HIV-1-induced cytokines deplete homeostatic innate lymphoid cells and expand TCF7-dependent memory NK cells

Yetao Wang, Lawrence Lifshitz, Kyle Gellatly, Carol L Vinton, Kathleen Busman-Sahay, Sean McCauley, Pranitha Vangala, Kyusik Kim, Alan Derr, Smita Jaiswal, Alper Kucukural, Patrick McDonel, Peter W Hunt, Thomas Greenough, JeanMarie Houghton, Ma Somsouk, Jacob D Estes, Jason M Brenchley, Manuel Garber, Steven G Deeks, Jeremy Luban, Yetao Wang, Lawrence Lifshitz, Kyle Gellatly, Carol L Vinton, Kathleen Busman-Sahay, Sean McCauley, Pranitha Vangala, Kyusik Kim, Alan Derr, Smita Jaiswal, Alper Kucukural, Patrick McDonel, Peter W Hunt, Thomas Greenough, JeanMarie Houghton, Ma Somsouk, Jacob D Estes, Jason M Brenchley, Manuel Garber, Steven G Deeks, Jeremy Luban

Abstract

Human immunodeficiency virus 1 (HIV-1) infection is associated with heightened inflammation and excess risk of cardiovascular disease, cancer and other complications. These pathologies persist despite antiretroviral therapy. In two independent cohorts, we found that innate lymphoid cells (ILCs) were depleted in the blood and gut of people with HIV-1, even with effective antiretroviral therapy. ILC depletion was associated with neutrophil infiltration of the gut lamina propria, type 1 interferon activation, increased microbial translocation and natural killer (NK) cell skewing towards an inflammatory state, with chromatin structure and phenotype typical of WNT transcription factor TCF7-dependent memory T cells. Cytokines that are elevated during acute HIV-1 infection reproduced the ILC and NK cell abnormalities ex vivo. These results show that inflammatory cytokines associated with HIV-1 infection irreversibly disrupt ILCs. This results in loss of gut epithelial integrity, microbial translocation and memory NK cells with heightened inflammatory potential, and explains the chronic inflammation in people with HIV-1.

Conflict of interest statement

Declaration of interests

The authors declare no competing financial interests.

Figures

Extended Data Fig. 1
Extended Data Fig. 1
Extended Data Fig. 2
Extended Data Fig. 2
Extended Data Fig. 3
Extended Data Fig. 3
Extended Data Fig. 4
Extended Data Fig. 4
Extended Data Fig. 5
Extended Data Fig. 5
Extended Data Fig. 6
Extended Data Fig. 6
Extended Data Fig. 7
Extended Data Fig. 7
Fig. 1 |. HIV-1 infection decreases ILCs…
Fig. 1 |. HIV-1 infection decreases ILCs in blood and colon lamina propria.
a, Percent ILCs (Lin−CD56−CD16−CD127+ PBMCs; Extended Data Fig. 1a) from 40 HIV-1−, 36 HIV-1+ viremic, 39 HIV-1+ on ART, and 38 HIV-1+ spontaneous controllers (Supplementary Table 1). b-d, ILCs as in a, correlation with CD4s (b), CD4/CD8 ratio (c), and plasma sCD14 (d). Correlation coefficient (R) by Pearson, zero slope p value determined by the F-test (n=80). e,f, Percent Lin−CD127+ILCs (e) and Lin−CD127+RORγT+ILC3s (f), among colon lamina propria lymphoid cells from 6 HIV-1− and 7 HIV-1+ on ART with undetectable viremia (Supplementary Table 2). g, CD3−CD117+ ILC3s in rectosigmoid tissue from HIV-1− or HIV-1+ individuals on ART (Supplementary Table 1). White arrowheads, ILCs. Scale bar = 100 μm. h,i, As in g, percent ILC3s (h) and CD3−CD117+/CD3+CD117− (i) from 8 HIV-1− and 16 HIV-1+ on ART. j, MX1+ cells as in g. k, As in j, percent MX1+ as in h. l, MPO+ cells as in g. m, As in l, percent MPO+ cells as in h. n, CD127+ among Lin−CD56−CD16− PBMCs (HIV-1−), 16 hrs IL-15 with ruxolitinib (JAK1/2i, n=5), CP-690550 (JAK3i, n=11), rapamycin (RAPA, n=7), or no IL-15 (n=11). Data are mean ± s.e.m., a, e, f, h, i, k, m two-tailed unpaired t-test; n, two-tailed paired t-test. ns, not significant, *p<0.05, **p<0.01, ***p<0.001. g, j, l, three independent blocks for each donor with similar results.
Fig. 2 |. HIV-1 infection increases the…
Fig. 2 |. HIV-1 infection increases the proportion of CD94+ NK cells.
a, Lin−TBX21+ NK cells from HIV-1−, HIV-1+ viremic, HIV-1+ ART suppressed, and HIV-1+ spontaneous controllers (cohort description in Supplementary Table 1). b, Percent NK cells as in a (n=20 for each group). c, CD94 on NK cells (Lin−CD56+) in PBMCs from HIV-1−, HIV-1+ viremic, HIV-1+ ART suppressed, and HIV-1+ spontaneous controllers (Supplementary Table 1). d, As in c, percent of CD94+NK cells among NK cells from HIV-1− (n=37), HIV-1+ viremic (n=38), HIV-1+ on ART (n=38), and HIV-1+ spontaneous controllers (n=38). e, Percent CD94+ cells after 16 hrs PMA/ionomycin stimulation of Lin−CD56+CD94−NK cells sorted from HIV-1− PBMCs. f, As in e, percent CD94+NK cells after 16 hrs stimulation with PMA/ionomycin or IL-12 and IL-15 (n=6). g, Heatmap of differentially expressed genes by RNA-Seq, sorted Lin−CD56+CD94− versus Lin−CD56+CD94+NK cells, from PBMCs of two HIV-1− donors (log2 fold change >1, p<0.05 determined by DESeq2). h, Percent indicated proteins encoded by differentially expressed genes in Lin−CD56+CD94− and Lin−CD56+CD94+ NK cells from HIV-1− PBMCs, as in g, GZMK (n=21), TCF7 (n=15), CXCR3 (n=27), CD44 (n=26), CD2 (n=11), SELL (n=27), KIR2DL1 (n=18). Data are mean ± s.e.m. b, d, two-tailed unpaired t-test; f, h, two-tailed paired t-test. ns, not significant, *p<0.05, **p<0.01, ***p<0.001.
Fig. 3 |. TCF7 expression correlates with…
Fig. 3 |. TCF7 expression correlates with pseudotime trajectory from CD94−NK cells to CD94+NK Cells.
a, Two-dimensional tSNE plot of single cell RNA-Seq of 986 Lin−CD56+CD94−NK cells (pink) and 767 Lin−CD56+CD94+NK cells (aqua), sorted from HIV-1− blood. Data are representative of 2 donors. b, Spectral clustering of single cell transcriptomes from all cells in a, independent of CD94, k-nearest neighbor, search=2. c, Heatmap of 1,729 CD94−NK cells (blue) and 1,548 CD94+NK cells (yellow), sorted from 2 HIV-1− anonymous blood donors, using all differentially expressed genes from the spectral cluster analysis. d, Minimum spanning tree based on the transcriptome of individual cells from a, showing pseudotime trajectory (black line). e, TCF7 expression along the pseudotime trajectory. f, Expression and density of the indicated genes within t-SNE plots. g, Flow cytometry for CD56 and TCF7 on Lin− PBMCs. h, Sorted CD94−NK cells were untreated (No Stim) or treated with IL-15 (5 ng/ml) for 5 days, and IL-12 (50 ng/ml) and IL-15 (50 ng/ml) for 16 hrs (Stimulated). TCF7, CD44, CXCR3, and CXCR6 were detected by flow cytometry. TCF7, representative of 8 donors; CD44, CXCR3, and CXCR6, representative of 4 HIV-1− donors. All data were generated using blood from HIV-1− donors.
Fig. 4 |. TCF and WNT signaling…
Fig. 4 |. TCF and WNT signaling pathway enrichment in CD94+CD56hiNK cells.
a, Sorting strategy for CD94−CD56dim, CD94+CD56dim, and CD94+CD56hi NK cell subsets. b, Heatmap of differentially expressed genes by RNA-Seq (fold change of normalized counts, log2 >1, p<0.05 determined by DESeq2) for the indicated NK cell subsets sorted from four HIV-1− blood donors. c-e, Pairwise comparison of the indicated NK cell subsets based on differentially expressed genes. f, PCA based on RNA-Seq data from the indicated NK cell subsets. g, Reactome pathway analysis based on 152 differentially expressed genes in CD94+CD56hi NK cells. GENEONTOLOGY produces p value from Fisher’s exact test, followed by BH false discovery rate (in parentheses). All data were generated using blood from HIV-1− donors.
Fig. 5 |. Distinct chromatin landscape in…
Fig. 5 |. Distinct chromatin landscape in CD94+CD56hi NK cells.
a-c, Heatmap showing differential enrichment (>2 fold change of normalized counts) for H3K4me1 (a) and H3K4me3 (b) by CUT&RUN, or accessible chromatin by ATAC-Seq (c), in sorted CD94−CD56dim, CD94+CD56dim, and CD94+CD56hi NK cell subsets. Data is representative of two HIV-1− blood donors. d,e, H3K4me1, H3K4me3, and ATAC-Seq signal on TCF7 (d) and CD6 (e), in the indicated NK cell subsets. f,g,De novo analysis of transcription factor binding motifs enriched in open chromatin from CD56hi NK cells (f), or CD56dim NK cells (g), using HOMER. h, ATAC-Seq and TCF7 CUT&RUN signal at the indicated loci from the three NK cell subsets. All data were generated using blood from HIV-1− anonymous donors.
Fig. 6 |. Chromatin, transcriptome, and phenotype…
Fig. 6 |. Chromatin, transcriptome, and phenotype define CD94+CD56hi NK cells as memory cells.
a, CUT&RUN and ATAC-Seq showing memory gene-associated loci in sorted CD94−CD56dim, CD94+CD56dim, and CD94+CD56hi NK cells. b,c, Gene set enrichment analysis comparing memory CD8+ T cell expression signature (GSE9650) with sorted CD94+CD56hi and CD94−CD56dim NK cells (b), or with sorted CD94+CD56hi and CD94+CD56dim NK cells (c). p value and FDR determined using GSEAPreranked module from GenePattern with 1000 permutations. d, Sorted NK cells labelled with CFSE and cultured 5 days in IL-15 (5 ng/ml). CFSE and TCF7 detected by flow cytometry. e, Percent IFN-γ+ cells by flow cytometry after stimulation of sorted NK cell populations with IL-12 and IL-15 (n=4). f, Fold increase of surface CD107a on bead-enriched NK cells after incubation with K562 cells (n=4). g, bead-enriched NK cells incubated with IL-15 (5ng/ml) for 5 days, then with NL4-3-nef-GFP infected CD4+ T cells. Fold increase of surface CD107a on indicated NK populations, as in f (0h, n=5; 0.5h, 1h, 2h, n=3; 3h, n=5). h, IFNG-AS1 expression relative to GAPDH by RT-qPCR for the indicated NK cell populations sorted from 3 donors. Each population contains 2 technical replicates. i, IFNG-AS1 chromatin analysis as in a. Data are mean ± s.e.m. e-g, two-way ANOVA; h, two-tailed unpaired t-test. *p<0.05, **p<0.01, ***p<0.001. All data were generated using blood from HIV-1− anonymous donors.
Fig. 7 |. WNT inhibition blocks cytokine…
Fig. 7 |. WNT inhibition blocks cytokine induced NK memory response.
a, Experimental scheme for generation of memory NK cells ex vivo from sorted CD94−CD56dimNK cells. b, IFNG-AS1 expression relative to GAPDH by RT-qPCR using RNA extracted from sorted CD94−CD56dim NK cells just prior to secondary stimulation, after primary stimulation, and resting in low dose IL-15 (n=4). c,d, Gene set enrichment analysis (GSEA) comparing CD8+ memory T cell expression signature (GSE9650) by RNA-Seq from sorted CD94−CD56dim NK cells using RNA harvested as in b. p value and FDR determined using GSEAPreranked module from GenePattern with 1000 permutations. e, Heatmap of memory related genes by RNA-Seq that were also found in GSEA list (Supplementary Table 9,10). Fold change of normalized counts Log2>0.5, n=2. f, Heatmap of genes by RNA-Seq that distinguish memory from effector or naive cells reported by literature but that were not identified in GSEA list. Fold change of normalized counts Log2>0.5, n=2. g, Percent IFN-γ+ from sorted CD94−CD56dimNK cells after secondary stimulation. Data are mean ± s.e.m.; two-tailed paired t-test; *p<0.05, **p<0.01. All samples used were from HIV-1− anonymous blood donors.
Fig. 8 |. Expansion of TCF7 +…
Fig. 8 |. Expansion of TCF7+NK cells during HIV-1 infection.
a, Percent TCF7+NK cells among Lin−CD56+ PBMCs in HIV-1− (n=60), HIV-1+ viremic (n=46), HIV-1+ on ART (n=50), and HIV-1+ spontaneous controllers (n=40). Cohort characteristics described in Supplementary Table 1,2. b, Percent TCF7+NK cells from Lin−CD56+CD94−NK subsets of HIV-1− (n=61), HIV-1+ viremic (n=49), HIV-1+ on ART (n=57), and HIV-1+ spontaneous controllers (n=40) (Supplementary Table 1,2) c, Correlation of ILCs with CD56hiNK cells. Samples are detected from HIV-1+ viremic individuals, ART suppressed HIV-1+ individuals (ART), and HIV-1+ individuals who spontaneously control viremia without ART (Supplementary Table 1) (n=113). The correlation coefficient (R) was determined by Pearson, p value for the slope being zero was determined by the F-test. d, Percent NKG2C on Lin−CD56+ PBMCs from HIV-1− (n=9) and HIV-1+ (n=10, under ART) individuals (Supplementary Table 2). e, Model for effect of HIV-1 infection on NK cell subsets. Data are mean ± s.e.m.; a, b and d, two-tailed unpaired t-test. ns, not significant, *p<0.01, **p<0.001.

References

    1. Deeks SG, Tracy R & Douek DC Systemic effects of inflammation on health during chronic HIV infection. Immunity 39, 633–645 (2013).
    1. Brenchley JM et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med 12, 1365–1371 (2006).
    1. Vivier E et al. Innate Lymphoid Cells: 10 Years On. Cell 174, 1054–1066 (2018).
    1. Kløverpris HN et al. Innate Lymphoid Cells Are Depleted Irreversibly during Acute HIV-1 Infection in the Absence of Viral Suppression. Immunity 44, 391–405 (2016).
    1. Mudd JC et al. Hallmarks of primate lentiviral immunodeficiency infection recapitulate loss of innate lymphoid cells. Nat. Commun 9, 3967 (2018).
    1. Bruel T et al. Elimination of HIV-1-infected cells by broadly neutralizing antibodies. Nat. Commun 7, 10844 (2016).
    1. Alter G et al. HIV-1 adaptation to NK-cell-mediated immune pressure. Nature 476, 96–100 (2011).
    1. Cerwenka A & Lanier LL Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol 16, 112–123 (2016).
    1. Lim AI et al. Systemic Human ILC Precursors Provide a Substrate for Tissue ILC Differentiation. Cell 168, 1086–1100.e10 (2017).
    1. Colonna M Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in Immunity. Immunity 48, 1104–1117 (2018).
    1. Mazzucchelli R & Durum SK Interleukin-7 receptor expression: intelligent design. Nat. Rev. Immunol 7, 144–154 (2007).
    1. Leonard WJ & O’Shea JJ Jaks and STATs: biological implications. Annu. Rev. Immunol 16, 293–322 (1998).
    1. Mavilio D et al. Characterization of CD56–/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc. Natl. Acad. Sci. U. S. A 102, 2886–2891 (2005).
    1. Fang M et al. CD94 is essential for NK cell-mediated resistance to a lethal viral disease. Immunity 34, 579–589 (2011).
    1. Jeevan-Raj B et al. The Transcription Factor Tcf1 Contributes to Normal NK Cell Development and Function by Limiting the Expression of Granzymes. Cell Rep. 20, 613–626 (2017).
    1. Wendel M, Galani IE, Suri-Payer E & Cerwenka A Natural killer cell accumulation in tumors is dependent on IFN-gamma and CXCR3 ligands. Cancer Res. 68, 8437–8445 (2008).
    1. Sconocchia G, Titus JA & Segal DM Signaling pathways regulating CD44-dependent cytolysis in natural killer cells. Blood 90, 716–725 (1997).
    1. Liu LL et al. Critical Role of CD2 Co-stimulation in Adaptive Natural Killer Cell Responses Revealed in NKG2C-Deficient Humans. Cell Rep. 15, 1088–1099 (2016).
    1. Juelke K et al. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood 116, 1299–1307 (2010).
    1. Gazit R et al. Expression of KIR2DL1 on the entire NK cell population: a possible novel immunodeficiency syndrome. Blood 103, 1965–1966 (2004).
    1. Klein AM et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).
    1. Tibshirani R & Walther G Cluster Validation by Prediction Strength. J. Comput. Graph. Stat 14, 511–528 (2005).
    1. Trapnell C et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol 32, 381–386 (2014).
    1. Utzschneider DT et al. T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. Immunity 45, 415–427 (2016).
    1. Jeannet G et al. Essential role of the Wnt pathway effector Tcf-1 for the establishment of functional CD8 T cell memory. Proc. Natl. Acad. Sci. U. S. A 107, 9777–9782 (2010).
    1. Yang Q et al. TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow. Nat. Immunol 16, 1044–1050 (2015).
    1. Aksoy I et al. Self-Renewal of Murine Embryonic Stem Cells Is Supported by the Serine/Threonine Kinases Pim-1 and Pim-3. Stem Cells 25, 2996–3004 (2007).
    1. Baaten BJG et al. CD44 regulates survival and memory development in Th1 cells. Immunity 32, 104–115 (2010).
    1. Weng N-P, Araki Y & Subedi K The molecular basis of the memory T cell response: differential gene expression and its epigenetic regulation. Nat. Rev. Immunol 12, 306–315 (2012).
    1. Moretta L Dissecting CD56dim human NK cells. Blood vol. 116 3689–3691 (2010).
    1. Paust S et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat. Immunol 11, 1127–1135 (2010).
    1. Roychoudhuri R et al. BACH2 regulates CD8(+) T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol 17, 851–860 (2016).
    1. Shin HM et al. Epigenetic Modifications Induced by Blimp-1 Regulate CD8+ T Cell Memory Progression during Acute Virus Infection. Immunity 39, 661–675 (2013).
    1. Kamimura Y & Lanier LL Homeostatic control of memory cell progenitors in the natural killer cell lineage. Cell Rep. 10, 280–291 (2015).
    1. Brenchley JM et al. Expression of CD57 defines replicative senescence and antigen-induced apoptotic death of CD8+ T cells. Blood 101, 2711–2720 (2003).
    1. Wherry EJ et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27, 670–684 (2007).
    1. Lin W-HW et al. CD8+ T Lymphocyte Self-Renewal during Effector Cell Determination. Cell Rep. 17, 1773–1782 (2016).
    1. Vigneau S, Rohrlich P-S, Brahic M & Bureau J-F Tmevpg1, a candidate gene for the control of Theiler’s virus persistence, could be implicated in the regulation of gamma interferon. J. Virol 77, 5632–5638 (2003).
    1. Gomez JA et al. The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell 152, 743–754 (2013).
    1. Walker W, Aste-Amezaga M, Kastelein RA, Trinchieri G & Hunter CA IL-18 and CD28 use distinct molecular mechanisms to enhance NK cell production of IL-12-induced IFN-gamma. J. Immunol 162, 5894–5901 (1999).
    1. Schoenborn JR & Wilson CB Regulation of Interferon‐γ During Innate and Adaptive Immune Responses in Advances in Immunology vol. 96 41–101 (Academic Press, 2007).
    1. Björkström NK et al. Expression patterns of NKG2A, KIR, and CD57 define a process of CD56dim NK-cell differentiation uncoupled from NK-cell education. Blood 116, 3853–3864 (2010).
    1. Romee R et al. Cytokine activation induces human memory-like NK cells. Blood 120, 4751–4760 (2012).
    1. Cooper MA et al. Cytokine-induced memory-like natural killer cells. Proc. Natl. Acad. Sci. U. S. A 106, 1915–1919 (2009).
    1. Henning AN, Roychoudhuri R & Restifo NP Epigenetic control of CD8+ T cell differentiation. Nat. Rev. Immunol 18, 340–356 (2018).
    1. Hu G & Chen J A genome-wide regulatory network identifies key transcription factors for memory CD8+ T-cell development. Nat. Commun 4, 2830 (2013).
    1. O’Sullivan TE, Sun JC & Lanier LL Natural Killer Cell Memory. Immunity 43, 634–645 (2015).
    1. Xing S et al. Tcf1 and Lef1 transcription factors establish CD8(+) T cell identity through intrinsic HDAC activity. Nat. Immunol 17, 695–703 (2016).
    1. Thomas R et al. NKG2C deletion is a risk factor of HIV infection. AIDS Res. Hum. Retroviruses 28, 844–851 (2012).
    1. Fregni G et al. High number of CD56(bright) NK-cells and persistently low CD4+ T-cells in a hemophiliac HIV/HCV co-infected patient without opportunistic infections. Virol. J 10, 33 (2013).
References for Methods
    1. Davis ZB et al. A Conserved HIV-1-Derived Peptide Presented by HLA-E Renders Infected T-cells Highly Susceptible to Attack by NKG2A/CD94-Bearing Natural Killer Cells. PLoS Pathog. 12, e1005421 (2016).
    1. Neri S, Mariani E, Meneghetti A, Cattini L & Facchini A Calcein-acetyoxymethyl cytotoxicity assay: standardization of a method allowing additional analyses on recovered effector cells and supernatants. Clin. Diagn. Lab. Immunol 8, 1131–1135 (2001).
    1. Pertel T et al. TRIM5 is an innate immune sensor for the retrovirus capsid lattice. Nature 472, 361–365 (2011).
    1. Poli A et al. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology 126, 458–465 (2009).
    1. Hashimshony T et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).
    1. Skene PJ & Henikoff S An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites. Elife 6, (2017).
    1. Hainer SJ, Boskovic A, Rando OJ & Fazzio TG Profiling of pluripotency factors in individual stem cells and early embryos. bioRxiv (2018).
    1. Skene PJ, Henikoff JG & Henikoff S Targeted in situ genome-wide profiling with high efficiency for low cell numbers. Nat. Protoc 13, 1006–1019 (2018).
    1. Buenrostro JD, Wu B, Chang HY & Greenleaf WJ ATAC-seq: A Method for Assaying Chromatin Accessibility Genome-Wide. Curr. Protoc. Mol. Biol 109, 21.29.1–9 (2015).
    1. Buenrostro JD, Giresi PG, Zaba LC, Chang HY & Greenleaf WJ Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).
    1. Kim D et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).
    1. Derr A et al. End Sequence Analysis Toolkit (ESAT) expands the extractable information from single-cell RNA-seq data. Genome Res. 26, 1397–1410 (2016).
    1. Love M, Anders S & Huber W Differential analysis of count data--the DESeq2 package. Genome Biol. 15, 550 (2014).
    1. Hyvärinen A & Oja E Independent component analysis: algorithms and applications. Neural Netw. 13, 411–430 (2000).
    1. Hechenbichler K & Schliep K Weighted k-Nearest-Neighbor Techniques and Ordinal Classification. 399, (2004).
    1. Robinson MD, McCarthy DJ & Smyth GK edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).
    1. Van Der Maaten L Accelerating t-SNE using tree-based algorithms. J. Mach. Learn. Res 15, 3221–3245 (2014).
    1. Zhang Y et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).
    1. Robinson JT et al. Integrative genomics viewer. Nat. Biotechnol 29, 24–26 (2011).

Source: PubMed

3
Tilaa