Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(14): 2395-2397, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35803242

RESUMEN

Flaviviruses, such as Dengue and Zika viruses, infect millions of people worldwide using mosquitos as vectors. In this issue of Cell, Zhang et al. reveal how these viruses manipulate the skin microbiome of infected hosts in a way that increases vector recruitment and viral spread. They propose vitamin A as a way to counteract the virus and decrease transmission.


Asunto(s)
Infecciones por Flavivirus , Flavivirus , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Piel , Animales , Culicidae/virología , Dengue , Flavivirus/fisiología , Infecciones por Flavivirus/microbiología , Infecciones por Flavivirus/transmisión , Humanos , Publicaciones Periódicas como Asunto , Piel/metabolismo , Piel/microbiología , Enfermedades Transmitidas por Vectores , Infección por el Virus Zika
2.
Nucleic Acids Res ; 50(D1): D701-D709, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34634810

RESUMEN

Signaling networks represent the molecular mechanisms controlling a cell's response to various internal or external stimuli. Most currently available signaling databases contain only a part of the complex network of intertwining pathways, leaving out key interactions or processes. Hence, we have developed SignaLink3 (http://signalink.org/), a value-added knowledge-base that provides manually curated data on signaling pathways and integrated data from several types of databases (interaction, regulation, localisation, disease, etc.) for humans, and three major animal model organisms. SignaLink3 contains over 400 000 newly added human protein-protein interactions resulting in a total of 700 000 interactions for Homo sapiens, making it one of the largest integrated signaling network resources. Next to H. sapiens, SignaLink3 is the only current signaling network resource to provide regulatory information for the model species Caenorhabditis elegans and Danio rerio, and the largest resource for Drosophila melanogaster. Compared to previous versions, we have integrated gene expression data as well as subcellular localization of the interactors, therefore uniquely allowing tissue-, or compartment-specific pathway interaction analysis to create more accurate models. Data is freely available for download in widely used formats, including CSV, PSI-MI TAB or SQL.


Asunto(s)
Bases de Datos Genéticas , Redes Reguladoras de Genes/genética , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética , Animales , Caenorhabditis elegans/genética , Drosophila melanogaster/genética , Humanos , Pez Cebra/genética
3.
Int J Mol Sci ; 24(8)2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37108830

RESUMEN

One of the main inducers of autophagy-dependent self-cannibalism, called ULK1, is tightly regulated by the two sensor molecules of nutrient conditions and energy status, known as mTOR and AMPK kinases, respectively. Recently, we developed a freely available mathematical model to explore the oscillatory characteristic of the AMPK-mTOR-ULK1 regulatory triangle. Here, we introduce a systems biology analysis to explain in detail the dynamical features of the essential negative and double-negative feedback loops and also the periodic repeat of autophagy induction upon cellular stress. We propose an additional regulatory molecule in the autophagy control network that delays some of AMPK's effect on the system, making the model output more consistent with experimental results. Furthermore, a network analysis on AutophagyNet was carried out to identify which proteins could be the proposed regulatory components in the system. These regulatory proteins should satisfy the following rules: (1) they are induced by AMPK; (2) they promote ULK1; (3) they down-regulate mTOR upon cellular stress. We have found 16 such regulatory components that have been experimentally proven to satisfy at least two of the given rules. Identifying such critical regulators of autophagy induction could support anti-cancer- and ageing-related therapeutic efforts.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas Activadas por AMP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/farmacología , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Biología de Sistemas , Serina-Treonina Quinasas TOR/metabolismo , Autofagia
4.
Appl Environ Microbiol ; 88(16): e0053322, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35916501

RESUMEN

Bacterial extracellular vesicles (BEVs) released from both Gram-negative and Gram-positive bacteria provide an effective means of communication and trafficking of cell signaling molecules. In the gastrointestinal tract (GIT) BEVs produced by members of the intestinal microbiota can impact host health by mediating microbe-host cell interactions. A major unresolved question, however, is what factors influence the composition of BEV proteins and whether the host influences protein packaging into BEVs and secretion into the GIT. To address this, we have analyzed the proteome of BEVs produced by the major human gut symbiont Bacteroides thetaiotaomicron both in vitro and in vivo in the murine GIT in order to identify proteins specifically enriched in BEVs produced in vivo. We identified 113 proteins enriched in BEVs produced in vivo, the majority (62/113) of which accumulated in BEVs in the absence of any changes in their expression by the parental cells. Among these selectively enriched proteins, we identified dipeptidyl peptidases and an asparaginase and confirmed their increased activity in BEVs produced in vivo. We also showed that intact BEVs are capable of degrading bile acids via a bile salt hydrolase. Collectively these findings provide additional evidence for the dynamic interplay of host-microbe interactions in the GIT and the existence of an active mechanism to drive and enrich a selected group of proteins for secretion into BEVs in the GIT. IMPORTANCE The gastrointestinal tract (GIT) harbors a complex community of microbes termed the microbiota that plays a role in maintaining the host's health and wellbeing. How this comes about and the nature of microbe-host cell interactions in the GIT is still unclear. Recently, nanosized vesicles naturally produced by bacterial constituents of the microbiota have been shown to influence responses of different host cells although the molecular basis and identity of vesicle-born bacterial proteins that mediate these interactions is unclear. We show here that bacterial extracellular vesicles (BEVs) produced by the human symbiont Bacteroides thetaiotaomicron in the GIT are enriched in a set of proteins and enzymes, including dipeptidyl peptidases, an asparaginase and a bile salt hydrolase that can influence host cell biosynthetic pathways. Our results provide new insights into the molecular basis of microbiota-host interactions that are central to maintaining GIT homeostasis and health.


Asunto(s)
Bacteroides thetaiotaomicron , Vesículas Extracelulares , Animales , Asparaginasa/metabolismo , Bacterias , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Vesículas Extracelulares/metabolismo , Microbioma Gastrointestinal , Humanos , Ratones , Proteoma/metabolismo
5.
Mol Syst Biol ; 17(3): e9923, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33749993

RESUMEN

Molecular knowledge of biological processes is a cornerstone in omics data analysis. Applied to single-cell data, such analyses provide mechanistic insights into individual cells and their interactions. However, knowledge of intercellular communication is scarce, scattered across resources, and not linked to intracellular processes. To address this gap, we combined over 100 resources covering interactions and roles of proteins in inter- and intracellular signaling, as well as transcriptional and post-transcriptional regulation. We added protein complex information and annotations on function, localization, and role in diseases for each protein. The resource is available for human, and via homology translation for mouse and rat. The data are accessible via OmniPath's web service (https://omnipathdb.org/), a Cytoscape plug-in, and packages in R/Bioconductor and Python, providing access options for computational and experimental scientists. We created workflows with tutorials to facilitate the analysis of cell-cell interactions and affected downstream intracellular signaling processes. OmniPath provides a single access point to knowledge spanning intra- and intercellular processes for data analysis, as we demonstrate in applications studying SARS-CoV-2 infection and ulcerative colitis.


Asunto(s)
COVID-19/metabolismo , Colitis Ulcerosa/metabolismo , Biología Computacional/métodos , Proteínas/metabolismo , Transducción de Señal , Animales , Comunicación Celular , Colitis Ulcerosa/patología , Bases de Datos Factuales , Enzimas/metabolismo , Humanos , Ratones , Procesamiento Proteico-Postraduccional , Proteínas/genética , Ratas , Análisis de la Célula Individual , Programas Informáticos , Flujo de Trabajo
6.
PLoS Comput Biol ; 17(2): e1008685, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33534793

RESUMEN

The SARS-CoV-2 pandemic of 2020 has mobilised scientists around the globe to research all aspects of the coronavirus virus and its infection. For fruitful and rapid investigation of viral pathomechanisms, a collaborative and interdisciplinary approach is required. Therefore, we have developed ViralLink: a systems biology workflow which reconstructs and analyses networks representing the effect of viruses on intracellular signalling. These networks trace the flow of signal from intracellular viral proteins through their human binding proteins and downstream signalling pathways, ending with transcription factors regulating genes differentially expressed upon viral exposure. In this way, the workflow provides a mechanistic insight from previously identified knowledge of virally infected cells. By default, the workflow is set up to analyse the intracellular effects of SARS-CoV-2, requiring only transcriptomics counts data as input from the user: thus, encouraging and enabling rapid multidisciplinary research. However, the wide-ranging applicability and modularity of the workflow facilitates customisation of viral context, a priori interactions and analysis methods. Through a case study of SARS-CoV-2 infected bronchial/tracheal epithelial cells, we evidence the functionality of the workflow and its ability to identify key pathways and proteins in the cellular response to infection. The application of ViralLink to different viral infections in a context specific manner using different available transcriptomics datasets will uncover key mechanisms in viral pathogenesis.


Asunto(s)
COVID-19/metabolismo , Biología Computacional/métodos , Regulación Viral de la Expresión Génica , SARS-CoV-2/patogenicidad , Transducción de Señal , Algoritmos , Bronquios/virología , Análisis por Conglomerados , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Investigación Interdisciplinaria , Pulmón/virología , Modelos Estadísticos , Biología de Sistemas , Transcriptoma , Flujo de Trabajo
7.
Ecotoxicol Environ Saf ; 230: 113130, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34968797

RESUMEN

Stress-responsive microRNAs (miRNAs) contribute to the regulation of cellular homeostasis or pathological processes, including carcinogenesis, by reprogramming target gene expression following human exposure to environmental or dietary xenobiotics. Herein, we predicted the targets of carcinogenic mycotoxin-responsive miRNAs and analyzed their association with disease and functionality. miRNA target-derived prediction indicated potent associations of oncogenic mycotoxin exposure with metabolism- or hormone-related diseases, including sex hormone-linked cancers. Mechanistically, the signaling network evaluation suggested androgen receptor (AR)-linked signaling as a common pivotal cluster associated with metabolism- or hormone-related tumorigenesis in response to aflatoxin B1 and ochratoxin A co-exposure. Particularly, high levels of AR and AR-linked genes for the retinol and xenobiotic metabolic enzymes were positively associated with attenuated disease biomarkers and good prognosis in patients with liver or kidney cancers. Moreover, AR-linked signaling was protective against OTA-induced genetic insults in human hepatocytes whereas it was positively involved in AFB1-induced genotoxic actions. Collectively, miRNA target network-based predictions provide novel clinical insights into the progression or intervention against malignant adverse outcomes of human exposure to environmental oncogenic insults.

8.
Brief Bioinform ; 20(1): 89-101, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28968712

RESUMEN

Biomarkers with high reproducibility and accurate prediction performance can contribute to comprehending the underlying pathogenesis of related complex diseases and further facilitate disease diagnosis and therapy. Techniques integrating gene expression profiles and biological networks for the identification of network-based disease biomarkers are receiving increasing interest. The biomarkers for heterogeneous diseases often exhibit strong cooperative effects, which implies that a set of genes may achieve more accurate outcome prediction than any single gene. In this study, we evaluated various biomarker identification methods that consider gene cooperative effects implicitly or explicitly, and proposed the gene cooperation network to explicitly model the cooperative effects of gene combinations. The gene cooperation network-enhanced method, named as MarkRank, achieves superior performance compared with traditional biomarker identification methods in both simulation studies and real data sets. The biomarkers identified by MarkRank not only have a better prediction accuracy but also have stronger topological relationships in the biological network and exhibit high specificity associated with the related diseases. Furthermore, the top genes identified by MarkRank involve crucial biological processes of related diseases and give a good prioritization for known disease genes. In conclusion, MarkRank suggests that explicit modeling of gene cooperative effects can greatly improve biomarker identification for complex diseases, especially for diseases with high heterogeneity.


Asunto(s)
Redes Reguladoras de Genes , Marcadores Genéticos , Herencia Multifactorial , Algoritmos , Biomarcadores de Tumor/genética , Biología Computacional , Simulación por Computador , Bases de Datos Genéticas/estadística & datos numéricos , Perfilación de la Expresión Génica/estadística & datos numéricos , Humanos , Modelos Genéticos , Modelos Estadísticos , Neoplasias/genética , Programas Informáticos , Biología de Sistemas
9.
Cell Commun Signal ; 19(1): 7, 2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33441142

RESUMEN

The cytokine release syndrome or cytokine storm, which is the hyper-induction of inflammatory responses has a central role in the mortality rate of COVID-19 and some other viral infections. Interleukin-6 (IL-6) is a key player in the development of cytokine storms. Shedding of interleukin-6 receptor (IL-6Rα) results in the accumulation of soluble interleukin-6 receptors (sIL-6R). Only relatively few cells express membrane-bound IL-6Rα. However, sIL-6R can act on potentially all cells and organs through the ubiquitously expressed gp130, the coreceptor of IL-6Rα. Through this, so-called trans-signaling, IL-6-sIL-6R is a powerful factor in the development of cytokine storms and multiorgan involvement. Some bacteria (e.g., Serratia marcescens, Staphylococcus aureus, Pseudomonas aeruginosa, Listeria monocytogenes), commonly considered to cause co-infections during viral pneumonia, can directly induce the shedding of membrane receptors, including IL-6Rα, or enhance endogenous shedding mechanisms causing the increase of sIL-6R level. Here we hypothesise that bacteria promoting shedding and increase the sIL-6R level can be an important contributing factor for the development of cytokine storms. Therefore, inhibition of IL-6Rα shedding by drastically reducing the number of relevant bacteria may be a critical element in reducing the chance of a cytokine storm. Validation of this hypothesis can support the consideration of the prophylactic use of antibiotics more widely and at an earlier stage of infection to decrease the mortality rate of COVID-19. Video abstract.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/metabolismo , COVID-19/patología , Síndrome de Liberación de Citoquinas/etiología , Metaloproteasas/metabolismo , COVID-19/complicaciones , COVID-19/virología , Síndrome de Liberación de Citoquinas/microbiología , Humanos , Interleucina-6/metabolismo , Receptores de Interleucina-6/metabolismo , SARS-CoV-2/aislamiento & purificación , Transducción de Señal
10.
Gut ; 69(8): 1520-1532, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32111636

RESUMEN

IBD is a complex multifactorial inflammatory disease of the gut driven by extrinsic and intrinsic factors, including host genetics, the immune system, environmental factors and the gut microbiome. Technological advancements such as next-generation sequencing, high-throughput omics data generation and molecular networks have catalysed IBD research. The advent of artificial intelligence, in particular, machine learning, and systems biology has opened the avenue for the efficient integration and interpretation of big datasets for discovering clinically translatable knowledge. In this narrative review, we discuss how big data integration and machine learning have been applied to translational IBD research. Approaches such as machine learning may enable patient stratification, prediction of disease progression and therapy responses for fine-tuning treatment options with positive impacts on cost, health and safety. We also outline the challenges and opportunities presented by machine learning and big data in clinical IBD research.


Asunto(s)
Macrodatos , Genómica , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Aprendizaje Automático , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Humanos , Interpretación de Imagen Asistida por Computador , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Metagenómica , Medicina de Precisión , Pronóstico , Proteómica , Medición de Riesgo , Investigación Biomédica Traslacional
11.
FASEB J ; 33(2): 2372-2387, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30277819

RESUMEN

NF-E2-related factor 2 (NRF2) transcription factor has a fundamental role in cell homeostasis maintenance as one of the master regulators of oxidative and electrophilic stress responses. Previous studies have shown that a regulatory connection exists between NRF2 and autophagy during reactive oxygen species-generated oxidative stress. The aim of the present study was to investigate how autophagy is turned off during prolonged oxidative stress, to avoid overeating and destruction of essential cellular components. AMPK is a key cellular energy sensor highly conserved in eukaryotic organisms, and it has an essential role in autophagy activation at various stress events. Here the role of human AMPK and its Caenorhabditis elegans counterpart AAK-2 was explored upon oxidative stress. We investigated the regulatory connection between NRF2 and AMPK during oxidative stress induced by tert-butyl hydroperoxide (TBHP) in HEK293T cells and C. elegans. Putative conserved NRF2/protein skinhead-1 binding sites were found in AMPK/aak-2 genes by in silico analysis and were later confirmed experimentally by using EMSA. After addition of TBHP, NRF2 and AMPK showed a quick activation; AMPK was later down-regulated, however, while NRF2 level remained high. Autophagosome formation and Unc-51-like autophagy activating kinase 1 phosphorylation were initially stimulated, but they returned to basal values after 4 h of TBHP treatment. The silencing of NRF2 resulted in a constant activation of AMPK leading to hyperactivation of autophagy during oxidative stress. We observed the same effects in C. elegans demonstrating the conservation of this self-defense mechanism to save cells from hyperactivated autophagy upon prolonged oxidative stress. We conclude that NRF2 negatively regulates autophagy through delayed down-regulation of the expression of AMPK upon prolonged oxidative stress. This regulatory connection between NRF2 and AMPK may have an important role in understanding how autophagy is regulated in chronic human morbidities characterized by oxidative stress, such as neurodegenerative diseases, certain cancer types, and in metabolic diseases.-Kosztelnik, M., Kurucz, A., Papp, D., Jones, E., Sigmond, T., Barna, J., Traka, M. H., Lorincz, T., Szarka, A., Banhegyi, G., Vellai, T., Korcsmaros, T., Kapuy, O. Suppression of AMPK/aak-2 by NRF2/SKN-1 down-regulates autophagy during prolonged oxidative stress.


Asunto(s)
Autofagia , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Células HEK293 , Humanos , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética
12.
Semin Cell Dev Biol ; 58: 55-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27395026

RESUMEN

Cancer initiation and development are increasingly perceived as systems-level phenomena, where intra- and inter-cellular signaling networks of the ecosystem of cancer and stromal cells offer efficient methodologies for outcome prediction and intervention design. Within this framework, RAS emerges as a 'contextual signaling hub', i.e. the final result of RAS activation or inhibition is determined by the signaling network context. Current therapies often 'train' cancer cells shifting them to a novel attractor, which has increased metastatic potential and drug resistance. The few therapy-surviving cancer cells are surrounded by massive cell death triggering a primordial adaptive and reparative general wound healing response. Overall, dynamic analysis of patient- and disease-stage specific intracellular and intercellular signaling networks may open new areas of anticancer therapy using multitarget drugs, drugs combinations, edgetic drugs, as well as help design 'gentler', differentiation and maintenance therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinogénesis/metabolismo , Carcinogénesis/patología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Transducción de Señal , Proteínas ras/metabolismo , Animales , Antineoplásicos/farmacología , Carcinogénesis/efectos de los fármacos , Humanos , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Semin Cell Dev Biol ; 58: 79-85, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27058752

RESUMEN

Why are YAP1 and c-Myc often overexpressed (or activated) in KRAS-driven cancers and drug resistance? Here, we propose that there are two independent pathways in tumor proliferation: one includes MAPK/ERK and PI3K/A kt/mTOR; and the other consists of pathways leading to the expression (or activation) of YAP1 and c-Myc. KRAS contributes through the first. MYC is regulated by e.g. ß-catenin, Notch and Hedgehog. We propose that YAP1 and ERK accomplish similar roles in cell cycle control, as do ß-catenin and PI3K. This point is compelling, since the question of how YAP1 rescues K-Ras or B-Raf ablation has recently captured much attention, as well as the mechanism of resistance to PI3K inhibitors. The similarity in cell cycle actions of ß-catenin and PI3K can also clarify the increased aggressiveness of lung cancer when both K-Ras and ß-catenin operate. Thus, we propose that the two pathways can substitute one another - or together amplify each other - in promoting proliferation. This new understanding of the independence and correspondence of the two pathways in cancer - MAPK/ERK and PI3K/Akt/mTOR; and YAP1 and c-Myc - provide a coherent and significant picture of signaling-driven oncogenic proliferation and may help in judicious, pathway-based drug discovery.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ciclo Celular , Transducción de Señal , beta Catenina/metabolismo , Proteínas ras/metabolismo , Animales , Humanos
14.
Nucleic Acids Res ; 43(Database issue): D485-93, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25348397

RESUMEN

Here we present ComPPI, a cellular compartment-specific database of proteins and their interactions enabling an extensive, compartmentalized protein-protein interaction network analysis (URL: http://ComPPI.LinkGroup.hu). ComPPI enables the user to filter biologically unlikely interactions, where the two interacting proteins have no common subcellular localizations and to predict novel properties, such as compartment-specific biological functions. ComPPI is an integrated database covering four species (S. cerevisiae, C. elegans, D. melanogaster and H. sapiens). The compilation of nine protein-protein interaction and eight subcellular localization data sets had four curation steps including a manually built, comprehensive hierarchical structure of >1600 subcellular localizations. ComPPI provides confidence scores for protein subcellular localizations and protein-protein interactions. ComPPI has user-friendly search options for individual proteins giving their subcellular localization, their interactions and the likelihood of their interactions considering the subcellular localization of their interacting partners. Download options of search results, whole-proteomes, organelle-specific interactomes and subcellular localization data are available on its website. Due to its novel features, ComPPI is useful for the analysis of experimental results in biochemistry and molecular biology, as well as for proteome-wide studies in bioinformatics and network science helping cellular biology, medicine and drug design.


Asunto(s)
Bases de Datos de Proteínas , Mapeo de Interacción de Proteínas , Animales , Compartimento Celular , Humanos , Internet , Proteínas/análisis , Proteínas/metabolismo
15.
Semin Cancer Biol ; 30: 42-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24412105

RESUMEN

Cancer is increasingly perceived as a systems-level, network phenomenon. The major trend of malignant transformation can be described as a two-phase process, where an initial increase of network plasticity is followed by a decrease of plasticity at late stages of tumor development. The fluctuating intensity of stress factors, like hypoxia, inflammation and the either cooperative or hostile interactions of tumor inter-cellular networks, all increase the adaptation potential of cancer cells. This may lead to the bypass of cellular senescence, and to the development of cancer stem cells. We propose that the central tenet of cancer stem cell definition lies exactly in the indefinability of cancer stem cells. Actual properties of cancer stem cells depend on the individual "stress-history" of the given tumor. Cancer stem cells are characterized by an extremely large evolvability (i.e. a capacity to generate heritable phenotypic variation), which corresponds well with the defining hallmarks of cancer stem cells: the possession of the capacity to self-renew and to repeatedly re-build the heterogeneous lineages of cancer cells that comprise a tumor in new environments. Cancer stem cells represent a cell population, which is adapted to adapt. We argue that the high evolvability of cancer stem cells is helped by their repeated transitions between plastic (proliferative, symmetrically dividing) and rigid (quiescent, asymmetrically dividing, often more invasive) phenotypes having plastic and rigid networks. Thus, cancer stem cells reverse and replay cancer development multiple times. We describe network models potentially explaining cancer stem cell-like behavior. Finally, we propose novel strategies including combination therapies and multi-target drugs to overcome the Nietzschean dilemma of cancer stem cell targeting: "what does not kill me makes me stronger".


Asunto(s)
Hipoxia de la Célula/fisiología , Transformación Celular Neoplásica/patología , Senescencia Celular/fisiología , Inflamación/patología , Células Madre Neoplásicas/patología , Humanos
16.
Brief Bioinform ; 14(5): 618-32, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23640570

RESUMEN

The number of bioinformatics tools and resources that support molecular and cell biology approaches is continuously expanding. Moreover, systems and network biology analyses are accompanied more and more by integrated bioinformatics methods. Traditional information-centered university teaching methods often fail, as (1) it is impossible to cover all existing approaches in the frame of a single course, and (2) a large segment of the current bioinformation can become obsolete in a few years. Signaling network offers an excellent example for teaching bioinformatics resources and tools, as it is both focused and complex at the same time. Here, we present an outline of a university bioinformatics course with four sample practices to demonstrate how signaling network studies can integrate biochemistry, genetics, cell biology and network sciences. We show that several bioinformatics resources and tools, as well as important concepts and current trends, can also be integrated to signaling network studies. The research-type hands-on experiences we show enable the students to improve key competences such as teamworking, creative and critical thinking and problem solving. Our classroom course curriculum can be re-formulated as an e-learning material or applied as a part of a specific training course. The multi-disciplinary approach and the mosaic setup of the course have the additional benefit to support the advanced teaching of talented students.


Asunto(s)
Biología Computacional/educación , Biología Celular/educación , Curriculum , Comunicación Interdisciplinaria , Aprendizaje , Biología Molecular/educación , Transducción de Señal , Biología de Sistemas/educación , Universidades
17.
Semin Cancer Biol ; 23(4): 209-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23831276

RESUMEN

Cancer is increasingly described as a systems-level, network phenomenon. Genetic methods, such as next generation sequencing and RNA interference uncovered the complexity tumor-specific mutation-induced effects and the identification of multiple target sets. Network analysis of cancer-specific metabolic and signaling pathways highlighted the structural features of cancer-related proteins and their complexes to develop next-generation protein kinase inhibitors, as well as the modulation of inflammatory and autophagic pathways in anti-cancer therapies. Importantly, malignant transformation can be described as a two-phase process, where an initial increase of system plasticity is followed by a decrease of plasticity at late stages of tumor development. Late-stage tumors should be attacked by an indirect network influence strategy. On the contrary, the attack of early-stage tumors may target central network nodes. Cancer stem cells need special diagnosis and targeting, since they potentially have an extremely high ability to change the rigidity/plasticity of their networks. The early warning signals of the activation of fast growing tumor cell clones are important in personalized diagnosis and therapy. Multi-target attacks are needed to perturb cancer-specific networks to exit from cancer attractors and re-enter a normal attractor. However, the dynamic non-genetic heterogeneity of cancer cell population induces the replenishment of the cancer attractor with surviving, non-responsive cells from neighboring abnormal attractors. The development of drug resistance is further complicated by interactions of tumor clones and their microenvironment. Network analysis of intercellular cooperation using game theory approaches may open new areas of understanding tumor complexity. In conclusion, the above applications of the network approach open up new, and highly promising avenues in anti-cancer drug design.


Asunto(s)
Autofagia/fisiología , Transformación Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Transducción de Señal/fisiología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Autofagia/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Biología de Sistemas/métodos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
18.
Semin Cancer Biol ; 23(4): 262-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23796463

RESUMEN

There is a widening recognition that cancer cells are products of complex developmental processes. Carcinogenesis and metastasis formation are increasingly described as systems-level, network phenomena. Here we propose that malignant transformation is a two-phase process, where an initial increase of system plasticity is followed by a decrease of plasticity at late stages of carcinogenesis as a model of cellular learning. We describe the hallmarks of increased system plasticity of early, tumor initiating cells, such as increased noise, entropy, conformational and phenotypic plasticity, physical deformability, cell heterogeneity and network rearrangements. Finally, we argue that the large structural changes of molecular networks during cancer development necessitate a rather different targeting strategy in early and late phase of carcinogenesis. Plastic networks of early phase cancer development need a central hit, while rigid networks of late stage primary tumors or established metastases should be attacked by the network influence strategy, such as by edgetic, multi-target, or allo-network drugs. Cancer stem cells need special diagnosis and targeting, since their dormant and rapidly proliferating forms may have more rigid, or more plastic networks, respectively. The extremely high ability of cancer stem cells to change the rigidity/plasticity of their networks may be their key hallmark. The application of early stage-optimized anti-cancer drugs to late-stage patients may be a reason of many failures in anti-cancer therapies. Our hypotheses presented here underlie the need for patient-specific multi-target therapies applying the correct ratio of central hits and network influences - in an optimized sequence.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología
19.
Semin Cancer Biol ; 23(4): 252-61, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23810837

RESUMEN

Autophagy, a highly regulated self-degradation process of eukaryotic cells, is a context-dependent tumor-suppressing mechanism that can also promote tumor cell survival upon stress and treatment resistance. Because of this ambiguity, autophagy is considered as a double-edged sword in oncology, making anti-cancer therapeutic approaches highly challenging. In this review, we present how systems-level knowledge on autophagy regulation can help to develop new strategies and efficiently select novel anti-cancer drug targets. We focus on the protein interactors and transcriptional/post-transcriptional regulators of autophagy as the protein and regulatory networks significantly influence the activity of core autophagy proteins during tumor progression. We list several network resources to identify interactors and regulators of autophagy proteins. As in silico analysis of such networks often necessitates experimental validation, we briefly summarize tractable model organisms to examine the role of autophagy in cancer. We also discuss fluorescence techniques for high-throughput monitoring of autophagy in humans. Finally, the challenges of pharmacological modulation of autophagy are reviewed. We suggest network-based concepts to overcome these difficulties. We point out that a context-dependent modulation of autophagy would be favored in anti-cancer therapy, where autophagy is stimulated in normal cells, while inhibited only in stressed cancer cells. To achieve this goal, we introduce the concept of regulo-network drugs targeting specific transcription factors or miRNA families identified with network analysis. The effect of regulo-network drugs propagates indirectly through transcriptional or post-transcriptional regulation of autophagy proteins, and, as a multi-directional intervention tool, they can both activate and inhibit specific proteins in the same time. The future identification and validation of such regulo-network drug targets may serve as novel intervention points, where autophagy can be effectively modulated in cancer therapy.


Asunto(s)
Autofagia/fisiología , Neoplasias/metabolismo , Mapas de Interacción de Proteínas/fisiología , Transducción de Señal/fisiología , Animales , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Autofagia/genética , Modelos Animales de Enfermedad , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA