Volume 21, Issue 12, Pages (December 2017)

Slides:



Advertisements
Similar presentations
Molecular Therapy - Nucleic Acids
Advertisements

M. Fu, G. Huang, Z. Zhang, J. Liu, Z. Zhang, Z. Huang, B. Yu, F. Meng 
Volume 44, Issue 1, Pages (January 2016)
Volume 21, Issue 11, Pages (December 2017)
Bart Tummers, Renske Goedemans, Veena Jha, Craig Meyers, Cornelis J. M
Volume 16, Issue 1, Pages (July 2014)
Volume 15, Issue 2, Pages (February 2014)
Volume 21, Issue 13, Pages (December 2017)
Volume 20, Issue 13, Pages (September 2017)
Volume 11, Issue 11, Pages (June 2015)
Volume 40, Issue 6, Pages (June 2014)
Expression profiling of snoRNAs in normal hematopoiesis and AML
Volume 14, Issue 3, Pages (September 2013)
Volume 20, Issue 12, Pages (September 2017)
Shiran Bar, Maya Schachter, Talia Eldar-Geva, Nissim Benvenisty 
Kobe C. Yuen, Baoshan Xu, Ian D. Krantz, Jennifer L. Gerton 
Volume 20, Issue 3, Pages (July 2017)
Volume 19, Issue 3, Pages (April 2017)
Role of monocytes in NK cell activity.
Volume 23, Issue 4, Pages (April 2018)
Transcriptional Landscape of Cardiomyocyte Maturation
Volume 31, Issue 2, Pages (February 2017)
Volume 21, Issue 1, Pages (October 2017)
Volume 33, Issue 4, Pages e6 (April 2018)
Dynamic Gene Regulatory Networks of Human Myeloid Differentiation
Volume 18, Issue 5, Pages (January 2017)
Transcriptional Profiling of Quiescent Muscle Stem Cells In Vivo
Volume 9, Issue 5, Pages (November 2017)
Volume 15, Issue 8, Pages (May 2016)
Volume 8, Issue 4, Pages (April 2017)
Volume 12, Issue 6, Pages (December 2003)
Volume 18, Issue 1, Pages (January 2017)
Volume 33, Issue 6, Pages (December 2010)
Volume 24, Issue 11, Pages e3 (September 2018)
Volume 21, Issue 11, Pages (December 2017)
Volume 23, Issue 7, Pages (May 2018)
An RpaA-Dependent Sigma Factor Cascade Sets the Timing of Circadian Transcriptional Rhythms in Synechococcus elongatus  Kathleen E. Fleming, Erin K. O’Shea 
Volume 21, Issue 6, Pages (November 2017)
Volume 22, Issue 9, Pages (February 2018)
Volume 16, Issue 11, Pages (September 2016)
Volume 21, Issue 13, Pages (December 2017)
Wei Jiang, Yuting Liu, Rui Liu, Kun Zhang, Yi Zhang  Cell Reports 
Volume 9, Issue 5, Pages (November 2017)
Volume 64, Issue 6, Pages (December 2016)
Signatures of the Immune Response
Volume 11, Issue 11, Pages (June 2015)
Shiran Bar, Maya Schachter, Talia Eldar-Geva, Nissim Benvenisty 
Volume 22, Issue 3, Pages (January 2018)
Volume 14, Issue 2, Pages (January 2016)
Volume 27, Issue 5, Pages (November 2007)
Alterations in mRNA 3′ UTR Isoform Abundance Accompany Gene Expression Changes in Human Huntington’s Disease Brains  Lindsay Romo, Ami Ashar-Patel, Edith.
Global Hypertranscription in the Mouse Embryonic Germline
Volume 39, Issue 5, Pages (November 2013)
Volume 11, Issue 11, Pages (June 2015)
Zika Virus Infects Human Placental Macrophages
Urtzi Garaigorta, Francis V. Chisari  Cell Host & Microbe 
Volume 21, Issue 4, Pages (October 2017)
Volume 1, Issue 1, Pages (July 2015)
Effects of a Single Escape Mutation on T Cell and HIV-1 Co-adaptation
Volume 56, Issue 6, Pages (December 2014)
Volume 15, Issue 12, Pages (June 2016)
Differential protein, mRNA, lncRNA and miRNA regulation by p53.
Volume 9, Issue 5, Pages (November 2017)
Maria S. Robles, Sean J. Humphrey, Matthias Mann  Cell Metabolism 
Volume 2, Issue 3, Pages (March 2016)
Volume 10, Issue 7, Pages (February 2015)
Genome-wide Functional Analysis Reveals Factors Needed at the Transition Steps of Induced Reprogramming  Chao-Shun Yang, Kung-Yen Chang, Tariq M. Rana 
Volume 12, Issue 12, Pages (September 2015)
Volume 18, Issue 5, Pages (January 2017)
Volume 20, Issue 2, Pages (August 2016)
Presentation transcript:

Volume 21, Issue 12, Pages 3471-3482 (December 2017) Transcriptional Changes during Naturally Acquired Zika Virus Infection Render Dendritic Cells Highly Conducive to Viral Replication  Xiaoming Sun, Stephane Hua, Hsiao-Rong Chen, Zhengyu Ouyang, Kevin Einkauf, Samantha Tse, Kevin Ard, Andrea Ciaranello, Sigal Yawetz, Paul Sax, Eric S. Rosenberg, Mathias Lichterfeld, Xu G. Yu  Cell Reports  Volume 21, Issue 12, Pages 3471-3482 (December 2017) DOI: 10.1016/j.celrep.2017.11.087 Copyright © 2017 Terms and Conditions

Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions

Figure 1 ZIKV Replication in In-Vitro-Infected Cell Subsets from Adult PBMCs and Cord Blood (A) PBMCs and CBMCs were infected with ZIKV at an MOI of 1, and viral replication was measured by qRT-PCR at indicated hours post-infection. Positive-strand viral RNA expression is shown after normalization to beta-actin (n = 10). (B) Comparison between ZIKV replication in in-vivo-infected PBMCs (blue) and CBMCs (red). Data indicate mean ± SD from 10 donors (n = 10). (C) ZIKV replication in sorted cell populations from PBMCs and CBMCs after infection for 24 hr with ZIKV at an MOI of 1 (n = 9). Mono, monocyte. (D) Comparison between ZIKV replication in B cells (orange), NK cells (pink), and mDCs (purple) from PBMCs and CBMCs (n = 9). (E) Analysis of ZIKV replication in pDCs and mDCs at indicated time points. (F) Analysis of negative-strand ZIKV RNA in pDCs and mDCs from PBMCs and CBMCs, as determined by qPCR at 24 hr post infection (p.i.). Horizontal lines reflect the median. ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions

Figure 2 Profound Changes in the Global Transcriptional Landscape of mDCs Infected with ZIKV In Vivo (A) Principal-component analysis (PCA) of RNA-seq data from mDCs isolated from three patients with acute ZIKV infection (red dots) and 5 healthy donors in mDCs (gray dots). The expression values are normalized across the entire dataset. (B) Hierarchical clustering and heatmap of genes that are differentially expressed (DEGs) between ZIKV-infected patients and healthy donors. Displayed DEGs had log2 FC ≥ 1.5 fold changes in gene expression intensity and an FDR-adjusted p value < 0.05, relative to the control cohort. (C) Volcano plot displaying genes detected by RNA-seq. Pink dots represent genes that show an FDR-adjusted p < 0.05; red dots indicate genes with an FDR-adjusted p < 0.05 and log2 FC > 1.5. (D) Functional annotations of differentially expressed genes, as determined using Ingenuity Pathway Analysis. Negative Z scores indicate decreased functional activity (blue); gray bars reflect Z scores with unknown functional activity changes. (E) Zika viral genome coverage detected by RNA-seq in in-vitro-infected mDCs at indicated hours post-infection. Alignment was performed using bedtools software. (F) Waterfall plot representing the total number of upregulated and downregulated genes between in-vitro-infected mDCs and their respective controls at indicated time points. Transcripts with a nominal p value < 0.05 were considered as DEGs. (G) Predicted functional pathways of indicated DEGs, as analyzed by Ingenuity Pathway Analysis. Z scores > 2 indicate functional pathway activation (red), whereas negative Z scores indicated pathway de-activation (blue). n.s, not significant; n.d, not detected. (H) Venn diagram indicates shared DEGs between in-vivo- and in-vitro-infected mDC samples; heatmap represents an expression pattern of 199 genes that were differentially expressed in both in-vivo- and in-vitro-infected mDCs. (I) Predicted functional pathways of shared 199 DEGs analyzed by Ingenuity Pathway Analysis. Directional changes in gene expression intensity were not determined in this analysis. Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions

Figure 3 ZIKV Infection Leads to Downregulation of IFN-Stimulated Genes and Impaired Dendritic Cell Function (A) Heatmap representing IFN-stimulated genes (ISGs) that were differentially expressed between mDCs isolated from ZIKV-infected patients and healthy controls. (B) Volcano plot reflecting all differentially expressed ISGs (FDR-adjusted p < 0.05 and log2 FC > 1.5, blue dots). Differentially expressed ISGs with known antiviral function are highlighted in orange. (C) Heatmap reflecting expression intensity of a known list of antiviral ISGs during in vivo and in vitro infection of mDCs with ZIKV. (D and E) Changes in mRNA expression of IFN-α or IFN-β from (D) in-vivo-infected compared to healthy donors and from (E) in-vitro-infected normalized to uninfected cells. (F) Surface expression of indicated dendritic cell immune activation (CD83) and maturation markers (CD80 and CD86) at 0 hr, 24 hr, and 48 hr post-infection (+). Data from uninfected controls (-) are shown for comparison. M.F.I., median fluorescence intensity. Horizontal lines reflect the median. ∗p < 0.05; ∗∗p < 0.01. Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions

Figure 4 Upregulation of Viral Dependency Genes during Natural ZIKV Infection (A) Pie chart of the viral dependency genes reported by Savidis et al. (2016a). Colors indicate expression intensity of these genes in mDCs from ZIKV-infected patients or healthy donors (left panel). Heatmaps reflect the relative ratio of gene expression intensity between ZIKV-infected patients and controls for 169 viral dependency genes meeting the criteria for DEGs and for 502 dependency genes that were expressed in mDCs from ZIKV patients and controls but did not meet the criteria for DEGs (right panel). (B) Venn diagram reflecting overlay between DEGs in in-vivo- and in-vitro-infected mDCs, and ZIKV dependency genes reported by Savidis et al. (2016a). Numbers of ISGs are listed in parentheses. Gene identities of the 10 transcripts detected in all three gene sets are listed; the green box highlights ISGs. (C) Heatmap representing gene expression changes of the viral dependency genes (n = 10) with differential gene expression in both in-vitro- and in-vivo-infected mDCs described in (B). Color code reflects the fold changes in ZIKV-infected samples relative to corresponding controls. (D) Fold changes of SOCS3 and AXL expression following siRNA-mediated gene silencing relative to control samples (left and middle panels) and intensity of reactive oxidative species (ROS) production following the inhibition of IDO-1 activity (right panel). M.F.I., median fluorescence intensity. (E) Monocyte-derived DCs were pretreated with SOCS3 siRNA or AXL siRNA for 48 hr or pretreated with 100 μM of the IDO1 inhibitor (NLG919) for 2 hr. Cells were then infected with ZIKV at an MOI of 1. Negative-strand Zika RNA was measured after 24 hr p.i. and normalized to untreated controls. Horizontal lines reflect the median. ∗p < 0.05; ∗∗p < 0.01. Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions

Figure 5 AXL, SOCS3, and IDO-1 Alter Type I IFN Responses during ZIKV Infection (A) Expression of IFN-α after 24 hr post-infection with ZIKV in MDDCs manipulated as described in Figure 4E. Fold changes normalized to untreated controls are shown. (B) Correlations between the expression levels of IFN-α and the corresponding ZIKV negative-strand RNA levels after silencing SOCS3 (left; n = 10), silencing AXL (middle; n = 7), or the addition of the IDO1 inhibitor NLG99 (right; n = 7). Cumulative data were analyzed using generalized estimated equations adjusted for repeated measures. (C and D) Volcano plots reflecting changes in the expression of host genes involved in flavivirus pathogenesis following downregulation of SOCS3 (C) or inhibition of IDO1 (D). Differentially expressed genes (nominal p < 0.05) are indicated in red. Heatmaps reflect gene expression changes of the differentially expressed genes; color coding indicates gene expression intensity. S1, sample 1. Horizontal lines reflect the median. ∗p < 0.05. Cell Reports 2017 21, 3471-3482DOI: (10.1016/j.celrep.2017.11.087) Copyright © 2017 Terms and Conditions