics studies, coupled to computational systems biology, to elucidate global patterns of phosphatase activation and understand the variations in response to PP2A activation across genetically similar NSCLC cell lines.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfoproteínas/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteómica/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Espectrometría de Masas , Fosforilación , Transducción de Señal
7.
PLoS Comput Biol ; 12(11): e1005195, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27835645

RESUMEN

Susceptibility loci identified by GWAS generally account for a limited fraction of heritability. Predictive models based on identified loci also have modest success in risk assessment and therefore are of limited practical use. Many methods have been developed to overcome these limitations by incorporating prior biological knowledge. However, most of the information utilized by these methods is at the level of genes, limiting analyses to variants that are in or proximate to coding regions. We propose a new method that integrates protein protein interaction (PPI) as well as expression quantitative trait loci (eQTL) data to identify sets of functionally related loci that are collectively associated with a trait of interest. We call such sets of loci "population covering locus sets" (PoCos). The contributions of the proposed approach are three-fold: 1) We consider all possible genotype models for each locus, thereby enabling identification of combinatorial relationships between multiple loci. 2) We develop a framework for the integration of PPI and eQTL into a heterogenous network model, enabling efficient identification of functionally related variants that are associated with the disease. 3) We develop a novel method to integrate the genotypes of multiple loci in a PoCo into a representative genotype to be used in risk assessment. We test the proposed framework in the context of risk assessment for seven complex diseases, type 1 diabetes (T1D), type 2 diabetes (T2D), psoriasis (PS), bipolar disorder (BD), coronary artery disease (CAD), hypertension (HT), and multiple sclerosis (MS). Our results show that the proposed method significantly outperforms individual variant based risk assessment models as well as the state-of-the-art polygenic score. We also show that incorporation of eQTL data improves the performance of identified POCOs in risk assessment. We also assess the biological relevance of PoCos for three diseases that have similar biological mechanisms and identify novel candidate genes. The resulting software is publicly available at http://compbio. CASE: edu/pocos/.


Asunto(s)
Estudios de Asociación Genética/métodos , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Sitios de Carácter Cuantitativo/genética , Medición de Riesgo/métodos , Algoritmos , Humanos , Prevalencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
8.
J Clin Med ; 13(5)2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38592374

RESUMEN

Background: The mechanism of lithium treatment responsiveness in bipolar disorder (BD) remains unclear. The aim of this study was to explore the utility of correlation coefficients and protein-to-protein interaction (PPI) network analyses of intracellular proteins in monocytes and CD4+ lymphocytes of patients with BD in studying the potential mechanism of lithium treatment responsiveness. Methods: Patients with bipolar I or II disorder who were diagnosed with the MINI for DSM-5 and at any phase of the illness with at least mild symptom severity and received lithium (serum level ≥ 0.6 mEq/L) for 16 weeks were divided into two groups, responders (≥50% improvement in Montgomery-Asberg Depression Rating Scale and/or Young Mania Rating Scale scores from baseline) and non-responders. Twenty-eight intracellular proteins/analytes in CD4+ lymphocytes and monocytes were analyzed with a tyramine-based signal-amplified flow cytometry procedure. Correlation coefficients between analytes at baseline were estimated in both responders and non-responders and before and after lithium treatment in responders. PPI network, subnetwork, and pathway analyses were generated based on fold change/difference in studied proteins/analytes between responders and non-responders. Results: Of the 28 analytes from 12 lithium-responders and 11 lithium-non-responders, there were more significant correlations between analytes in responders than in non-responders at baseline. Of the nine lithium responders with pre- and post-lithium blood samples available, the correlations between most analytes were weakened after lithium treatment with cell-type specific patterns in CD4+ lymphocytes and monocytes. PPI network/subnetwork and pathway analyses showed that lithium response was involved in four pathways, including prolactin, leptin, neurotrophin, and brain-derived neurotrophic factor pathways. Glycogen synthase kinase 3 beta and nuclear factor NF-kappa-B p65 subunit genes were found in all four pathways. Conclusions: Using correlation coefficients, PPI network/subnetwork, and pathway analysis with multiple intracellular proteins appears to be a workable concept for studying the mechanism of lithium responsiveness in BD. Larger sample size studies are necessary to determine its utility.

9.
Pac Symp Biocomput ; 28: 73-84, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36540966

RESUMEN

Protein phosphorylation is a key post-translational modification that plays a central role in many cellular processes. With recent advances in biotechnology, thousands of phosphorylated sites can be identified and quantified in a given sample, enabling proteome-wide screening of cellular signaling. However, for most (> 90%) of the phosphorylation sites that are identified in these experiments, the kinase(s) that target these sites are unknown. To broadly utilize available structural, functional, evolutionary, and contextual information in predicting kinase-substrate associations (KSAs), we develop a network-based machine learning framework. Our framework integrates a multitude of data sources to characterize the landscape of functional relationships and associations among phosphosites and kinases. To construct a phosphosite-phosphosite association network, we use sequence similarity, shared biological pathways, co-evolution, co-occurrence, and co-phosphorylation of phosphosites across different biological states. To construct a kinase-kinase association network, we integrate protein-protein interactions, shared biological pathways, and membership in common kinase families. We use node embeddings computed from these heterogeneous networks to train machine learning models for predicting kinase-substrate associations. Our systematic computational experiments using the PhosphositePLUS database shows that the resulting algorithm, NetKSA, outperforms two state-of-the-art algorithms, including KinomeXplorer and LinkPhinder, in overall KSA prediction. By stratifying the ranking of kinases, NetKSA also enables annotation of phosphosites that are targeted by relatively less-studied kinases.Availability: The code and data are available at compbio.case.edu/NetKSA/.


Asunto(s)
Biología Computacional , Proteínas Quinasas , Humanos , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Biología Computacional/métodos , Algoritmos
10.
J Affect Disord ; 328: 116-127, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36806598

RESUMEN

BACKGROUND: Molecular biomarkers for bipolar disorder (BD) that distinguish it from other manifestations of depressive symptoms remain unknown. The aim of this study was to determine if a very sensitive tyramine-based signal-amplification technology for flow cytometry (CellPrint™) could facilitate the identification of cell-specific analyte expression profiles of peripheral blood cells for bipolar depression (BPD) versus healthy controls (HCs). METHODS: The diagnosis of psychiatric disorders was ascertained with Mini International Neuropsychiatric Interview for DSM-5. Expression levels for eighteen protein analytes previously shown to be related to bipolar disorder were assessed with CellPrint™ in CD4+ T cells and monocytes of bipolar patients and HCs. Implementation of protein-protein interaction (PPI) network and pathway analysis was subsequently used to identify new analytes and pathways for subsequent interrogations. RESULTS: Fourteen drug-naïve or -free patients with bipolar I or II depression and 17 healthy controls (HCs) were enrolled. The most distinguishable changes in analyte expression based on t-tests included GSK3ß, HMGB1, IRS2, phospho-GSK3αß, phospho-RELA, and TSPO in CD4+ T cells and calmodulin, GSK3ß, IRS2, and phospho-HS1 in monocytes. Subsequent PPI and pathway analysis indicated that prolactin, leptin, BDNF, and interleukin-3 signal pathways were significantly different between bipolar patients and HCs. LIMITATION: The sample size of the study was small and 2 patients were on medications. CONCLUSION: In this pilot study, CellPrint™ was able to detect differences in cell-specific protein levels between BPD patients and HCs. A subsequent study including samples from patients with BPD, major depressive disorder, and HCs is warranted.


Asunto(s)
Trastorno Bipolar , Trastorno Depresivo Mayor , Humanos , Trastorno Bipolar/psicología , Monocitos/metabolismo , Proyectos Piloto , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Citometría de Flujo , Linfocitos T CD4-Positivos/metabolismo , Receptores de GABA/metabolismo
11.
Psychopharmacol Bull ; 52(1): 8-35, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35342205

RESUMEN

Purpose: To determine if enhanced flow cytometry (CellPrint™) can identify intracellular proteins of lithium responsiveness in monocytes and CD4+ lymphocytes from patients with bipolar disorder. Methods: Eligible bipolar I or II patients were openly treated with lithium for 16-weeks. Baseline levels of Bcl2, BDNF, calmodulin, Fyn, phospho-Fyn/phospho-Yes, GSK3ß, phospho-GSK3αß, HMGB1, iNOS, IRS2, mTor, NLPR3, PGM1, PKA C-α, PPAR-γ, phospho-RelA, and TPH1 in monocytes and CD4+ lymphocytes of lithium responders and non-responders were measured with CellPrint™. Their utility of discriminating responders from non-responders was explored. Protein-protein network and pathway enrichment analyses were conducted. Results: Of the 24 intent-to-treat patients, 12 patients completed the 16-week study. Eleven of 13 responders and 8 of 11 non-responders were available for this analysis. The levels of the majority of analytes in lithium responders were lower than non-responders in both cell types, but only the level of GSK3ß in monocytes was significantly different (p = 0.034). The combination of GSK3ß and phospho-GSK3αß levels in monocytes correctly classified 11/11 responders and 5/8 non-responders. Combination of GSK3ß, phospho-RelA, TPH1 and PGM1 correctly classified 10/11 responders and 6/7 non-responders, both with a likelihood of ≥ 85%. Prolactin, leptin, BDNF, neurotrophin, and epidermal growth factor/epidermal growth factor receptor signaling pathways are involved in the lithium treatment response. GSK3ß and RelA genes are involved in 4 of 5 these pathways. Conclusion: CellPrint™ flow cytometry was able to detect differences in multiple proteins in monocytes and CD4+ lymphocytes between lithium responders and non-responders. A large study is warranted to confirm or refute these findings.


Asunto(s)
Trastorno Bipolar , Biomarcadores , Trastorno Bipolar/tratamiento farmacológico , Factor Neurotrófico Derivado del Encéfalo , Linfocitos T CD4-Positivos , Estudios de Factibilidad , Citometría de Flujo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Litio/farmacología , Litio/uso terapéutico , Compuestos de Litio , Monocitos , Tiramina
12.
Commun Biol ; 5(1): 1251, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36380187

RESUMEN

Alterations of serine/threonine phosphorylation of the cardiac proteome are a hallmark of heart failure. However, the contribution of tyrosine phosphorylation (pTyr) to the pathogenesis of cardiac hypertrophy remains unclear. We use global mapping to discover and quantify site-specific pTyr in two cardiac hypertrophic mouse models, i.e., cardiac overexpression of ErbB2 (TgErbB2) and α myosin heavy chain R403Q (R403Q-αMyHC Tg), compared to control hearts. From this, there are significant phosphoproteomic alterations in TgErbB2 mice in right ventricular cardiomyopathy, hypertrophic cardiomyopathy (HCM), and dilated cardiomyopathy (DCM) pathways. On the other hand, R403Q-αMyHC Tg mice indicated that the EGFR1 pathway is central for cardiac hypertrophy, along with angiopoietin, ErbB, growth hormone, and chemokine signaling pathways activation. Surprisingly, most myofilament proteins have downregulation of pTyr rather than upregulation. Kinase-substrate enrichment analysis (KSEA) shows a marked downregulation of MAPK pathway activity downstream of k-Ras in TgErbB2 mice and activation of EGFR, focal adhesion, PDGFR, and actin cytoskeleton pathways. In vivo ErbB2 inhibition by AG-825 decreases cardiomyocyte disarray. Serine/threonine and tyrosine phosphoproteome confirm the above-described pathways and the effectiveness of AG-825 Treatment. Thus, altered pTyr may play a regulatory role in cardiac hypertrophic models.


Asunto(s)
Cardiomiopatía Hipertrófica , Proteoma , Ratones , Animales , Proteoma/metabolismo , Fosforilación , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Cardiomegalia , Serina/metabolismo , Treonina/metabolismo , Tirosina/metabolismo
13.
Cells ; 10(9)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34571868

RESUMEN

Plants and insect herbivores are in a relentless battle to outwit each other. Plants have evolved various strategies to detect herbivores and mount an effective defense system against them. These defenses include physical and structural barriers such as spines, trichomes, cuticle, or chemical compounds, including secondary metabolites such as phenolics and terpenes. Plants perceive herbivory by both mechanical and chemical means. Mechanical sensing can occur through the perception of insect biting, piercing, or chewing, while chemical signaling occurs through the perception of various herbivore-derived compounds such as oral secretions (OS) or regurgitant, insect excreta (frass), or oviposition fluids. Interestingly, ion channels or transporters are the first responders for the perception of these mechanical and chemical cues. These transmembrane pore proteins can play an important role in plant defense through the induction of early signaling components such as plasma transmembrane potential (Vm) fluctuation, intracellular calcium (Ca2+), and reactive oxygen species (ROS) generation, followed by defense gene expression, and, ultimately, plant defense responses. In recent years, studies on early plant defense signaling in response to herbivory have been gaining momentum with the application of genetically encoded GFP-based sensors for real-time monitoring of early signaling events and genetic tools to manipulate ion channels involved in plant-herbivore interactions. In this review, we provide an update on recent developments and advances on early signaling events in plant-herbivore interactions, with an emphasis on the role of ion channels in early plant defense signaling.


Asunto(s)
Herbivoria/fisiología , Insectos/metabolismo , Canales Iónicos/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Potenciales de la Membrana/fisiología , Plantas/metabolismo
14.
Nat Commun ; 12(1): 1177, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33608514

RESUMEN

Mass spectrometry enables high-throughput screening of phosphoproteins across a broad range of biological contexts. When complemented by computational algorithms, phospho-proteomic data allows the inference of kinase activity, facilitating the identification of dysregulated kinases in various diseases including cancer, Alzheimer's disease and Parkinson's disease. To enhance the reliability of kinase activity inference, we present a network-based framework, RoKAI, that integrates various sources of functional information to capture coordinated changes in signaling. Through computational experiments, we show that phosphorylation of sites in the functional neighborhood of a kinase are significantly predictive of its activity. The incorporation of this knowledge in RoKAI consistently enhances the accuracy of kinase activity inference methods while making them more robust to missing annotations and quantifications. This enables the identification of understudied kinases and will likely lead to the development of novel kinase inhibitors for targeted therapy of many diseases. RoKAI is available as web-based tool at http://rokai.io .


Asunto(s)
Biología Computacional/métodos , Redes y Vías Metabólicas , Fosfotransferasas/metabolismo , Transducción de Señal/fisiología , Algoritmos , Enfermedad de Alzheimer/metabolismo , Redes Reguladoras de Genes/fisiología , Humanos , Espectrometría de Masas , Neoplasias/metabolismo , Enfermedad de Parkinson/metabolismo , Fosfoproteínas , Fosforilación , Fosfotransferasas/genética , Proteómica/métodos , Reproducibilidad de los Resultados , Programas Informáticos , Biología de Sistemas/métodos
15.
Int J Bioinform Res Appl ; 11(1): 72-89, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25667386

RESUMEN

Removal of proteins on an essential pathway of a pathogen is expected to prohibit the pathogen from performing a vital function. To disrupt these pathways, we consider a cut set S of simple graph G, where G representing the PPI network of the pathogen. After removing S, if the difference of sizes of two partitions is high, the probability of existence of a functioning pathway is increased. We need to partition the graph into balanced partitions and approximate it with spectral bipartitioning. We consider two scenarios: in the first, we do not have any information on drug targets; in second, we consider information on drug targets. Our databases are E. coli and C. jejuni. In the first scenario, 20% and 17% of proteins in cut of E. coli and C. jejuni are drug targets and in the second scenario 53% and 63% of proteins in cut are drug targets respectively.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Diseño de Fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Modelos Biológicos , Mapeo de Interacción de Proteínas/métodos , Simulación por Computador , Farmacorresistencia Bacteriana/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
EURASIP J Bioinform Syst Biol ; 2015: 7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28194175

RESUMEN

Network-based analyses are commonly used as powerful tools to interpret the findings of genome-wide association studies (GWAS) in a functional context. In particular, identification of disease-associated functional modules, i.e., highly connected protein-protein interaction (PPI) subnetworks with high aggregate disease association, are shown to be promising in uncovering the functional relationships among genes and proteins associated with diseases. An important issue in this regard is the scoring of subnetworks by integrating two quantities: disease association of individual gene products and network connectivity among proteins. Current scoring schemes either disregard the level of connectivity and focus on the aggregate disease association of connected proteins or use a linear combination of these two quantities. However, such scoring schemes may produce arbitrarily large subnetworks which are often not statistically significant or require tuning of parameters that are used to weigh the contributions of network connectivity and disease association. Here, we propose a parameter-free scoring scheme that aims to score subnetworks by assessing the disease association of interactions between pairs of gene products. We also incorporate the statistical significance of network connectivity and disease association into the scoring function. We test the proposed scoring scheme on a GWAS dataset for two complex diseases type II diabetes (T2D) and psoriasis (PS). Our results suggest that subnetworks identified by commonly used methods may fail tests of statistical significance after correction for multiple hypothesis testing. In contrast, the proposed scoring scheme yields highly significant subnetworks, which contain biologically relevant proteins that cannot be identified by analysis of genome-wide association data alone. We also show that the proposed scoring scheme identifies subnetworks that are reproducible across different cohorts, and it can robustly recover relevant subnetworks at lower sampling rates.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA