Skip to main content
Frontiers in Genetics logoLink to Frontiers in Genetics
. 2014 Mar 3;5:49. doi: 10.3389/fgene.2014.00049

Signaling pathway cloud regulation for in silico screening and ranking of the potential geroprotective drugs

Alex Zhavoronkov 1,2,3,4,*, Anton A Buzdin 1,2,4,5, Andrey V Garazha 1,2,4,5, Nikolay M Borisov 1,2,6, Alexey A Moskalev 1,7,8,*
PMCID: PMC3940060  PMID: 24624136

Abstract

The major challenges of aging research include absence of the comprehensive set of aging biomarkers, the time it takes to evaluate the effects of various interventions on longevity in humans and the difficulty extrapolating the results from model organisms to humans. To address these challenges we propose the in silico method for screening and ranking the possible geroprotectors followed by the high-throughput in vivo and in vitro validation. The proposed method evaluates the changes in the collection of activated or suppressed signaling pathways involved in aging and longevity, termed signaling pathway cloud, constructed using the gene expression data and epigenetic profiles of young and old patients' tissues. The possible interventions are selected and rated according to their ability to regulate age-related changes and minimize differences in the signaling pathway cloud. While many algorithmic solutions to simulating the induction of the old into young metabolic profiles in silico are possible, this flexible and scalable approach may potentially be used to predict the efficacy of the many drugs that may extend human longevity before conducting pre-clinical work and expensive clinical trials.

Keywords: geroprotector, aging-suppressive drug, signaling pathway cloud, validation of drugs, transcriptomics


The increasing burden of the aging on the economies of the developed countries is turning the quest to increase healthy life spans from an altruistic cause into a pressing economic priority required to maintain the current standards of living and facilitate economic growth (Zhavoronkov and Litovchenko, 2013). There is an urgent need to develop and validate interventions with geroprotective properties to increase the productive health spans of the working population and maintaining performance and avoiding loss of function (Kennedy, 2012).

While no doubt exists that aging is a complex multifactorial process with no single cause or treatment (Zhavoronkov and Cantor, 2011; Trindade et al., 2013), the issue whether aging can be classified as the disease is widely debated (Rattan, 2013). However, many strategies for extending organismal life spans have been proposed including replacing cells (Rodgerson and Harris, 2011) and organs, comprehensive strategies for repairing the accumulated damage, using hormetins to activate endogenous repair processes (Gems and Partridge, 2008; Gaman et al., 2011), modulating the aging processes through specific mutations, gene therapy (Bernardes De Jesus et al., 2012) and small molecule drugs (Kennedy and Pennypacker, 2013). An animal's survival strongly depends on its ability to maintain homeostasis and achieved through intracellular and intercellular communication within and among different tissues (Alcedo et al., 2013). Many strategies for the development and validation of drugs with geroprotective properties have been proposed to help maintain the homeostasis including drugs that act on specific targets or combinations of molecular pathways (Moskalev and Shaposhnikov, 2010, 2011; Zhavoronkov et al., 2012; Danilov et al., 2013) and epigenetic drugs (Vaiserman and Pasyukova, 2012). However, none of the proposed strategies for aging-suppressive drug development provide a roadmap for rapid screening, validation, and clinical deployment. No methods currently exist to predict the effects of currently available drugs on human longevity and health span in a timely manner. This is partly due to the absence of the clear panel of human processes involved in aging to effectively run clinical trials.

Many processes are involved in the aging of cells and organisms including telomere length (Lehmann et al., 2013), intracellular and extracellular aggregates, racemization of the amino acids and genetic instability. Both gene expression (Wolters and Schumacher, 2013) and DNA methylation profiles (Horvath et al., 2012; Horvath, 2013; Mendelsohn and Larrick, 2013) change during aging and may be used as biomarkers of aging. Many studies analyzing transcriptomes of biopsies in a variety of diseases indicated that age and sex of the patient had significant effects on gene expression (Chowers et al., 2003) and that there are noticeable changes in gene expression with age in mice (Weindruch et al., 2002; Park et al., 2009) resulting in development of mouse aging gene expression databases (Zahn et al., 2007) and in humans (Blalock et al., 2003; Welle et al., 2003; Park and Prolla, 2005; Hong et al., 2008; De Magalhaes et al., 2009).

Combination of protein-protein interaction and gene expression in both flies and humans demonstrated that aging is mainly associated with a small number of biological processes, might preferentially attack key regulatory nodes that are important for network stability (Xue et al., 2007).

Our prior work with gene expression and epigenetics of various solid tumors (Kuzmin et al., 2010; Mityaev et al., 2010; Zabolotneva et al., 2012a,b) using the OncoFinder system (www.oncofinder.com), provided clues that transcription profiles of cancer cells mapped onto the signaling pathways may be used to screen for and rate the targeted drugs that regulate pathways directly and indirectly related to aging and longevity. Instead of focusing on individual network elements, this approach involves creating the signaling pathway cloud, a collection of signaling pathways involved in aging and longevity each comprised of multiple network elements and evaluating the individual pathway activation strength. Despite significant advances in aging research, the knowledge of the aging processes is still poor, and combining all available factors involved in cellular aging, aging of the organisms, age-related diseases, stress-resistance, and stress-response along the many other factors into a comprehensive signaling pathway cloud may be more beneficial than focusing on the narrow collection of elements. The creation of the pathway cloud may allow for the annotated databases of molecules and other factors to be screened for effectiveness of individual compounds in replicating the “young” signaling activation profiles in silico.

Several new methods evaluating the robustness and response ability of the gene regulatory network have been developed and applied to gene expression data sets from young and old patients (Tu and Chen, 2013). Prior studies suggested that a combination of pathways, termed pathway cloud, instead of one element of the pathway or the whole pathway might be responsible for pathological changes in the cell (Voronkov and Krauss, 2013). Long-lived species like the sea urchin (Strongylocentrotus franciscanus) and naked mole rat (Heterocephalus glaber) that senesce at a slower rate than members of the same order show less transcriptome changes with age (Kim et al., 2011; Loram and Bodnar, 2012). Gene network analysis using gene expression data was effective in identifying the possible drug targets (Imoto et al., 2003; Savoie et al., 2003). In silico drug discovery algorithms that attempt to transform the metabolism to the healthy state have been proposed and validated (Yizhak et al., 2013). We theorize that in order to be effective, the geroprotector or a combination of aging-suppressive drugs must regulate the pathway cloud in a way that minimizes the difference in the net differences in pathway cloud activation or downregulation between samples of young and old patients. Small molecules and other factors that may influence gene expression may be ranked by their ability to minimize the net difference between the pathway activation profiles of young and old cells. The algorithms for calculating the ability of the potential geroprotector to minimize signaling disturbance may be parametric and account for the effects on specific targets within signaling pathways or machine learned.

Despite the differences in life span and aging phenotypes, many molecular mechanisms of aging are common in all eukaryotes. Pathway analysis revealed that there are many common age-related transcriptomic changes between different species, including yeasts, worms, flies, rodents, and human (Murphy et al., 2006). Hypothetically, the human orthologs of aging-related genes of model organisms are also involved in aging process. To select longevity-associated pathways for future analysis we perform the following procedure. Using (Tacutu et al., 2013) database we selected genes, where knockout, loss-of-function mutation, deletion or RNA interference significantly extended lifespan in several model organisms (yeasts Schizosaccharomyces pombe and Saccharomyces cerevisiae, nematode Caenorhabditis elegans, fruitfly Drosophila melanogaster and mouse Mus musculus) from 10 to 200%. We converted the obtained gene lists from different models to the general list of human orthologs where it is possible. 226 genes of 315 from our set were subjected to over-representation pathway analysis in (Kamburov et al., 2011). P-value and corrected by FDR P-value are calculated according to the hypergeometric test based on the number of physical entities present in both the predefined set of genes to each (Kanehisa et al., 2004) pathway and our list of aging-associated genes (Table 1). We established limits of 2 genes of minimum overlap with input list and 0.01 p-value cut off threshold.

Table 1.

KEGG pathways and list of aging-associated genes.

KEGG pathway name Background quantity of genes in the pathway Overlap with candidates list % p-Value FDR
Citrate cycle (TCA cycle) 30 5 16.7 0.000401 0.0037
Ribosome 136 21 15.7 7.52E-13 6.21E-11
Parkinson‘s disease 131 20 15.4 3.87E-12 2.02E-10
Aldosterone-regulated sodium reabsorption 39 6 15.4 0.000165 0.00199
Type II diabetes mellitus 48 7 14.6 6.66E-05 0.00105
Oxidative phosphorylation 133 19 14.4 4.55E-11 1.78E-09
mTOR signaling pathway 60 8 13.3 3.89E-05 0.000764
Huntington‘s disease 183 24 13.2 7.92E-13 6.21E-11
Progesterone-mediated oocyte maturation 86 11 12.8 2.08E-06 4.66E-05
Insulin signaling pathway 142 17 12.1 8.01E-09 2.10E-07
Ovarian steroidogenesis 51 6 11.8 0.000735 0.00525
Long-term depression 60 7 11.7 0.000281 0.00294
Amyotrophic lateral sclerosis (ALS) 53 6 11.3 0.000905 0.00592
Alzheimer‘s disease 170 19 11.2 3.36E-09 1.05E-07
Cardiac muscle contraction 77 8 10.5 0.000214 0.0024
Gap junction 89 9 10.1 0.000118 0.00155
Prostate cancer 89 9 10.1 0.000118 0.00155
Estrogen signaling pathway 100 10 10.0 5.44E-05 0.00095
Colorectal cancer 62 6 9.7 0.00206 0.0101
Glioma 65 6 9.2 0.00263 0.012
Pancreatic cancer 66 6 9.1 0.00284 0.0124
GABAergic synapse 90 8 9.0 0.000631 0.00524
Adipocytokine signaling pathway 71 6 8.6 0.00382 0.0154
PPAR signaling pathway 71 6 8.5 0.0041 0.0157
Circadian entrainment 97 8 8.3 0.00104 0.00629
Prolactin signaling pathway 72 6 8.3 0.0044 0.0157
Chronic myeloid leukemia 73 6 8.2 0.0047 0.0164
Cholinergic synapse 113 9 8.0 0.000667 0.00524
Oocyte meiosis 112 9 8.0 0.000667 0.00524
Serotonergic synapse 114 9 8.0 0.000711 0.00525
Insulin secretion 87 7 8.0 0.00261 0.012
Gastric acid secretion 75 6 8.0 0.00537 0.0183
HIF-1 signaling pathway 106 8 7.5 0.00198 0.01
Peroxisome 81 6 7.5 0.00734 0.0226
TGF-beta signaling pathway 80 6 7.5 0.00734 0.0226
Cell cycle 124 9 7.3 0.00138 0.00803
Dopaminergic synapse 131 9 6.9 0.00192 0.01
Glutamatergic synapse 118 8 6.8 0.00366 0.0151
Proteoglycans in cancer 225 14 6.2 0.000348 0.00342
RNA transport 165 10 6.1 0.00268 0.012
Hepatitis B 147 9 6.1 0.00439 0.0157
HTLV-I infection 267 14 5.3 0.00161 0.00869
Chemokine signaling pathway 192 10 5.3 0.00732 0.0226
PI3K-Akt signaling pathway 347 16 4.6 0.00312 0.0133

As a result we revealed overrepresented cell signaling pathways (mTOR, insulin/IGF-1, PI3K-Akt, PPAR, HIF-1, TGF-beta, chemokines, adipocytokine, prolactin, estrogen), general metabolism (TCA cycle, ribosome, oxidative phosphorylation), RNA transport, cell cycle and meiosis, gap junction, peroxisome, cyrcadian rhythm, different synapse types (dopaminergic, glutamatergic, cholinergic, serotonergic, GABAergic), gastric acid secretion as well as age-related diseases pathways (Parkinson's disease, type II diabetes mellitus, Huntington's disease, long-term depression, amyotrophic lateral sclerosis, Alzheimer's disease), Hepatitis B, HTLV-I infection and cancer pathways (prostate cancer, colorectal cancer, glioma, pancreatic cancer, chronic myeloid leukemia, proteoglycans in cancer). We considered obtained such a way pathways as probably associated with the human longevity. Human genes known as key activators/repressors of these pathways may be used in provided further mathematical model.

The methods that may be applied for the possible analysis of geroprotector efficiency by pathways regulation have been arisen from our research experience of cell signaling pathways. As far as we have seen before (Kiyatkin et al., 2006; Borisov et al., 2009; Kuzmina and Borisov, 2011), most signal transduction proteins are essentially far from saturation even at the peak concentrations of the activated form in comparison with the total protein abundances. Thus, we can consider that all activator/repressor genes/proteins have equal importance for the pathway activation/downregulation, and then arrive at the following assessment function for the overall signal pathway cloud disturbance outcome (SPCD) is proportional to the following estimator function,

SPCD=i=1N[AGEL]ij=1M[RGEL]j

Here the multiplication is done over all possible activator and repressor proteins in the pathway, and [AGEL]i and [RGEL]j are gene expression levels of an activator i and repressor j, respectively. To obtain an additive rather than multiplicative value, it is enough just turn from the absolute values of the expression levels to their logarithms, arriving at the pathway activation strength (PAS) value for each pathway (see Figure 1). To obtain the values of Old (case)-to-Young ratio, YORn, one just has to divide the expression levels for a gene n in the sample taken for the senescent person by the same average value for the normalized young group. The discrete value of ARR (activator/repressor role) equals to the following numbers:

Figure 1.

Figure 1

Gene expression-based approach to in silico screening for drugs with geroprotective properties and estimating the predicted efficacy. (A) Using signaling pathway cloud regulation for theoretical in silico aging-suppressive drug identification and ranking. The proposed method for identifying and ranking of geroprotective drugs by evaluating the net effect on the many elements of signaling pathway cloud that brings the “old” metabolic state closer to the “young.” (B) An example of how multiple pathways are activated and down-regulated during aging. (C) Pathway Activation Strength (PAS) is the logarithmic additive value that characterizes the up-/downregulation of signaling pathways. (D) Function for the overall signal pathway cloud disturbance outcome (SPCD).

−1, when the gene/protein n is a repressor of pathway excitation;

1, if the gene/protein n is an activator of pathway excitation;

0, when the gene/protein n can be both an activator and a repressor of signal transduction;

0.5 and −0.5, respectively, if the gene/protein n is more an activator or repressor of the signaling pathway p.

The information about the activator/repressor role of a particular gene/protein may be obtained from the analysis of open-access or customized pathway databases and from the literature.

The Boolean flag of BTIF (beyond tolerance interval flag) equals to zero when the OYR value lies within the tolerance limit, and to one when otherwise. During the current study, we have admitted that the OYR lies beyond the tolerance limit if it satisfies simultaneously the two criteria. First, it either higher than 3/2 or lower than 2/3, and, second, the expression level for a corresponding gene from an old patient of an individual patient differs by more than two standard deviations from the average expression level for the same gene from a set of analogous young tissue/organ samples.

We propose a new computational approach for identifying and rating the variety of factors including small molecules, peptides, stress factors and conditions with the known effects on the transcriptomes at different ages of one or more cell or tissue types or known targets (Figure 1A). The approach may be used for general geroprotector screening, but after the validation of the algorithms in vivo and in vitro may be expanded to identify and predict the efficacy of personalized aging-suppressive intervention regimens for individual patients based on the transcriptome information from various tissue biopsies and blood samples.

The generic geroprotector rating approach involves collecting the transcriptome data sets from young and old patients and normalizing the data for each cell and tissue type, evaluating the pathway activation strength (PAS) for each individual pathway (Figure 1B) and constructing the pathway cloud (PC, Figure 1C) and screen for drugs or combinations that minimize the signaling pathway cloud disturbance (SPCD, Figure 1D) by acting on one or multiple elements of the pathway cloud. Drugs and combinations may be rated by their ability to compensate the changes in signaling pathway activation patterns that are related to aging, thus bringing all the PAS values for the general set of the pathways as close to zero as possible. Since many of the drugs approved for use in humans have known molecular targets and some have been screened for the impact on longevity in model organisms (Ye et al., 2013), the predictions may be then tested both in vitro and in vivo on human cells and on model organisms such as rodents, nematodes and flies to validate the screening and rating algorithms.

Conclusion

Longevity studies of aging-suppressive drug efficiency in higher mammals take several years and decades and may cost millions of dollars. An intelligent process for predicting the activity and ranking the geroprotective activity of various factors and strengthening the prediction in rapid and cost-effective studies on cell cultures and model organisms may help increase the longevity dividend of these studies. In this paper we propose a method for in silico screening and ranking of drugs and other factors that act on many signaling pathways implicated in aging processes by calculating their ability to minimize the difference between signaling pathway activation patterns in cells of young and old patients and confirming the results using in vivo and in vitro studies.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We would like to thank the reviewers for the many useful comments, corrections, and recommendations. We would like to thank Charles R. Cantor, Brian Kennedy, Robert Shmookler-Reis and many others for helpful comments and discussions on strategies for geroprotector screening. We thank UMA Foundation for their help in preparation of this manuscript and Dr. Kristen Swithers from Yale University for her assistance with editing the manuscript. We would like to thank Alex Kim and ASUSTek for equipment and support of this research.

References

  1. Alcedo J., Flatt T., Pasyukova E. G. (2013). Neuronal inputs and outputs of aging and longevity. Front. Genet. 4:71 10.3389/fgene.2013.00071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Bernardes De Jesus B., Vera E., Schneeberger K., Tejera A. M., Ayuso E., Bosch F., et al. (2012). Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol. Med. 4, 691–704 10.1002/emmm.201200245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Blalock E. M., Chen K. C., Sharrow K., Herman J. P., Porter N. M., Foster T. C., et al. (2003). Gene microarrays in hippocampal aging: statistical profiling identifies novel processes correlated with cognitive impairment. J. Neurosci. 23, 3807–3819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Borisov N., Aksamitiene E., Kiyatkin A., Legewie S., Berkhout J., Maiwald T., et al. (2009). Systems-level interactions between insulin-EGF networks amplify mitogenic signaling. Mol. Syst. Biol. 5, 256 10.1038/msb.2009.19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Chowers I., Liu D., Farkas R. H., Gunatilaka T. L., Hackam A. S., Bernstein S. L., et al. (2003). Gene expression variation in the adult human retina. Hum. Mol. Genet. 12, 2881–2893 10.1093/hmg/ddg326 [DOI] [PubMed] [Google Scholar]
  6. Danilov A., Shaposhnikov M., Plyusnina E., Kogan V., Fedichev P., Moskalev A. (2013). Selective anticancer agents suppress aging in Drosophila. Oncotarget 4, 1507–1526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. De Magalhaes J. P., Curado J., Church G. M. (2009). Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics 25, 875–881 10.1093/bioinformatics/btp073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Gaman L., Stoian I., Atanasiu V. (2011). Can ageing be slowed?: Hormetic and redox perspectives. J. Med. Life 4, 346–351 [PMC free article] [PubMed] [Google Scholar]
  9. Gems D., Partridge L. (2008). Stress-response hormesis and aging: “that which does not kill us makes us stronger.” Cell Metab. 7, 200–203 10.1016/j.cmet.2008.01.001 [DOI] [PubMed] [Google Scholar]
  10. Hong M. G., Myers A. J., Magnusson P. K., Prince J. A. (2008). Transcriptome-wide assessment of human brain and lymphocyte senescence. PLoS ONE 3:e3024 10.1371/journal.pone.0003024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Horvath S. (2013). DNA methylation age of human tissues and cell types. Genome Biol. 14:R115 10.1186/gb-2013-14-10-r115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Horvath S., Zhang Y., Langfelder P., Kahn R. S., Boks M. P., Van Eijk K., et al. (2012). Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol. 13:R97 10.1186/gb-2012-13-10-r97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Imoto S., Savoie C. J., Aburatani S., Kim S., Tashiro K., Kuhara S., et al. (2003). Use of gene networks for identifying and validating drug targets. J. Bioinform. Comput. Biol. 1, 459–474 10.1142/S0219720003000290 [DOI] [PubMed] [Google Scholar]
  14. Kamburov A., Pentchev K., Galicka H., Wierling C., Lehrach H., Herwig R. (2011). ConsensusPathDB: toward a more complete picture of cell biology. Nucleic Acids Res. 39, D712–D717 10.1093/nar/gkq1156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Kanehisa M., Goto S., Kawashima S., Okuno Y., Hattori M. (2004). The KEGG resource for deciphering the genome. Nucleic Acids Res. 32, D277–D280 10.1093/nar/gkh063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Kennedy B. (2012). Gerontology: more funding for studies of ageing. Nature 487:39 10.1038/487039a [DOI] [PubMed] [Google Scholar]
  17. Kennedy B. K., Pennypacker J. K. (2013). Drugs that modulate aging: the promising yet difficult path ahead. Transl. Res. [Epub ahead of print]. 10.1016/j.trsl.2013.11.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Kim E. B., Fang X., Fushan A. A., Huang Z., Lobanov A. V., Han L., et al. (2011). Genome sequencing reveals insights into physiology and longevity of the naked mole rat. Nature 479, 223–227 10.1038/nature10533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Kiyatkin A., Aksamitiene E., Markevich N. I., Borisov N. M., Hoek J. B., Kholodenko B. N. (2006). Scaffolding protein Grb2-associated binder 1 sustains epidermal growth factor-induced mitogenic and survival signaling by multiple positive feedback loops. J. Biol. Chem. 281, 19925–19938 10.1074/jbc.M600482200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Kuzmin D., Gogvadze E., Kholodenko R., Grzela D. P., Mityaev M., Vinogradova T., et al. (2010). Novel strong tissue specific promoter for gene expression in human germ cells. BMC Biotechnol. 10:58 10.1186/1472-6750-10-58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Kuzmina N. B., Borisov N. M. (2011). Handling complex rule-based models of mitogenic cell signaling (on the example of ERK Activation upon EGF Stimulation). Int. Proc. Chem. Biol. Environ. Eng. 5, 67–82 [Google Scholar]
  22. Lehmann G., Muradian K. K., Fraifeld V. E. (2013). Telomere length and body temperature-independent determinants of mammalian longevity? Front. Genet. 4:111 10.3389/fgene.2013.00111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Loram J., Bodnar A. (2012). Age-related changes in gene expression in tissues of the sea urchin Strongylocentrotus purpuratus. Mech. Ageing Dev. 133, 338–347 10.1016/j.mad.2012.03.012 [DOI] [PubMed] [Google Scholar]
  24. Mendelsohn A. R., Larrick J. W. (2013). The DNA methylome as a biomarker for epigenetic instability and human aging. Rejuvenation Res. 16, 74–77 10.1089/rej.2013.1414 [DOI] [PubMed] [Google Scholar]
  25. Mityaev M. V., Kopantzev E. P., Buzdin A. A., Vinogradova T. V., Sverdlov E. D. (2010). Enhancer element potentially involved in human survivin gene promoter regulation in lung cancer cell lines. Biochemistry (Mosc.) 75, 182–191 10.1134/S0006297910020082 [DOI] [PubMed] [Google Scholar]
  26. Moskalev A. A., Shaposhnikov M. V. (2010). Pharmacological inhibition of phosphoinositide 3 and TOR kinases improves survival of Drosophila melanogaster. Rejuvenation Res. 13, 246–247 10.1089/rej.2009.0903 [DOI] [PubMed] [Google Scholar]
  27. Moskalev A., Shaposhnikov M. (2011). Pharmacological inhibition of NF-kappaB prolongs lifespan of Drosophila melanogaster. Aging (Albany NY) 3, 391–394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Murphy G. G., Shah V., Hell J. W., Silva A. J. (2006). Investigation of age-related cognitive decline using mice as a model system: neurophysiological correlates. Am. J. Geriatr. Psychiatry 14, 1012–1021 10.1097/01.JGP.0000209404.54310.b3 [DOI] [PubMed] [Google Scholar]
  29. Park S. K., Kim K., Page G. P., Allison D. B., Weindruch R., Prolla T. A. (2009). Gene expression profiling of aging in multiple mouse strains: identification of aging biomarkers and impact of dietary antioxidants. Aging Cell 8, 484–495 10.1111/j.1474-9726.2009.00496.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Park S. K., Prolla T. A. (2005). Gene expression profiling studies of aging in cardiac and skeletal muscles. Cardiovasc. Res. 66, 205–212 10.1016/j.cardiores.2005.01.005 [DOI] [PubMed] [Google Scholar]
  31. Rattan S. (2013). Aging is not a disease: implications for intervention. Aging Dis. 5 [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Rodgerson D. O., Harris A. G. (2011). A comparison of stem cells for therapeutic use. Stem Cell Rev. 7, 782–796 10.1007/s12015-011-9241-y [DOI] [PubMed] [Google Scholar]
  33. Savoie C. J., Aburatani S., Watanabe S., Eguchi Y., Muta S., Imoto S., et al. (2003). Use of gene networks from full genome microarray libraries to identify functionally relevant drug-affected genes and gene regulation cascades. DNA Res. 10, 19–25 10.1093/dnares/10.1.19 [DOI] [PubMed] [Google Scholar]
