Transcriptomic Profile of Whole Blood Cells from Elderly Subjects Fed Probiotic Bacteria Lactobacillus rhamnosus GG ATCC 53103 (LGG) in a Phase I Open Label Study

Gloria Solano-Aguilar, Aleksey Molokin, Christine Botelho, Anne-Maria Fiorino, Bryan Vinyard, Robert Li, Celine Chen, Joseph Urban Jr, Harry Dawson, Irina Andreyeva, Miriam Haverkamp, Patricia L Hibberd, Gloria Solano-Aguilar, Aleksey Molokin, Christine Botelho, Anne-Maria Fiorino, Bryan Vinyard, Robert Li, Celine Chen, Joseph Urban Jr, Harry Dawson, Irina Andreyeva, Miriam Haverkamp, Patricia L Hibberd

Abstract

We examined gene expression of whole blood cells (WBC) from 11 healthy elderly volunteers participating on a Phase I open label study before and after oral treatment with Lactobacillus rhamnosus GG-ATCC 53103 (LGG)) using RNA-sequencing (RNA-Seq). Elderly patients (65-80 yrs) completed a clinical assessment for health status and had blood drawn for cellular RNA extraction at study admission (Baseline), after 28 days of daily LGG treatment (Day 28) and at the end of the study (Day 56) after LGG treatment had been suspended for 28 days. Treatment compliance was verified by measuring LGG-DNA copy levels detected in host fecal samples. Normalized gene expression levels in WBC RNA were analyzed using a paired design built within three analysis platforms (edgeR, DESeq2 and TSPM) commonly used for gene count data analysis. From the 25,990 transcripts detected, 95 differentially expressed genes (DEGs) were detected in common by all analysis platforms with a nominal significant difference in gene expression at Day 28 following LGG treatment (FDR<0.1; 77 decreased and 18 increased). With a more stringent significance threshold (FDR<0.05), only two genes (FCER2 and LY86), were down-regulated more than 1.5 fold and met the criteria for differential expression across two analysis platforms. The remaining 93 genes were only detected at this threshold level with DESeq2 platform. Data analysis for biological interpretation of DEGs with an absolute fold change of 1.5 revealed down-regulation of overlapping genes involved with Cellular movement, Cell to cell signaling interactions, Immune cell trafficking and Inflammatory response. These data provide evidence for LGG-induced transcriptional modulation in healthy elderly volunteers because pre-treatment transcription levels were restored at 28 days after LGG treatment was stopped. To gain insight into the signaling pathways affected in response to LGG treatment, DEG were mapped using biological pathways and genomic data mining packages to indicate significant biological relevance.

Trial registration: ClinicalTrials.gov NCT01274598.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Participant flow diagram.
Fig 1. Participant flow diagram.
Fig 2. Differential Expression Analysis of RNA-seq…
Fig 2. Differential Expression Analysis of RNA-seq Data.
Volcano plots depicting the fold difference in gene expression levels after consumption of LGG for 28 days. Volcano plots with DEGs generated from edge-R (Panel A), DESeq2 (Panel B) or TSPM (Panel C) analysis platforms. Colored points in red refer to down-regulated genes green for up-regulated genes according to their fold change (Log FC) in x-axis and p value (log 10 p-value) p<0.05 or p<0.1 in y-axis.
Fig 3. Ingenuity top gene network interaction…
Fig 3. Ingenuity top gene network interaction reflecting immune response-related transcriptome changes after consumption of LGG.
Nodes in the interaction network are encoded by differentially expressed genes detected by edge-R in blood from subjects consuming LGG for 28 days, up-regulated genes are depicted in shades of green and down-regulated genes are in shades of red. Transcriptional information derived from IPA knowledge database on interactions between the nodes (activation, expression, molecular cleavage or phosphorylation) was projected onto the interaction map with predicted downregulation effects represented with blue dashed lines and upregulation effects with orange lines. From this interaction map it can be seen that several downstream genes including growth factors, peptidases, G-coupled receptors and cytokines that are known to be regulated by NF-KB transcription factor are down-regulated.
Fig 4. Downstream effect analysis (DEA) on…
Fig 4. Downstream effect analysis (DEA) on whole blood cells of subjects consuming LGG for 28 days.
(A).The visualization is a hierarchical heat-map generated from edgeR analysis with filtered data where the major boxes represent a family (or category) of related functions. Each individual colored rectangle is a particular biological function or disease and the color indicates its predicted state: Increasing (orange), or decreasing (blue). Darker colors indicate higher absolute Z-scores. In this view the size of the rectangle is correlated with increasing overlap significance (p-value). The image has been cropped for better readability. (B) Heat-map comparison of Diseases and Biofunctions affected across all 4 analysis (edgeR 0.1 cpm/all, edgeR 0.1cpm/ 22, DESEq2, TSPM). Similarly color represents predicted state. (C). Individual Z-scores and mean Z-scores per each Bio Function affected. The Z-score algorithm is designed to reduce the chance that random data will generate significant predictions. Negative Z-scores indicate a down-regulation of Biofunction, positives Z-scores indicate an up-regulation of function. Absolute Z-score values higher than 2.0 can be used to make biological predictions.

References

    1. Lievin-Le Moal V, Servin AL (2014) Anti-infective activities of lactobacillus strains in the human intestinal microbiota: from probiotics to gastrointestinal anti-infectious biotherapeutic agents. Clin Microbiol Rev 27: 167–199. 10.1128/CMR.00080-13
    1. Chiba E, Tomosada Y, Vizoso-Pinto MG, Salva S, Takahashi T, Tsukida K, et al. (2013) Immunobiotic Lactobacillus rhamnosus improves resistance of infant mice against respiratory syncytial virus infection. Int Immunopharmacol 17: 373–382. 10.1016/j.intimp.2013.06.024
    1. Luoto R, Ruuskanen O, Waris M, Kalliomaki M, Salminen S, Isolauri E. (2014) Prebiotic and probiotic supplementation prevents rhinovirus infections in preterm infants: a randomized, placebo-controlled trial. J Allergy Clin Immunol 133: 405–413. 10.1016/j.jaci.2013.08.020
    1. Miettinen M, Pietila TE, Kekkonen RA, Kankainen M, Latvala S, Pirhonen J, et al. (2012) Nonpathogenic Lactobacillus rhamnosus activates the inflammasome and antiviral responses in human macrophages. Gut Microbes 3: 510–522. 10.4161/gmic.21736
    1. Kawase M, He F, Kubota A, Harata G, Hiramatsu M (2010) Oral administration of lactobacilli from human intestinal tract protects mice against influenza virus infection. Lett Appl Microbiol 51: 6–10. 10.1111/j.1472-765X.2010.02849.x
    1. Zelaya H, Tsukida K, Chiba E, Marranzino G, Alvarez S, Kitazawa H, et al. (2014) Immunobiotic lactobacilli reduce viral-associated pulmonary damage through the modulation of inflammation-coagulation interactions. Int Immunopharmacol 19: 161–173. 10.1016/j.intimp.2013.12.020
    1. Klaenhammer TR, Kleerebezem M, Kopp MV, Rescigno M (2012) The impact of probiotics and prebiotics on the immune system. Nat Rev Immunol 12: 728–734. 10.1038/nri3312
    1. van Baarlen P, Troost F, van der Meer C, Hooiveld G, Boekschoten M, Brummer RJ, et al. (2011) Human mucosal in vivo transcriptome responses to three lactobacilli indicate how probiotics may modulate human cellular pathways. Proc Natl Acad Sci U S A 108 Suppl 1: 4562–4569. 10.1073/pnas.1000079107
    1. Segers ME, Lebeer S (2014) Towards a better understanding of Lactobacillus rhamnosus GG—host interactions. Microb Cell Fact 13 Suppl 1: S7 10.1186/1475-2859-13-S1-S7
    1. Ammoscato F, Scirocco A, Altomare A, Matarrese P, Petitta C, Ascione B, et al. (2013) Lactobacillus rhamnosus protects human colonic muscle from pathogen lipopolysaccharide-induced damage. Neurogastroenterol Motil 25: 984–e777. 10.1111/nmo.12232
    1. Mirpuri J, Sotnikov I, Myers L, Denning TL, Yarovinsky F, Parkos CA, et al. (2012) Lactobacillus rhamnosus (LGG) regulates IL-10 signaling in the developing murine colon through upregulation of the IL-10R2 receptor subunit. PLoS One 7: e51955 10.1371/journal.pone.0051955
    1. Rodes L, Khan A, Paul A, Coussa-Charley M, Marinescu D, Tomaro-Puchesneau C, et al. (2013) Effect of probiotics Lactobacillus and Bifidobacterium on gut-derived lipopolysaccharides and inflammatory cytokines: an in vitro study using a human colonic microbiota model. J Microbiol Biotechnol 23: 518–526.
