Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Septic Shock in Advanced Age: Transcriptome Analysis Reveals Altered Molecular Signatures in Neutrophil Granulocytes

Abstract

Sepsis is one of the highest causes of mortality in hospitalized people and a common complication in both surgical and clinical patients admitted to hospital for non-infectious reasons. Sepsis is especially common in older people and its incidence is likely to increase substantially as a population ages. Despite its increased prevalence and mortality in older people, immune responses in the elderly during septic shock appear similar to that in younger patients. The purpose of this study was to conduct a genome-wide gene expression analysis of circulating neutrophils from old and young septic patients to better understand how aged individuals respond to severe infectious insult. We detected several genes whose expression could be used to differentiate immune responses of the elderly from those of young people, including genes related to oxidative phosphorylation, mitochondrial dysfunction and TGF-β signaling, among others. Our results identify major molecular pathways that are particularly affected in the elderly during sepsis, which might have a pivotal role in worsening clinical outcomes compared with young people with sepsis.

Introduction

Sepsis is a complex syndrome triggered by infection and characterized by the massive deregulation of immunological networks [1]. Septic patients have a mean age of approximately 65 years [2] and the incidence of sepsis and its risk of mortality increase significantly with advanced age [3, 4]. Factors that might contribute to the increased risk and incidence in the elderly include defects in the integrity of epithelial barriers, decreased gag and cough reflexes, altered levels of consciousness, immobility, concomitant medical diseases, a dependency on invasive medical devices, diminished physiological reserves, endocrine deficiencies, and malnutrition [5, 6].

Age-related defects in immunity are caused by major defects in cell-mediated and humoral effector functions [7]. Aging causes changes in adaptive immunity associated with a shift of the T cell repertoire from a naive phenotype to memory T cells [3] and from type 1 to type 2 responses [8, 9]. Numbers of B and plasma cells gradually decrease with age, while immunoglobulin levels increase [10].

Innate immunity was previously considered to be well-preserved in the elderly [11], but recent studies have also demonstrated significant alterations in these components [3]. Studies have also indicated the altered expression and function of Toll-like receptors (TLRs) caused by aging, altering the host’s response to pathogens [12]. Elevated levels of basal inflammation [13, 14], defective activation of mitogen-activated protein kinases (MAPKs) [15], increased number of apoptotic cells [16], defects in phagocytosis, generation of reactive oxygen species (ROS) and impaired costimulatory molecule expression have also been reported in the elderly [17]. Indeed, recent studies have indicated that older adults have elevated levels of pro-inflammatory cytokines, clotting factors and acute phase reactants in the steady state [1820].

The inflammatory response of the elderly following infection, however, remains under debate. Animal studies demonstrated that mortality, inflammation, hypothermia, apoptosis and disseminated intravascular coagulation are increased in aged animals subjected to experimental models of sepsis [21]. It is intriguing that despite the well characterized aspects of immunosenescence and the exaggerated inflammatory response detected in septic aged rodents [22, 23], clinical studies conducted in humans (including those from our group) observed a similar immune profile when old and young septic critically ill patients were compared [2427].

The purpose of this study was to perform a genome-wide gene expression analysis of neutrophils obtained from elderly and young patients in septic shock, to investigate the potential differences in cell activation that might explain the altered status of immune and inflammatory systems in advanced aged patients with sepsis. We decided to focus on neutrophils because recent reports suggested their function was particularly altered in the elderly [28].

Patients and Methods

Study design

The current study was a prospective cohort study, conducted in the Hospital das Clínicas Intensive Care Units (University of Sao Paulo, Brazil). Blood samples were obtained from six aged septic patients (age range 65–78 years old), six young septic patients (age range 22–35 years old), six healthy aged volunteers (age range 60–82 years old) and six healthy younger individuals (age range 20–35 years old). Patients’ profiles are described in the S1 Table. All cases of sepsis were in patients with a clinical illness and did not include patients admitted for trauma or surgical reasons. Reasons for the majority of admissions included in this study were sepsis, stroke, altered levels of consciousness, pulmonary edema, and asthma and/or chronic obstructive pulmonary disease. Patients who were less than 18 years old, pregnant, HIV-positive, or in end-of-life conditions were excluded. Patients with disseminated malignancies or advanced hepatic disease, those receiving chemotherapy and those who refused to participate in this study were also excluded. Septic shock was defined according to the criteria of the American College of Chest Physicians/Society of Critical Care Medicine (ACCP/SCCM) Consensus Conference Committee proposed in 1992 [29].

The study protocol was approved by the Hospital das Clínicas Ethics Committee. Patients (or their close relatives) received detailed explanations and provided written consent prior to inclusion in the study (Protocol # 1207/09).

RNA extraction and microarray experiments

All blood samples were sent to our laboratory for processing immediately after collection. The anticoagulant-treated blood was layered on the Ficoll-Paque PLUS solution (GE Healthcare) and centrifuged for a short period of time. Differential migration during centrifugation results in the formation of layers containing different cell types. The bottom layer contains erythrocytes that have been aggregated by the Ficoll and, therefore, sediment completely through the Ficoll-Paque PLUS. The layer immediately above the erythrocyte layer contains the granulocytes, which at the osmotic pressure of the Ficoll-Paque PLUS solution attain a density great enough to migrate through the Ficoll-Paque PLUS layer.

After Ficoll-Paque PLUS (GE Healthcare) density gradient centrifugation, we separated the second layer containing the granulocytes. This layer was transferred to new tubes, diluted in lysis buffer and kept on ice for 10 minutes. After centrifugation at 290 x g for 10 minutes at 4°C the pellet was resuspended in lysis buffer and kept on ice for additional 10 minutes. A new centrifugation step was performed at 2500 x g for 2 minutes at room temperature and the samples were washed with phosphate-buffered saline (PBS). Finally, the samples were centrifuged at 1500 x g for 2 minutes at room temperature and the pellet was resuspended in Trizol (Life Technology, Carlsbad, USA) and stored at -80°C.