  34. Tacutu R., Craig T., Budovsky A., Wuttke D., Lehmann G., Taranukha D., et al. (2013). Human ageing genomic resources: integrated databases and tools for the biology and genetics of ageing. Nucleic Acids Res. 41, D1027–D1033 10.1093/nar/gks1155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Trindade L. S., Aigaki T., Peixoto A. A., Balduino A., Manica Da Cruz I. B., Heddle J. G. (2013). A novel classification system for evolutionary aging theories. Front. Genet. 4:25 10.3389/fgene.2013.00025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Tu C.-T., Chen B.-S. (2013). New measurement methods of network robustness and response ability via microarray data. PLoS ONE 8:e55230 10.1371/journal.pone.0055230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Vaiserman A. M., Pasyukova E. G. (2012). Epigenetic drugs: a novel anti-aging strategy? Front. Genet. 3:224 10.3389/fgene.2012.00224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Voronkov A., Krauss S. (2013). Wnt/beta-catenin signaling and small molecule inhibitors. Curr. Pharm. Des. 19, 634–664 10.2174/138161213804581837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Weindruch R., Kayo T., Lee C.-K., Prolla T. A. (2002). Gene expression profiling of aging using DNA microarrays. Mech. Ageing Dev. 123, 177–193 10.1016/S0047-6374(01)00344-X [DOI] [PubMed] [Google Scholar]
  40. Welle S., Brooks A. I., Delehanty J. M., Needler N., Thornton C. A. (2003). Gene expression profile of aging in human muscle. Physiol. Genomics 14, 149–159 10.1152/physiolgenomics.00049.2003 [DOI] [PubMed] [Google Scholar]
  41. Wolters S., Schumacher B. (2013). Genome maintenance and transcription integrity in aging and disease. Front. Genet. 4:19 10.3389/fgene.2013.00019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Xue H., Xian B., Dong D., Xia K., Zhu S., Zhang Z., et al. (2007). A modular network model of aging. Mol. Syst. Biol. 3, 147 10.1038/msb4100189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ye X., Linton J. M., Schork N. J., Buck L. B., Petrascheck M. (2013). A pharmacological network for lifespan extension in Caenorhabditis elegans. Aging Cell. [Epub ahead of print]. 10.1111/acel.12163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Yizhak K., Gabay O., Cohen H., Ruppin E. (2013). Model-based identification of drug targets that revert disrupted metabolism and its application to ageing. Nat. Commun. 4:2632 10.1038/ncomms3632 [DOI] [PubMed] [Google Scholar]
  45. Zabolotneva A. A., Bantysh O., Suntsova M. V., Efimova N., Malakhova G. V., Schumann G. G., et al. (2012a). Transcriptional regulation of human-specific SVAF(1) retrotransposons by cis-regulatory MAST2 sequences. Gene 505, 128–136 10.1016/j.gene.2012.05.016 [DOI] [PubMed] [Google Scholar]
  46. Zabolotneva A. A., Zhavoronkov A., Garazha A. V., Roumiantsev S. A., Buzdin A. A. (2012b). Characteristic patterns of microRNA expression in human bladder cancer. Front. Genet. 3:310 10.3389/fgene.2012.00310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Zahn J. M., Poosala S., Owen A. B., Ingram D. K., Lustig A., Carter A., et al. (2007). AGEMAP: a gene expression database for aging in mice. PLoS Genet. 3:e201 10.1371/journal.pgen.0030201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Zhavoronkov A., Cantor C. R. (2011). Methods for structuring scientific knowledge from many areas related to aging research. PLoS ONE 6:e22597 10.1371/journal.pone.0022597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Zhavoronkov A., Litovchenko M. (2013). Biomedical progress rates as new parameters for models of economic growth in developed countries. Int. J. Environ. Res. Public Health 10, 5936–5952 10.3390/ijerph10115936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Zhavoronkov A., Smit-Mcbride Z., Guinan K. J., Litovchenko M., Moskalev A. (2012). Potential therapeutic approaches for modulating expression and accumulation of defective lamin A in laminopathies and age-related diseases. J. Mol. Med. (Berl.) 90, 1361–1389 10.1007/s00109-012-0962-4 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Frontiers in Genetics are provided here courtesy of Frontiers Media SA

RESOURCES