    1. Lee SK, Yang KM, Cheon JH, Kim TI, Kim WH (2012) [Anti-inflammatory mechanism of Lactobacillus rhamnosus GG in lipopolysaccharide- stimulated HT-29 cell]. Korean J Gastroenterol 60: 86–93.
    1. Douillard FP, Ribbera A, Kant R, Pietila TE, Jarvinen HM, Messing M, et al. (2013) Comparative genomic and functional analysis of 100 Lactobacillus rhamnosus strains and their comparison with strain GG. PLoS Genet 9: e1003683 10.1371/journal.pgen.1003683
    1. Kalliomaki M, Salminen S, Poussa T, Arvilommi H, Isolauri E (2003) Probiotics and prevention of atopic disease: 4-year follow-up of a randomised placebo-controlled trial. Lancet 361: 1869–1871.
    1. Ghadimi D, Vrese M, Heller KJ, Schrezenmeir J (2010) Effect of natural commensal-origin DNA on toll-like receptor 9 (TLR9) signaling cascade, chemokine IL-8 expression, and barrier integritiy of polarized intestinal epithelial cells. Inflamm Bowel Dis 16: 410–427. 10.1002/ibd.21057
    1. Bajaj JS, Heuman DM, Hylemon PB, Sanyal AJ, Puri P, Sterling LK, et al. (2014) Randomised clinical trial: Lactobacillus GG modulates gut microbiome, metabolome and endotoxemia in patients with cirrhosis. Aliment Pharmacol Ther 39: 1113–1125. 10.1111/apt.12695
    1. Hibberd PL, Kleimola L, Fiorino AM, Botelho C, Haverkamp M, Andreyeva I, et al. (2014) No Evidence of Harms of Probiotic Lactobacillus rhamnosus GG ATCC 53103 in Healthy Elderly-A Phase I Open Label Study to Assess Safety, Tolerability and Cytokine Responses. PLoS One 9: e113456 10.1371/journal.pone.0113456
    1. Schwochow D, Serieys LE, Wayne RK, Thalmann O (2012) Efficient recovery of whole blood RNA—a comparison of commercial RNA extraction protocols for high-throughput applications in wildlife species. BMC Biotechnol 12: 33 10.1186/1472-6750-12-33
    1. Mastrokolias A, den Dunnen JT, van Ommen GB, t Hoen PA, van Roon-Mom WM (2012) Increased sensitivity of next generation sequencing-based expression profiling after globin reduction in human blood RNA. BMC Genomics 13: 28 10.1186/1471-2164-13-28
    1. Robinson MD, McCarthy DJ, Smyth GK (2010) edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26: 139–140. 10.1093/bioinformatics/btp616
    1. Anders S, McCarthy DJ, Chen Y, Okoniewski M, Smyth GK, Huber W, et al. (2013) Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat Protoc 8: 1765–1786. 10.1038/nprot.2013.099
    1. Robles JA, Qureshi SE, Stephen SJ, Wilson SR, Burden CJ, Taylor JM (2012) Efficient experimental design and analysis strategies for the detection of differential expression using RNA-Sequencing. BMC Genomics 13: 484 10.1186/1471-2164-13-484
    1. Auer PL, Srivastava S Doerge RW Differential expression—the next generation and beyond. Brief Funct Genomics 11: 57–62. 10.1093/bfgp/elr041
    1. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I (2001) Controlling the false discovery rate in behavior genetics research. Behav Brain Res 125: 279–284.
    1. Storey JD, Tibshirani R (2003) Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 100: 9440–9445.
    1. Cox MP, Peterson DA, Biggs PJ (2010) SolexaQA: At-a-glance quality assessment of Illumina second-generation sequencing data. BMC Bioinformatics 11: 485 10.1186/1471-2105-11-485
    1. Trapnell C, Pachter L, Salzberg SL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111. 10.1093/bioinformatics/btp120
    1. Anders S, Huber W (2010) Differential expression analysis for sequence count data. Genome Biol 11: R106 10.1186/gb-2010-11-10-r106
    1. Peffers M, Liu X, Clegg P (2013) Transcriptomic signatures in cartilage ageing. Arthritis Res Ther 15: R98 10.1186/ar4278
    1. Alvarado S, Tajerian M, Millecamps M, Suderman M, Stone LS, Szyf M (2013) Peripheral nerve injury is accompanied by chronic transcriptome-wide changes in the mouse prefrontal cortex. Mol Pain 9: 21 10.1186/1744-8069-9-21
    1. Joosten SA, Fletcher HA, Ottenhoff TH (2013) A helicopter perspective on TB biomarkers: pathway and process based analysis of gene expression data provides new insight into TB pathogenesis. PLoS One 8: e73230 10.1371/journal.pone.0073230
    1. Li B, Tsoi LC, Swindell WR, Gudjonsson JE, Tejasvi T, Johnston A, et al. (2014) Transcriptome Analysis of Psoriasis in A Large Case-Control Sample: Rna-Seq Provides Insights Into Disease Mechanisms. J Invest Dermatol. 134: 1828–1838. 10.1038/jid.2014.28
    1. Kramer A, Green J, Pollard J Jr., Tugendreich S (2014) Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics 30: 523–530. 10.1093/bioinformatics/btt703
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545–15550.
    1. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al. (2006) The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313: 1929–1935.
    1. Solano-Aguilar G, Dawson H, Restrepo M, Andrews K, Vinyard B, Urban JF Jr. (2008) Detection of Bifidobacterium animalis subsp. lactis (Bb12) in the intestine after feeding of sows and their piglets. Appl Environ Microbiol 74: 6338–6347. 10.1128/AEM.00309-08
    1. Nadkarni MA, Martin FE, Jacques NA, Hunter N (2002) Determination of bacterial load by real-time PCR using a broad-range (universal) probe and primers set. Microbiology 148: 257–266.
    1. Delroisse JM, Boulvin AL, Parmentier I, Dauphin RD, Vandenbol M, Portetelle D (2008) Quantification of Bifidobacterium spp. and Lactobacillus spp. in rat fecal samples by real-time PCR. Microbiol Res 163: 663–670.
    1. Haarman M, Knol J (2006) Quantitative real-time PCR analysis of fecal Lactobacillus species in infants receiving a prebiotic infant formula. Appl Environ Microbiol 72: 2359–2365.
    1. Chavagnat F, Haueter M, Jimeno J, Casey MG (2002) Comparison of partial tuf gene sequences for the identification of lactobacilli. FEMS Microbiol Lett 217: 177–183.
    1. Ventura M, Canchaya C, Meylan V, Klaenhammer TR, Zink R (2003) Analysis, characterization, and loci of the tuf genes in lactobacillus and bifidobacterium species and their direct application for species identification. Appl Environ Microbiol 69: 6908–6922.
    1. Solano-Aguilar G, Fernandez KP, Ets H, Molokin A, Vinyard B, Urban JF Jr (2013) Characterization of fecal microbiota of children with diarrhea in 2 locations in Colombia. J Pediatr Gastroenterol Nutr 56: 503–511. 10.1097/MPG.0b013e318282aa12
    1. Thompson JD, Gibson TJ, Plewniak F, Jeanmougin F, Higgins DG (1997) The CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res 25: 4876–4882.
    1. Kim Y, Trace SE, Crowley JJ, Brownley KA, Hamer RM, Pisetsky DS, et al. (2013) Assessment of gene expression in peripheral blood using RNAseq before and after weight restoration in anorexia nervosa. Psychiatry Res 210: 287–293. 10.1016/j.psychres.2013.05.026
    1. Demasius W, Weikard R, Hadlich F, Muller KE, Kuhn C (2013) Monitoring the immune response to vaccination with an inactivated vaccine associated to bovine neonatal pancytopenia by deep sequencing transcriptome analysis in cattle. Vet Res 44: 93 10.1186/1297-9716-44-93
    1. Chung LM, Ferguson JP, Zheng W, Qian F, Bruno V, Montgomery RR, et al. (2013) Differential expression analysis for paired RNA-Seq data. BMC Bioinformatics 14: 110 10.1186/1471-2105-14-110
    1. Yendrek CR, Ainsworth EA, Thimmapuram J (2012) The bench scientist's guide to statistical analysis of RNA-Seq data. BMC Res Notes 5: 506 10.1186/1756-0500-5-506
    1. Acharya M, Borland G, Edkins AL, Maclellan LM, Matheson J, Ozanne BW, et al. (2010) CD23/FcepsilonRII: molecular multi-tasking. Clin Exp Immunol 162: 12–23. 10.1111/j.1365-2249.2010.04210.x
    1. Cooper AM, Hobson PS, Jutton MR, Kao MW, Drung B, Schmidt B, et al. (2012) Soluble CD23 controls IgE synthesis and homeostasis in human B cells. J Immunol 188: 3199–3207. 10.4049/jimmunol.1102689
    1. Yang D, Chen Q, Rosenberg HF, Rybak SM, Newton DL, Wang ZY, et al. (2004) Human ribonuclease A superfamily members, eosinophil-derived neurotoxin and pancreatic ribonuclease, induce dendritic cell maturation and activation. J Immunol 173: 6134–6142.
    1. Staples KJ, Smallie T, Williams LM, Foey A, Burke B, Foxwell BM, et al. (2007) IL-10 induces IL-10 in primary human monocyte-derived macrophages via the transcription factor Stat3. J Immunol 178: 4779–4785.
    1. van der Does AM, Hensbergen PJ, Bogaards SJ, Cansoy M, Deelder AM, van Leeuwen HC, et al. (2012) The human lactoferrin-derived peptide hLF1-11 exerts immunomodulatory effects by specific inhibition of myeloperoxidase activity. J Immunol 188: 5012–5019. 10.4049/jimmunol.1102777
    1. Gagro A, Toellner KM, Grafton G, Servis D, Branica S, Radojcic V, et al. (2003) Naive and memory B cells respond differentially to T-dependent signaling but display an equal potential for differentiation toward the centroblast-restricted CD77/globotriaosylceramide phenotype. Eur J Immunol 33: 1889–1898.
    1. Li D, Mehta JL (2000) Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arterioscler Thromb Vasc Biol 20: 1116–1122.
    1. Shu HB, Johnson H (2000) B cell maturation protein is a receptor for the tumor necrosis factor family member TALL-1. Proc Natl Acad Sci U S A 97: 9156–9161.
    1. Oh SM, Pyo CW, Kim Y, Choi SY (2004) Neutrophil lactoferrin upregulates the human p53 gene through induction of NF-kappaB activation cascade. Oncogene 23: 8282–8291.
    1. Walsh DE, Greene CM, Carroll TP, Taggart CC, Gallagher PM, O'Neill SJ, et al. (2001) Interleukin-8 up-regulation by neutrophil elastase is mediated by MyD88/IRAK/TRAF-6 in human bronchial epithelium. J Biol Chem 276: 35494–35499.
    1. Segev DL, Hoshiya Y, Stephen AE, Hoshiya M, Tran TT, MacLaughlin DT, et al. (2001) Mullerian inhibiting substance regulates NFkappaB signaling and growth of mammary epithelial cells in vivo. J Biol Chem 276: 26799–26806.
    1. Coward WR, Okayama Y, Sagara H, Wilson SJ, Holgate ST, Church MK (2002) NF-kappa B and TNF-alpha: a positive autocrine loop in human lung mast cells? J Immunol 169: 5287–5293.
    1. Gewurz BE, Towfic F, Mar JC, Shinners NP, Takasaki K, Zhao B, et al. (2012) Genome-wide siRNA screen for mediators of NF-kappaB activation. Proc Natl Acad Sci U S A 109: 2467–2472. 10.1073/pnas.1120542109
    1. Kraft S, Novak N, Katoh N, Bieber T, Rupec RA (2002) Aggregation of the high-affinity IgE receptor Fc(epsilon)RI on human monocytes and dendritic cells induces NF-kappaB activation. J Invest Dermatol 118: 830–837.
    1. Yu J, Mookherjee N, Wee K, Bowdish DM, Pistolic J, Li Y, et al. (2007) Host defense peptide LL-37, in synergy with inflammatory mediator IL-1beta, augments immune responses by multiple pathways. J Immunol 179: 7684–7691.
    1. Kasper B, Brandt E, Brandau S, Petersen F (2007) Platelet factor 4 (CXC chemokine ligand 4) differentially regulates respiratory burst, survival, and cytokine expression of human monocytes by using distinct signaling pathways. J Immunol 179: 2584–2591.
    1. Kim HJ, Kim YJ, Lee SH, Yu J, Jeong SK, Hong SJ (2014) Effects of Lactobacillus rhamnosus on allergic march model by suppressing Th2, Th17, and TSLP responses via CD4(+)CD25(+)Foxp3(+) Tregs. Clin Immunol 153: 178–186. 10.1016/j.clim.2014.04.008
    1. Rodes L, Coussa-Charley M, Marinescu D, Paul A, Fakhoury M, Abbasi S, et al. (2013) Design of a novel gut bacterial adhesion model for probiotic applications. Artif Cells Nanomed Biotechnol 41: 116–124. 10.3109/10731199.2012.712047
    1. Vong L, Lorentz RJ, Assa A, Glogauer M, Sherman PM (2014) Probiotic Lactobacillus rhamnosus inhibits the formation of neutrophil extracellular traps. J Immunol 192: 1870–1877. 10.4049/jimmunol.1302286
    1. Ashraf R, Vasiljevic T, Smith SC, Donkor ON (2014) Effect of cell-surface components and metabolites of lactic acid bacteria and probiotic organisms on cytokine production and induction of CD25 expression in human peripheral mononuclear cells. J Dairy Sci 97: 2542–2558. 10.3168/jds.2013-7459
    1. Rask C, Adlerberth I, Berggren A, Ahren IL, Wold AE (2013) Differential effect on cell-mediated immunity in human volunteers after intake of different lactobacilli. Clin Exp Immunol 172: 321–332. 10.1111/cei.12055
    1. Schultz M, Linde HJ, Lehn N, Zimmermann K, Grossmann J, Falk W, et al. (2003) Immunomodulatory consequences of oral administration of Lactobacillus rhamnosus strain GG in healthy volunteers. J Dairy Res 70: 165–173.
    1. Tomosada Y, Chiba E, Zelaya H, Takahashi T, Tsukida K, Kitasawa H, et al. (2013) Nasally administered Lactobacillus rhamnosus strains differentially modulate respiratory antiviral immune responses and induce protection against respiratory syncytial virus infection. BMC Immunol 14: 40 10.1186/1471-2172-14-40
    1. Wang Y, Liu Y, Kirpich I, Ma Z, Wang C, Zhang M, et al. (2013) Lactobacillus rhamnosus GG reduces hepatic TNFalpha production and inflammation in chronic alcohol-induced liver injury. J Nutr Biochem 24: 1609–1615. 10.1016/j.jnutbio.2013.02.001
    1. Marguerat S, Bahler J (2010) RNA-seq: from technology to biology. Cell Mol Life Sci 67: 569–579. 10.1007/s00018-009-0180-6
    1. St Laurent G, Shtokalo D, Tackett MR, Yang Z, Vyatkin Y, Milos PM, et al. (2013) On the importance of small changes in RNA expression. Methods 63: 18–24. 10.1016/j.ymeth.2013.03.027
    1. van Beveren NJ, Krab LC, Swagemakers S, Buitendijk GH, Boot E, van der Spek P, et al. (2012) Functional gene-expression analysis shows involvement of schizophrenia-relevant pathways in patients with 22q11 deletion syndrome. PLoS One 7: e33473 10.1371/journal.pone.0033473
    1. Lin D, Hollander Z, Ng RT, Imai C, Ignaszewski A, Balshaw R, et al. (2009) Whole blood genomic biomarkers of acute cardiac allograft rejection. J Heart Lung Transplant 28: 927–935. 10.1016/j.healun.2009.04.025
    1. Joseph P, Umbright C, Sellamuthu R (2013) Blood transcriptomics: applications in toxicology. J Appl Toxicol.33: 1193–1202. 10.1002/jat.2861
    1. Song HK, Hong SE, Kim T, Kim do H (2012) Deep RNA sequencing reveals novel cardiac transcriptomic signatures for physiological and pathological hypertrophy. PLoS One 7: e35552 10.1371/journal.pone.0035552
    1. Mele M, Ferreira PG, Reverter F, DeLuca DS, Monlong J, Sammeth M, et al. (2015) Human genomics. The human transcriptome across tissues and individuals. Science 348: 660–665. 10.1126/science.aaa0355
    1. Colitti M, Gaspardo B, Della Pria A, Scaini C, Stefanon B (2012) Transcriptome modification of white blood cells after dietary administration of curcumin and non-steroidal anti-inflammatory drug in osteoarthritic affected dogs. Vet Immunol Immunopathol 147: 136–146. 10.1016/j.vetimm.2012.04.001
    1. Dorr C, Wu B, Guan W, Muthusamy A, Sanghavi K, Schladt DP, et al. (2015) Differentially expressed gene transcripts using RNA sequencing from the blood of immunosuppressed kidney allograft recipients. PLoS One 10: e0125045 10.1371/journal.pone.0125045
    1. Li H, Lovci MT, Kwon YS, Rosenfeld MG, Fu XD, Yeo GW (2008) Determination of tag density required for digital transcriptome analysis: application to an androgen-sensitive prostate cancer model. Proc Natl Acad Sci U S A 105: 20179–20184. 10.1073/pnas.0807121105
    1. Liu Y, Zhou J, White KP (2013) RNA-seq differential expression studies: more sequence or more replication? Bioinformatics 30: 301–304. 10.1093/bioinformatics/btt688
    1. Dillies MA, Rau A, Aubert J, Hennequet-Antier C, Jeanmougin M, Servant N, et al. (2013) A comprehensive evaluation of normalization methods for Illumina high-throughput RNA sequencing data analysis. Brief Bioinform 14: 671–683. 10.1093/bib/bbs046
    1. Trapnell C, Hendrickson DG, Sauvageau M, Goff L, Rinn JL, Pachter L (2013) Differential analysis of gene regulation at transcript resolution with RNA-seq. Nat Biotechnol 31: 46–53. 10.1038/nbt.2450
    1. Guo Y, Li CI, Ye F, Shyr Y (2013) Evaluation of read count based RNAseq analysis methods. BMC Genomics 14 Suppl 8: S2 10.1186/1471-2164-14-S8-S2
    1. Kvam VM, Liu P, Si Y (2012) A comparison of statistical methods for detecting differentially expressed genes from RNA-seq data. Am J Bot 99: 248–256. 10.3732/ajb.1100340
    1. Zhang ZH, Jhaveri DJ, Marshall VM, Bauer DC, Edson J, Narayanan RK (2014) A Comparative Study of Techniques for Differential Expression Analysis on RNA-Seq Data. PLoS One 9: e103207 10.1371/journal.pone.0103207
    1. Shin H, Shannon CP, Fishbane N, Ruan J, Zhou M, Balshaw R (2014) Variation in RNA-Seq transcriptome profiles of peripheral whole blood from healthy individuals with and without globin depletion. PLoS One 9: e91041 10.1371/journal.pone.0091041
    1. Toung JM, Morley M, Li M, Cheung VG (2011) RNA-sequence analysis of human B-cells. Genome Res 21: 991–998. 10.1101/gr.116335.110
    1. Wright HL, Thomas HB, Moots RJ, Edwards SW (2013) RNA-seq reveals activation of both common and cytokine-specific pathways following neutrophil priming. PLoS One 8: e58598 10.1371/journal.pone.0058598
    1. Peng X, Sova P, Green RR, Thomas MJ, Korth MJ, Proll S, et al. (2014) Deep sequencing of HIV infected cells: insights into nascent transcription and host-directed therapy. J Virol. 88:8768–8782. 10.1128/JVI.00768-14
    1. Bullard JH, Purdom E, Hansen KD, Dudoit S (2010) Evaluation of statistical methods for normalization and differential expression in mRNA-Seq experiments. BMC Bioinformatics 11: 94 10.1186/1471-2105-11-94
    1. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008) Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5: 621–628. 10.1038/nmeth.1226
    1. Watanabe Y, Nakamura T, Ishikawa S, Fujisaka S, Usui I, Tsuneyama K, et al. (2012) The radioprotective 105/MD-1 complex contributes to diet-induced obesity and adipose tissue inflammation. Diabetes 61: 1199–1209. 10.2337/db11-1182
    1. Kalliomaki M, Salminen S, Arvilommi H, Kero P, Koskinen P, Isolauri E (2001) Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet 357: 1076–1079.
    1. Kalliomaki M, Isolauri E (2003) Role of intestinal flora in the development of allergy. Curr Opin Allergy Clin Immunol 3: 15–20.
    1. Lin WH, Wu CR, Lee HZ, Kuo YH, Wen HS, Lin TY. (2013) Induced apoptosis of Th2 lymphocytes and inhibition of airway hyperresponsiveness and inflammation by combined lactic acid bacteria treatment. Int Immunopharmacol 15: 703–711. 10.1016/j.intimp.2012.10.025
    1. Kim HJ, Kim YJ, Lee SH, Kang MJ, Yu HS, Jeong SK, et al. (2013) Effects of Lactobacillus rhamnosus on asthma with an adoptive transfer of dendritic cells in mice. J Appl Microbiol 115: 872–879. 10.1111/jam.12268
    1. Recchiuti A, Krishnamoorthy S, Fredman G, Chiang N, Serhan CN (2011) MicroRNAs in resolution of acute inflammation: identification of novel resolvin D1-miRNA circuits. FASEB J 25: 544–560. 10.1096/fj.10-169599
    1. Pujols L, Fernandez-Bertolin L, Fuentes-Prado M, Alobid I, Roca-Ferrer J, Agell N, et al. (2012) Proteasome inhibition reduces proliferation, collagen expression, and inflammatory cytokine production in nasal mucosa and polyp fibroblasts. J Pharmacol Exp Ther 343: 184–197. 10.1124/jpet.111.190710
    1. Bollrath J, Greten FR (2009) IKK/NF-kappaB and STAT3 pathways: central signalling hubs in inflammation-mediated tumour promotion and metastasis. EMBO Rep 10: 1314–1319. 10.1038/embor.2009.243

Source: PubMed

3
Suscribir