Total RNA was isolated following the manufacturer’s protocol and its integrity and concentration were assessed using the Agilent 2100 Bioanalyzer and the RNA 6000 Nano Kit (Agilent Technologies, Santa Clara, CA, USA). Expression levels of protein-coding genes and long noncoding RNAs (lncRNA) were evaluated using the SurePrint G3 Human Gene Expression 8x60K v2 Microarray (design ID # 039494) and the Low Input Quick Amp Labeling kit, following a two-color labeling protocol (Agilent Technologies). Cyanine-3 labeled RNA from each sample and cyanine-5 labeled reference RNA (Universal Human Reference RNA, Agilent, cat #740000) were combined and hybridized to the microarrays following the manufacturer’s protocols.

Data processing and filtering

Microarrays were scanned using the SureScan Microarray Scanner (Agilent Technologies) and images were processed using the Feature Extraction Software v12 (Agilent Technologies) for quality control, determination of feature intensities and ratios, and for background correction. The LOWESS procedure was used for data normalization and transformation [30]. We only considered features that were considered “well above background” in at least five (of six) subjects in at least one study group (young adults with sepsis, control young adults, elderly with sepsis, and control elderly). After data filtering and processing, 16,698 probes remained. A Pearson correlation between those filtered features was calculated for each pair of subjects. Overall, we observed a good correlation between any pair of samples (minimum r = 0.88) and a higher correlation between samples from the same group (mean intragroup correlation = 0.96) compared with different groups of samples (mean intergroup correlation = 0.93). Moreover, the samples grouped by disease status and age using unsupervised hierarchical clustering (S1 Fig) further indicating the good quality of the data.

A number of probes (8192) could not be assigned to the known or predicted lncRNA sequences annotated in public databases (Genbank RefSeq, Emsembl). These poorly annotated probes were categorized as “unannotated” and were not analyzed further. The raw and processed microarray datasets are deposited at the Gene Expression Omnibus (accession number GSE67652 associated to platform GPL16699).

Statistical analysis

We performed two sets of differential gene expression analyses. One analysis was to identify genes deregulated in young and elderly sepsis subjects compared with matched healthy controls. A second analysis searched for genes deregulated in septic or healthy elderly subjects compared with matched young controls. In each analysis, a gene was considered differentially expressed when it was identified by a combination of two statistical methods, namely Significance Analysis of Microarrays [31] and rank product [32], using publicly available R packages [33]. To limit the number of false-positives we only considered for further analysis genes identified as differentially expressed with a P-value of p ≤ 0.01 by both methods.

Functional annotation and pathway analysis

Lists of differentially expressed genes were annotated and investigated to identify the enrichment of particular gene categories or pathways using the Ingenuity Pathway Analysis suite (IPA, Qiagen, Redwood City, www.qiagen.com/ingenuity). IPA Upstream Analysis was used to identify putative upstream regulators and to predict whether they were activated or inhibited, given the observed gene expression changes in the experimental dataset, and the expected causal effects, which were compiled from the literature in Ingenuity’s Knowledge Base.

Relative Quantification of gene expression levels using Real Time-PCR

To validate the microarray expression data, selected genes were subjected to quantitative Real Time-PCR using the TaqMan assay system (Life Technologies). Briefly, 100 ng of total RNA was subjected to reverse transcription using the High-Capacity cDNA Reverse Transcription Kit (Life Technologies) following the manufacturer’s protocol. Specific TaqMan Gene Expression Assays (Life Technologies) were used for detecting gene expression levels of TGFB1 (Hs00998133), SRC (Hs1082246), HDAC4 (Hs01041638), CREBBP (Hs00231733), NDUFA4 (Hs00800172), INHBB (Hs00173582), BMP7 (Hs00233476) and SDHC (Hs01698067). PCR was carried out following the protocol recommended for the TaqMan Universal PCR Master Mix (Life Technologies) in an AB7500 thermocycler (Life Technologies). For each sample, the expression data of each candidate gene was normalized to 18S rRNA expression (Eukaryotic 18S rRNA Endogenous Control, Life Technologies). For relative quantification we used the comparative ∆Ct method [34]. The Student’s t-test (p-value < 0.05) was used to determine the statistical significance of expression differences between the sample groups.

Results

The microarray in this study contained 58,717 probes that included protein-coding mRNAs (36,075), long noncoding RNAs (14,450), and a number of poorly annotated transcripts (8192) (Table 1). Approximately 28.4% of all probes were detected in at least one group of samples. A smaller fraction of lncRNAs (10%) was detected compared with protein-coding mRNAs (82%) (Table 1), which is in line with the more tissue-specific and less abundant nature of these transcripts [35, 36].

thumbnail
Table 1. Expressed probes in the array according to gene type and sample group.

https://doi.org/10.1371/journal.pone.0128341.t001

We first compared gene expression profiles of neutrophils across the four groups of samples, grouped by septic status and age (Fig 1). S1 Table shows the clinical and laboratory characteristics of the study groups. As expected, the occurrence of sepsis affected the expression of a greater number of genes (4- to 5-fold) compared with aging (Table 1). We detected 421 genes that were differentially expressed between old and young subjects with sepsis, and 476 between old and young healthy controls (Fig 1). Only 1.4% of genes deregulated in old subjects compared with young subjects were common to both septic and healthy subjects. We also observed a number of lncRNAs that were deregulated in both sepsis and aging (Table 1).

thumbnail
Fig 1. Venn diagram of differentially expressed genes in each group according to age and disease status.

https://doi.org/10.1371/journal.pone.0128341.g001

To identify transcriptional changes that might explain the molecular basis of the clinical behavior in septic elderly patients, we examined the molecular pathways that were differentially expressed between elderly and young adults. First, we identified a number of canonical pathways that were enriched in genes deregulated in the elderly. Interestingly, most of these pathways were significantly enriched only in the sepsis group, i.e., genes were differentially expressed between elderly subjects with sepsis and younger adults with sepsis, with marginal enrichment in the healthy group (Table 2). The oxidative phosphorylation (p = 5.3 × 10−13) and mitochondrial dysfunction (p = 1.4 × 10−10) pathways were the most enriched in septic patients of advanced age when compared with the young septic group. Other pathways significantly enriched in the elderly were extracellular signal-regulated kinase 5 (ERK5) signaling (p = 9.4 × 10−4) and the NRF2-mediated oxidative stress response (p = 5.2 × 10−3) (Table 2). Table 3 lists the enzymes and transporters involved in oxidative phosphorylation that were differentially expressed in elderly septic patients.

thumbnail
Table 2. Differentially enriched canonical pathways between elderly and young individuals, with or without sepsis.

https://doi.org/10.1371/journal.pone.0128341.t002

thumbnail
Table 3. Genes from the oxidative phosphorylation pathway differentially expressed between elderly and young individuals with sepsis.

https://doi.org/10.1371/journal.pone.0128341.t003

Next, we performed a pathway enrichment analysis of the genes differentially expressed in septic patients compared with healthy controls. Many pathways were identified, but none showed significantly different enrichment in the elderly compared with younger adults. Pathways that are modulated during sepsis such as Cdc42 signaling [37], phospholipase C signaling [38], interleukin 17 (IL-17) signaling [39], protein ubiquitination pathway [40], glucocorticoid receptor [41] and p38 MAPK signaling [42] were observed.

To favor the identification of molecular pathways preferentially affected in the elderly with sepsis, we performed a similar analysis using a subset of genes differentially expressed between elderly sepsis and healthy subjects, but not in younger adults (944 genes, S2 Fig). Canonical pathways preferentially altered in elderly individuals with sepsis are shown in Table 4. These pathways included transforming growth factor β (TGF-β) signaling, Wnt/β-catenin signaling, calcium signaling and other pathways of interest relevant to sepsis. Notably, during sepsis, the TGF-β pathway was modulated both in the elderly and in younger adults. However, we noted an excess of TFG-β signaling genes upregulated in elderly septic individuals but not in younger septic individuals, as well as differences in the repertoire of genes modulated upon sepsis (Table 5).

thumbnail
Table 4. Canonical pathways enriched with genes altered in elderly individuals with sepsis compared with elderly controls, but not altered in young adult septic patients compared with young adult controls.

https://doi.org/10.1371/journal.pone.0128341.t004

thumbnail
Table 5. TFG-β signaling pathway genes exclusively differentially expressed in elderly individuals with sepsis compared with controls.

https://doi.org/10.1371/journal.pone.0128341.t005

A bioinformatics analysis based on the expression profiles of known targets identified by microarray identified several upstream regulators predicted to be either activated or inhibited in aging. Among those we focused on were putative regulators that had opposite activity in elderly septic individuals compared with healthy controls (Table 6). Interestingly, these included the upstream regulators of pathways detected in the previous analysis including ERK and TGF-β1, which were more active in septic elderly subjects compared with healthy elderly subjects, and when compared with matched young controls. This indicated that regulatory microRNAs might be predicted to be activated (miR-590-3p) or inhibited (miR-141-3p, miR-186-5p) in septic elders, as well as compounds that may exert an antagonic regulatory role in these patients compared with healthy controls (Table 6), suggesting novel potential therapeutic areas.

thumbnail
Table 6. Putative upstream regulators with inverted activity between elderly and young adults, with or without sepsis.

https://doi.org/10.1371/journal.pone.0128341.t006

To provide independent support of the DNA microarray data analysis, quantitative PCR experiments were performed for the following genes: 1) NDUFA4 and SDHC, to investigate respiratory chain activity and 2) INHBB, TGFB1 and CREBBP, to investigate TGF-β signaling. HDAC4 (calcium signaling) and SRC (Wnt/β-catenin signaling) were also investigated because these genes were significantly altered in elderly individuals with sepsis compared with healthy elderly controls, and were not expressed at all in young adults (Table 4). In general, we observed a good association (Pearson correlation = 0.68, p < 0.05) between qPCR and DNA microarray measurements (S3 Fig). NDUFA4, SDHC, and INHBB were significantly altered (p < 0.02). Although not statistically significant, TGFB1 and HDAC4 showed transcriptional changes in agreement with those observed in the microarray. Only CREBBP and SRC showed changes that were the reverse of those observed in the microarray, but these were not statistically significant, and therefore were inconclusive. Overall, these results support our observation that aged people had a reduced expression of genes related to respiratory chain activity and an increased activation of TGF-β-related genes during the course of overwhelming infection.

Discussion

Transcriptomics is a powerful technique used to identify therapeutic targets and biomarker signatures during infectious disease [43, 44]. A number of gene expression profiling studies were previously performed in sepsis, revealing a persistent repression of genes involved in adaptive immunity and the massive activation of innate immune pathways during septic shock [45]. Using whole blood-derived RNA, Wong et al. observed the upregulation of oxidative phosphorylation, IL-10 signaling, TLR signaling, NRF2-mediated oxidative stress responses, ubiquinone biosynthesis, TREM signaling, NF-κB signaling, protein ubiquitination pathways and IL-6 signaling in macrophages. They also observed the downregulation of T lymphocyte pathways and CCR5 signaling [46]. Similarly, Cvijanovich et al. detected the upregulation of TLR, IL-10, IL-6 and NF-κB signaling and the downregulation of T lymphocyte activation. Moreover, they detected the upregulation of acute phase responses, p38 MAPK, the complement system, and some nuclear receptor signaling molecules (LXR and PPAR) associated with the downregulation of antigen presentation pathways [47].

Using whole blood-derived RNA from patients in septic shock, the global gene expression experiments of Shanley et al. agreed with these studies. However, they also observed the upregulation of integrin, IGF-1, GM-CSF and insulin receptor signaling [48]. Similarly, Tang et al. observed the novel activation of many apoptotic genes, including CARD12, APAF1 and ELMOD2 in mononuclear cells from septic patients [49]. Surprisingly, a recent study in severe blunt trauma patients reported similar findings, with the activation of a large number of genes involved in inflammation, pattern recognition and antimicrobial functions with the simultaneous suppression of genes involved in antigen presentation and T cell proliferation, suggesting that severe physiological stress, regardless of its origin, may present common genomic signatures [50].

Sepsis affects very different groups of patients (including aged people, diabetics, patients with end-stage renal disease, trauma victims, surgical patients’, and obese people). Historically, it has been accepted that each specific subset of septic patients may have its own characteristic inflammatory immune response. In the course of sepsis, a transition from inflammation to immunosuppression has been suggested to explain the disappointing results obtained by clinical trials that investigated the use of anti-inflammatory drugs in this population [5153]. High-throughput transcriptome analysis has provided additional information and clustering algorithms using separate training and validation cohorts suggested the existence of gene expression subtypes in sepsis and septic shock [54, 55]. Furthermore, a study of gene expression in patients with sepsis secondary to community-acquired pneumonia distinguish survivors from nonsurvivors in part by variations in the expression of genes implicated in energy metabolism [56]. In contrast, a recent systematic review based on DNA microarray expression data was unable to detect a distinctive pro-inflammatory or anti-inflammatory phase in early or late sepsis or differences in the expression patterns between groups [57]. Thus, this subject remains highly controversial.

Most transcriptome studies in sepsis were performed in a pediatric population, and to our knowledge, our study is the first genome-wide expression analysis comparing septic shock patients of advanced age with young adults.

Initially, our results confirmed that both the young and the elderly responded alike regarding the production of tumor necrosis factor α (TNFα), IL-6, IL-1β, TLRs and other classical markers of cell activation, following severe infectious stress. Some specific pathways, however, appeared to be more critically affected in the elderly with sepsis, explaining the worse prognosis observed in these patients. Based on the data shown here, it is our opinion that defects in mitochondrial function/oxidative phosphorylation, TGF-β, Wnt/β-catenin, nerve growth factor (NGF) and calcium signaling may play a major role in the clinical features of sepsis in elderly patients.

We observed a marked decrease in the expression of genes encoding components of the mitochondrial respiratory chain in the septic elderly. Impairment of mitochondrial function significantly contributed to organ failure in septic patients [58]. Physiologically, small amounts of ROS are produced at complexes I and III of the respiratory chain. Sepsis is accompanied by increased oxidative stress caused by numerous factors, including the production of ROS by neutrophils, increased xanthine oxidase activity, increased nitric oxide plasma levels, and decreased antioxidant serum capacity [59]. Pro-inflammatory mediators and oxidative stress impair the function of the enzyme complexes of the respiratory chain and lead to structural damage to mitochondrial lipids, proteins and DNA [60, 61], promoting multiple organ failure [62]. Mitochondrial damage and dysfunction secondary to oxidative stress are also observed in aged cells [3, 63, 64]. Indeed, mice that present with defective editing functions of mitochondrial DNA polymerase exhibit a markedly shortened lifespan and increased features of aging, including alopecia, kyphosis, decreased activity, and a loss of reproductive function [65].

TGF-β has pleiotropic effects on adaptive immunity, especially for the regulation of effector and regulatory CD4+ T cell responses. TGF-β is a potent suppressor of Th1 and Th2 effector cell differentiation, a regulator of Foxp3+ regulatory T cells, and is a critical cytokine for the induction of Th9 and Th17 cells [6668]. TGF-β also has regulatory effects on CD8+ cells [69] and promotes phagocytosis [70]. Thus, we propose that increased activation of the TGF-β pathway in elderly septic patients may induce a prolonged downregulation of adaptive immunity and an increased Th17 response.

The Wnt/β-catenin pathway determines major developmental processes in the embryo and regulates maintenance, self-renewal and differentiation of adult mammalian tissue stem cells [71]. Many organs with a high cell turnover require continuous replenishment by somatic stem cells. Ageing of hematopoietic stem cells is associated with impaired hematopoiesis in the elderly [72, 73], which was attributed to a switch from canonical to non-canonical Wnt signaling [74]. In addition, a recent study reported that Wnt/β-catenin signaling induced the aging of mesenchymal stem cells through ROS generation [75]. These mechanisms might be responsible for defects in dendritic cell differentiation [76] and loss of T cell potential observed with age [77]. The Wnt/β-catenin pathway participates in the regulation of many other pathways, including NF-κB and PI3-kinase pathways, with important implications in the pathophysiology of septic shock [78]. In our opinion, the increased upregulation of the Wnt/β-catenin pathway in the elderly might be a critical detrimental factor.

Nerve growth factor (NGF) ensures the maintenance of phenotypic and functional characteristics of several populations of neurons and immune cells. The role of NGF has never been investigated in sepsis, but might be involved because neuropeptides possess a broad number of regulatory functions in immunity [79] and NGF directly affects the survival and differentiation of stem cells, granulocytes, lymphocytes and monocytes [8083]. Further studies should investigate its role in sepsis and in specific populations of septic patients, such as the elderly.

Calcium has a myriad of cellular functions, including effects on hormone secretion, enzyme activity, nerve conduction, and muscle contraction with obvious repercussions in sepsis. Calcium influx is a potent cell activator and, together with phosphate, alters electrostatic fields and protein conformation, dictating the majority of signal transduction [84].

The mechanisms that lead to the dysfunction of specific pathways in the immune responses of older people are complex and might involve multiple factors. We propose that environmental factors [85, 86], microRNAs [87] and epigenetic changes [88] might play a key role as modulating factors of the immune pathways that are particularly affected in older people.

Our data confirm previous reports that aging is accompanied by changes in the expression of genes related to immune responses [89], and further studies are required to support the involvement of pathways we identified and their relevance in sepsis. Interestingly, our analysis also suggests that a number of upstream regulators of molecular pathways display an inverted pattern of expression in elderly patients with sepsis compared with healthy controls, including microRNAs miR-590-3p (predicted activation in the septic elderly), miR-141-3p (predicted inhibition in the septic elderly) and miR-186-5p (predicted inhibition in the septic elderly). Another distinctive feature of this study was the observation for the first time that subsets of long noncoding RNAs are deregulated in the immune system in sepsis and aging, warranting additional studies aimed at investigating the biological roles exerted by this class of transcript in septic shock in patients of advanced age.

Conclusions

There is great hope that high-throughput screening technology will result in a better understanding of the complexity of systemic inflammation, identifying new therapeutic targets and groups of patients that may benefit from specific interventions.

Here, we identified major pathways preferentially deregulated in the elderly following severe infection, providing evidence that the systemic inflammatory response differs according to patient age. Thus, oxidative stress might have a major role in inducing multiple dysfunctions in the elderly. Furthermore, our study identified additional important genes and molecular pathways, thus helping to elucidate the mechanisms involved in signaling crosstalk during infection and in aging.

Supporting Information

S1 Fig. Hierarchical clustering of samples from sepsis (S) and healthy (H) subjects based on gene expression measurements.

After background filtering (see “Methods” for details), valid measurements from 16,698 genes were used to group samples using UPGMA hierarchical clustering and Pearson correlation as a distance measurement. Dendrograms are colored according to disease status (sepsis in red, healthy controls in blue) and age (elderly subjects are shown in darker colors whereas those from young adults are shown in lighter colors). Samples are well grouped by the disease status sepsis/healthy and moderately grouped by age.

https://doi.org/10.1371/journal.pone.0128341.s001

(DOCX)

S2 Fig. Venn diagram showing all possible intersections in differentially expressed genes between all groups.

https://doi.org/10.1371/journal.pone.0128341.s002

(DOCX)

S3 Fig. Confirmation of gene expression changes in sepsis and age detected by microarray using Real-Time PCR.

Genes with comparable changes in gene expression are shown in green and with an inverted pattern in red. Statistically significant results are shown in dark green. Values that follow a trend, but do not reach statistical significance are shown in light green.

https://doi.org/10.1371/journal.pone.0128341.s003

(DOCX)

Acknowledgments

MCCM is supported by FAPESP, the Sao Paulo Research Foundation (grant # 2012/03677-9). FPS is supported by CNPq, the National Council for Scientific and Technological Development (grant # 470539/2013-15). DVSP receives a M.Sc. fellowship from FAPESP (2014/03150-6) and EMR receives an established investigator fellowship from CNPq.

Author Contributions

Conceived and designed the experiments: FPS MCCM EMR PS HPS ITV. Performed the experiments: PS HVB FMA. Analyzed the data: DVSP EMR. Wrote the paper: FPS EMR DVSP PS MCCM.

References

  1. 1. Iskander KN, Osuchowski MF, Stearns-Kurosawa DJ, Kurosawa S, Stepien D, Valentine C, et al. Sepsis: multiple abnormalities, heterogeneous responses, and evolving understanding. Physiological reviews. 2013;93(3):1247–88. Epub 2013/08/01. pmid:23899564; PubMed Central PMCID: PMC3962548.
  2. 2. De Gaudio AR, Rinaldi S, Chelazzi C, Borracci T. Pathophysiology of sepsis in the elderly: clinical impact and therapeutic considerations. Current drug targets. 2009;10(1):60–70. Epub 2009/01/20 pmid:19149537.
  3. 3. Opal SM, Girard TD, Ely EW. The immunopathogenesis of sepsis in elderly patients. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2005;41 Suppl 7:S504–12. Epub 2005/10/21. pmid:16237654.
  4. 4. Heppner HJ, Cornel S, Peter W, Philipp B, Katrin S. Infections in the elderly. Critical care clinics. 2013;29(3):757–74. Epub 2013/07/09. pmid:23830661.
  5. 5. Nasa P, Juneja D, Singh O. Severe sepsis and septic shock in the elderly: An overview. World journal of critical care medicine. 2012;1(1):23–30. Epub 2012/02/04. pmid:24701398; PubMed Central PMCID: PMC3956061.
  6. 6. Girard TD, Opal SM, Ely EW. Insights into severe sepsis in older patients: from epidemiology to evidence-based management. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America. 2005;40(5):719–27. Epub 2005/02/17. pmid:15714419.
  7. 7. Grubeck-Loebenstein B, Wick G. The aging of the immune system. Advances in immunology. 2002;80:243–84. Epub 2002/06/25 pmid:12078483.
  8. 8. Plackett TP, Schilling EM, Faunce DE, Choudhry MA, Witte PL, Kovacs EJ. Aging enhances lymphocyte cytokine defects after injury. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2003;17(6):688–9. Epub 2003/02/21. pmid:12594182.
  9. 9. Mascarucci P, Taub D, Saccani S, Paloma MA, Dawson H, Roth GS, et al. Age-related changes in cytokine production by leukocytes in rhesus monkeys. Aging. 2001;13(2):85–94. Epub 2001/06/19 pmid:11405390.
  10. 10. Weksler ME, Goodhardt M, Szabo P. The effect of age on B cell development and humoral immunity. Springer seminars in immunopathology. 2002;24(1):35–52. Epub 2002/04/27 pmid:11974580.
  11. 11. Shinkai S, Konishi M, Shephard RJ. Aging and immune response to exercise. Canadian journal of physiology and pharmacology. 1998;76(5):562–72. Epub 1998/12/05 pmid:9839083.
  12. 12. Renshaw M, Rockwell J, Engleman C, Gewirtz A, Katz J, Sambhara S. Cutting edge: impaired Toll-like receptor expression and function in aging. Journal of immunology. 2002;169(9):4697–701. Epub 2002/10/23 pmid:12391175.
  13. 13. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nature reviews Immunology. 2013;13(12):875–87. Epub 2013/10/26. pmid:24157572; PubMed Central PMCID: PMC4096436.
  14. 14. Howcroft TK, Campisi J, Louis GB, Smith MT, Wise B, Wyss-Coray T, et al. The role of inflammation in age-related disease. Aging (Albany NY). 2013;5(1):84–93. Epub 2013/03/12 pmid:23474627; PubMed Central PMCID: PMC3616233.
  15. 15. Boehmer ED, Goral J, Faunce DE, Kovacs EJ. Age-dependent decrease in Toll-like receptor 4-mediated proinflammatory cytokine production and mitogen-activated protein kinase expression. Journal of leukocyte biology. 2004;75(2):342–9. Epub 2003/11/25. pmid:14634059.
  16. 16. Plackett TP, Boehmer ED, Faunce DE, Kovacs EJ. Aging and innate immune cells. Journal of leukocyte biology. 2004;76(2):291–9. Epub 2004/03/25. pmid:15039467.
  17. 17. Franceschi C, Bonafe M, Valensin S. Human immunosenescence: the prevailing of innate immunity, the failing of clonotypic immunity, and the filling of immunological space. Vaccine. 2000;18(16):1717–20. Epub 2000/02/26 pmid:10689155.
  18. 18. Fagiolo U, Cossarizza A, Scala E, Fanales-Belasio E, Ortolani C, Cozzi E, et al. Increased cytokine production in mononuclear cells of healthy elderly people. European journal of immunology. 1993;23(9):2375–8. Epub 1993/09/01. pmid:8370415.
  19. 19. Mari D, Mannucci PM, Coppola R, Bottasso B, Bauer KA, Rosenberg RD. Hypercoagulability in centenarians: the paradox of successful aging. Blood. 1995;85(11):3144–9. Epub 1995/06/01 pmid:7756646.
  20. 20. Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Current opinion in clinical nutrition and metabolic care. 2013;16(1):14–20. Epub 2012/11/08. pmid:23132168.
  21. 21. Starr ME, Saito H. Sepsis in old age: review of human and animal studies. Aging and disease. 2014;5(2):126–36. Epub 2014/04/15. pmid:24729938; PubMed Central PMCID: PMC3966671.
  22. 22. Tateda K, Matsumoto T, Miyazaki S, Yamaguchi K. Lipopolysaccharide-induced lethality and cytokine production in aged mice. Infection and immunity. 1996;64(3):769–74. Epub 1996/03/01 pmid:8641780; PubMed Central PMCID: PMC173836.
  23. 23. Turnbull IR, Wlzorek JJ, Osborne D, Hotchkiss RS, Coopersmith CM, Buchman TG. Effects of age on mortality and antibiotic efficacy in cecal ligation and puncture. Shock. 2003;19(4):310–3. Epub 2003/04/12 pmid:12688540.
  24. 24. Pinheiro da Silva F, Zampieri FG, Barbeiro DF, Barbeiro HV, Goulart AC, Torggler Filho F, et al. Septic shock in older people: a prospective cohort study. Immunity & ageing: I & A. 2013;10(1):21. Epub 2013/06/08. pmid:23742671; PubMed Central PMCID: PMC3685530.
  25. 25. Kale S, Yende S, Kong L, Perkins A, Kellum JA, Newman AB, et al. The effects of age on inflammatory and coagulation-fibrinolysis response in patients hospitalized for pneumonia. PloS one. 2010;5(11):e13852. Epub 2010/11/19. pmid:21085465; PubMed Central PMCID: PMC2973976.
  26. 26. Kelly E, MacRedmond RE, Cullen G, Greene CM, McElvaney NG, O'Neill SJ. Community-acquired pneumonia in older patients: does age influence systemic cytokine levels in community-acquired pneumonia? Respirology. 2009;14(2):210–6. Epub 2009/03/11. pmid:19272082.
  27. 27. Kale SS, Yende S. Effects of Aging on Inflammation and Hemostasis through the Continuum of Critical Illness. Aging and disease. 2011;2(6):501–11. Epub 2012/03/08 pmid:22396897; PubMed Central PMCID: PMC3295067.
  28. 28. Tseng CW, Liu GY. Expanding roles of neutrophils in aging hosts. Current opinion in immunology. 2014;29C:43–8. Epub 2014/04/30. pmid:24776646.
  29. 29. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992;101(6):1644–55. Epub 1992/06/01 1303622.
  30. 30. Quackenbush J. Microarray data normalization and transformation. Nature genetics. 2002;32 Suppl:496–501. Epub 2002/11/28. pmid:12454644.
  31. 31. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(9):5116–21. Epub 2001/04/20. pmid:11309499; PubMed Central PMCID: PMC33173.
  32. 32. Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL, Chory J. RankProd: a bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics. 2006;22(22):2825–7. Epub 2006/09/20. pmid:16982708.
  33. 33. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome biology. 2004;5(10):R80. Epub 2004/10/06. pmid:15461798; PubMed Central PMCID: PMC545600.
  34. 34. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. Epub 2002/02/16. pmid:11846609.
  35. 35. Nakaya HI, Amaral PP, Louro R, Lopes A, Fachel AA, Moreira YB, et al. Genome mapping and expression analyses of human intronic noncoding RNAs reveal tissue-specific patterns and enrichment in genes related to regulation of transcription. Genome biology. 2007;8(3):R43. Epub 2007/03/28. pmid:17386095; PubMed Central PMCID: PMC1868932.
  36. 36. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome research. 2012;22(9):1775–89. Epub 2012/09/08. pmid:22955988; PubMed Central PMCID: PMC3431493.
  37. 37. Cordero D, Fullenkamp CR, Pelly RR, Reed KM, Caffo LM, Zahrt AN, et al. Small Molecule Inhibitors Limit Endothelial Cell Invasion by Staphylococcus Aureus. Current pharmaceutical biotechnology. 2014. Epub 2014/09/13 pmid:25213310.
  38. 38. Kim SD, Kim HJ, Shim JW, Lee HY, Lee SK, Kwon S, et al. Phospholipase C activator m-3M3FBS protects against morbidity and mortality associated with sepsis. Journal of immunology. 2012;189(4):2000–5. Epub 2012/07/17. pmid:22798676.
  39. 39. Bosmann M, Ward PA. Therapeutic potential of targeting IL-17 and IL-23 in sepsis. Clinical and translational medicine. 2012;1(1):4. Epub 2013/02/02. pmid:23369364; PubMed Central PMCID: PMC3552565.
  40. 40. Zhong B, Liu X, Wang X, Liu X, Li H, Darnay BG, et al. Ubiquitin-specific protease 25 regulates TLR4-dependent innate immune responses through deubiquitination of the adaptor protein TRAF3. Science signaling. 2013;6(275):ra35. Epub 2013/05/16. pmid:23674823.
  41. 41. Deutschman CS. Steroid responses in sepsis: some novel thinking that may provide new insight. Critical care. 2013;17(3):147. Epub 2013/07/06. pmid:23826670; PubMed Central PMCID: PMC3706853.
  42. 42. Risco A, del Fresno C, Mambol A, Alsina-Beauchamp D, MacKenzie KF, Yang HT, et al. p38gamma and p38delta kinases regulate the Toll-like receptor 4 (TLR4)-induced cytokine production by controlling ERK1/2 protein kinase pathway activation. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(28):11200–5. Epub 2012/06/27. pmid:22733747; PubMed Central PMCID: PMC3396476.
  43. 43. Chaussabel D, Pascual V, Banchereau J. Assessing the human immune system through blood transcriptomics. BMC biology. 2010;8:84. Epub 2010/07/14. pmid:20619006; PubMed Central PMCID: PMC2895587.
  44. 44. Christie JD. Microarrays. Critical care medicine. 2005;33(12 Suppl):S449–52. Epub 2005/12/13 pmid:16340418.
  45. 45. Wong HR, Cvijanovich N, Allen GL, Lin R, Anas N, Meyer K, et al. Genomic expression profiling across the pediatric systemic inflammatory response syndrome, sepsis, and septic shock spectrum. Critical care medicine. 2009;37(5):1558–66. Epub 2009/03/28. pmid:19325468; PubMed Central PMCID: PMC2747356.
  46. 46. Wong HR, Freishtat RJ, Monaco M, Odoms K, Shanley TP. Leukocyte subset-derived genomewide expression profiles in pediatric septic shock. Pediatric critical care medicine: a journal of the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies. 2010;11(3):349–55. Epub 2009/12/17. pmid:20009785; PubMed Central PMCID: PMC2927840.
  47. 47. Cvijanovich N, Shanley TP, Lin R, Allen GL, Thomas NJ, Checchia P, et al. Validating the genomic signature of pediatric septic shock. Physiological genomics. 2008;34(1):127–34. Epub 2008/05/08. pmid:18460642; PubMed Central PMCID: PMC2440641.
  48. 48. Shanley TP, Cvijanovich N, Lin R, Allen GL, Thomas NJ, Doctor A, et al. Genome-level longitudinal expression of signaling pathways and gene networks in pediatric septic shock. Molecular medicine. 2007;13(9–10):495–508. Epub 2007/10/13. pmid:17932561; PubMed Central PMCID: PMC2014731.
  49. 49. Tang BM, McLean AS, Dawes IW, Huang SJ, Lin RC. Gene-expression profiling of peripheral blood mononuclear cells in sepsis. Critical care medicine. 2009;37(3):882–8. Epub 2009/02/25. pmid:19237892.
  50. 50. Xiao W, Mindrinos MN, Seok J, Cuschieri J, Cuenca AG, Gao H, et al. A genomic storm in critically injured humans. The Journal of experimental medicine. 2011;208(13):2581–90. Epub 2011/11/24. pmid:22110166; PubMed Central PMCID: PMC3244029.
  51. 51. Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Critical care medicine. 1996;24(7):1125–8. Epub 1996/07/01 pmid:8674323.
  52. 52. Bone RC. Why sepsis trials fail. JAMA: the journal of the American Medical Association. 1996;276(7):565–6. Epub 1996/08/21 pmid:8709407.
  53. 53. Bone RC, Grodzin CJ, Balk RA. Sepsis: a new hypothesis for pathogenesis of the disease process. Chest. 1997;112(1):235–43. Epub 1997/07/01 pmid:9228382.
  54. 54. Maslove DM, Wong HR. Gene expression profiling in sepsis: timing, tissue, and translational considerations. Trends in molecular medicine. 2014;20(4):204–13. Epub 2014/02/20. pmid:24548661; PubMed Central PMCID: PMC3976710.
  55. 55. Maslove DM, Tang BM, McLean AS. Identification of sepsis subtypes in critically ill adults using gene expression profiling. Critical care. 2012;16(5):R183. Epub 2012/10/06. pmid:23036193; PubMed Central PMCID: PMC3682285.
  56. 56. Severino P, Silva E, Baggio-Zappia GL, Brunialti MK, Nucci LA, Rigato O Jr., et al. Patterns of gene expression in peripheral blood mononuclear cells and outcomes from patients with sepsis secondary to community acquired pneumonia. PloS one. 2014;9(3):e91886. Epub 2014/03/29. pmid:24667684; PubMed Central PMCID: PMC3965402.
  57. 57. Tang BM, Huang SJ, McLean AS. Genome-wide transcription profiling of human sepsis: a systematic review. Critical care. 2010;14(6):R237. Epub 2010/12/31. pmid:21190579; PubMed Central PMCID: PMC3219990.
  58. 58. Tang BM, McLean AS, Dawes IW, Huang SJ, Lin RC. The use of gene-expression profiling to identify candidate genes in human sepsis. American journal of respiratory and critical care medicine. 2007;176(7):676–84. Epub 2007/06/19. pmid:17575094.
  59. 59. Wendel M, Heller AR. Mitochondrial function and dysfunction in sepsis. Wiener medizinische Wochenschrift. 2010;160(5–6):118–23. Epub 2010/04/07. pmid:20364414.
  60. 60. Suliman HB, Carraway MS, Piantadosi CA. Postlipopolysaccharide oxidative damage of mitochondrial DNA. American journal of respiratory and critical care medicine. 2003;167(4):570–9. Epub 2002/12/14. pmid:12480607.
  61. 61. Garcia-Ruiz C, Colell A, Mari M, Morales A, Fernandez-Checa JC. Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione. The Journal of biological chemistry. 1997;272(17):11369–77. Epub 1997/04/25 pmid:9111045.
  62. 62. Galley HF. Oxidative stress and mitochondrial dysfunction in sepsis. British journal of anaesthesia. 2011;107(1):57–64. Epub 2011/05/21. pmid:21596843.
  63. 63. Cannizzo ES, Clement CC, Sahu R, Follo C, Santambrogio L. Oxidative stress, inflamm-aging and immunosenescence. Journal of proteomics. 2011;74(11):2313–23. Epub 2011/07/02. pmid:21718814.
  64. 64. Floyd RA, Towner RA, He T, Hensley K, Maples KR. Translational research involving oxidative stress and diseases of aging. Free radical biology & medicine. 2011;51(5):931–41. Epub 2011/05/10. pmid:21549833; PubMed Central PMCID: PMC3156308.
  65. 65. Trifunovic A, Wredenberg A, Falkenberg M, Spelbrink JN, Rovio AT, Bruder CE, et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature. 2004;429(6990):417–23. Epub 2004/05/28. pmid:15164064.
  66. 66. Oh SA, Li MO. TGF-beta: guardian of T cell function. Journal of immunology. 2013;191(8):3973–9. Epub 2013/10/08. pmid:24098055; PubMed Central PMCID: PMC3856438.
  67. 67. Tu E, Chia PZ, Chen W. TGFbeta in T cell biology and tumor immunity: Angel or devil? Cytokine & growth factor reviews. 2014. Epub 2014/08/27. https://doi.org/10.1016/j.cytogfr.2014.07.014 pmid:25156420.
  68. 68. Hiraki S, Ono S, Tsujimoto H, Kinoshita M, Takahata R, Miyazaki H, et al. Neutralization of interleukin-10 or transforming growth factor-beta decreases the percentages of CD4+ CD25+ Foxp3+ regulatory T cells in septic mice, thereby leading to an improved survival. Surgery. 2012;151(2):313–22. Epub 2011/10/11. pmid:21982068.
  69. 69. Travis MA, Sheppard D. TGF-beta activation and function in immunity. Annual review of immunology. 2014;32:51–82. Epub 2013/12/10. pmid:24313777; PubMed Central PMCID: PMC4010192.
  70. 70. Sharma L, Wu W, Dholakiya SL, Gorasiya S, Wu J, Sitapara R, et al. Assessment of phagocytic activity of cultured macrophages using fluorescence microscopy and flow cytometry. Methods in molecular biology. 2014;1172:137–45. Epub 2014/06/09. pmid:24908301.
  71. 71. Van Camp JK, Beckers S, Zegers D, Van Hul W. Wnt signaling and the control of human stem cell fate. Stem cell reviews. 2014;10(2):207–29. Epub 2013/12/11. pmid:24323281.
  72. 72. Rossi DJ, Bryder D, Zahn JM, Ahlenius H, Sonu R, Wagers AJ, et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(26):9194–9. Epub 2005/06/22. pmid:15967997; PubMed Central PMCID: PMC1153718.
  73. 73. Geiger H, Van Zant G. The aging of lympho-hematopoietic stem cells. Nature immunology. 2002;3(4):329–33. Epub 2002/03/29. pmid:11919569.
  74. 74. Florian MC, Nattamai KJ, Dorr K, Marka G, Uberle B, Vas V, et al. A canonical to non-canonical Wnt signalling switch in haematopoietic stem-cell ageing. Nature. 2013;503(7476):392–6. Epub 2013/10/22. pmid:24141946; PubMed Central PMCID: PMC4078992.
  75. 75. Zhang DY, Pan Y, Zhang C, Yan BX, Yu SS, Wu DL, et al. Wnt/beta-catenin signaling induces the aging of mesenchymal stem cells through promoting the ROS production. Molecular and cellular biochemistry. 2013;374(1–2):13–20. Epub 2012/11/06. pmid:23124852.
  76. 76. Valencia J, Hernandez-Lopez C, Martinez VG, Hidalgo L, Zapata AG, Vicente A, et al. Wnt5a skews dendritic cell differentiation to an unconventional phenotype with tolerogenic features. Journal of immunology. 2011;187(8):4129–39. Epub 2011/09/16. pmid:21918189.
  77. 77. Khoo ML, Carlin SM, Lutherborrow MA, Jayaswal V, Ma DD, Moore JJ. Gene profiling reveals association between altered Wnt signaling and loss of T-cell potential with age in human hematopoietic stem cells. Aging cell. 2014;13(4):744–54. Epub 2014/06/04. pmid:24889652.
  78. 78. Silva-Garcia O, Valdez-Alarcon JJ, Baizabal-Aguirre VM. The Wnt/-Catenin Signaling Pathway Controls the Inflammatory Response in Infections Caused by Pathogenic Bacteria. Mediators of inflammation. 2014;2014:310183. Epub 2014/08/20. pmid:25136145; PubMed Central PMCID: PMC4127235.
  79. 79. Pinheiro da Silva F, Machado MC, Velasco IT. Neuropeptides in sepsis: from brain pathology to systemic inflammation. Peptides. 2013;44:135–8. Epub 2013/04/16. pmid:23583479.
  80. 80. Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. Journal of translational medicine. 2012;10:239. Epub 2012/11/30. pmid:23190582; PubMed Central PMCID: PMC3543237.
  81. 81. Shi Y, Jin Y, Guo W, Chen L, Liu C, Lv X. Blockage of nerve growth factor modulates T cell responses and inhibits allergic inflammation in a mouse model of asthma. Inflammation research: official journal of the European Histamine Research Society [et al]. 2012;61(12):1369–78. Epub 2012/08/09. pmid:22871964.
  82. 82. Manni L, Rocco ML, Bianchi P, Soligo M, Guaragna M, Barbaro SP, et al. Nerve growth factor: basic studies and possible therapeutic applications. Growth factors. 2013;31(4):115–22. Epub 2013/06/20. pmid:23777359.
  83. 83. Kritas SK, Caraffa A, Antinolfi P, Saggini A, Pantalone A, Rosati M, et al. Nerve growth factor interactions with mast cells. International journal of immunopathology and pharmacology. 2014;27(1):15–9. Epub 2014/03/29 pmid:24674674.
  84. 84. Clapham DE. Calcium signaling. Cell. 2007;131(6):1047–58. Epub 2007/12/18. pmid:18083096.
  85. 85. Su DM, Aw D, Palmer DB. Immunosenescence: a product of the environment? Current opinion in immunology. 2013;25(4):498–503. Epub 2013/07/03. pmid:23809359.
  86. 86. Muller L, Pawelec G. Aging and immunity—impact of behavioral intervention. Brain, behavior, and immunity. 2014;39:8–22. Epub 2013/12/10. pmid:24315935.
  87. 87. Harries LW. MicroRNAs as Mediators of the Ageing Process. Genes. 2014;5(3):656–70. Epub 2014/08/21. pmid:25140888.
  88. 88. Weidner CI, Wagner W. The epigenetic tracks of aging. Biological chemistry. 2014. Epub 2014/09/11. https://doi.org/10.1515/hsz-2014-0180 pmid:25205717.
  89. 89. de Magalhaes JP, Curado J, Church GM. Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics. 2009;25(7):875–81. Epub 2009/02/05. pmid:19189975; PubMed Central PMCID: PMC2732303.