Abstract
Background
Psoriasis patients have relatively infrequent cutaneous viral infections compared to atopic dermatitis patients. Increased expression of four antiviral proteins (MX1, BST2, ISG15, and OAS2) has been reported in psoriatic skin and genetic studies of psoriasis have identified susceptibility genes in antiviral pathways.
Objective
To determine if psoriasis is associated with pervasive expression of antiviral genes in skin and blood.
Methods
We performed RNA-sequencing on skin samples of 18 subjects with chronic plaque psoriasis and 16 healthy controls. We examined the expression of a pre-defined set of 42 antiviral genes, each of which has been shown in previous studies to inhibit viral replication. In parallel, we examined antiviral gene expression in atopic dermatitis, non-lesional psoriatic skin, and psoriatic blood. We performed HIV-1 infectivity assays in CD4+ peripheral blood T cells from psoriatic and healthy individuals.
Results
We observed significant overexpression of 16 antiviral genes in lesional psoriatic skin, with a greater than two-fold increase in ISG15, RSAD2, IRF7, MX2, and TRIM22 (p<1E-07). None of these genes was overexpressed in atopic dermatitis skin (p<0.0001) or non-lesional psoriatic skin. In contrast to the skin compartment, no differences in antiviral gene expression were detected in the peripheral blood of psoriasis cases compared to healthy controls. CD4+ T cells from both psoriatic and healthy patients supported HIV-1 infection at a similar rate.
Conclusion
Our findings highlight psoriasis as an inflammatory disease with cutaneous but not systemic immune activation against viral pathogens.
Introduction
Autoimmune and immune-mediated diseases arise from aberrant activation of host immune pathways. However, whether particular autoimmune diseases activate pathways specific for certain classes of pathogens remains unclear. Psoriasis, an immune-mediated inflammatory disease of the skin, has been recently associated with increased cutaneous expression of select antiviral proteins,1 which may explain the relative infrequency of cutaneous viral infections observed in these patients. Genetic studies performed by our group have shown that certain psoriasis susceptibility alleles such as HLA-B*57 are also associated with host control of HIV-1 infection.2 Moreover, a number of psoriasis susceptibility variants identified by genome-wide association studies (GWAS) map to genes associated with the innate antiviral response.3 Such genes include IFIH1 (MDA5) and DDX58 (RIG-I), which code for innate antiviral receptors recognizing cytosolic viral RNA, and RNF114, which regulates MDA5/RIG-I signaling.4 Therefore, it is of interest to determine whether psoriasis is associated with a systemic antiviral phenotype.
“Restriction factors” are innate antiviral proteins that are produced in the host and oppose or “restrict” viral replication. Our current knowledge of restriction factors stems from the large body of studies performed in HIV virology.5 The discovery of restriction factors and their antiviral nature is relatively recent, but has already been implicated in many host mechanisms of antiviral defense. Examples include BST2/Tetherin,6, 7 which blocks the release of enveloped viruses; SAMHD1,8 which is thought to inhibit viral replication by reducing the cellular levels of dNTP and mediating pro-inflammatory responses to TNF-alpha signaling; and TRIM5,9 which blocks the uncoating of viral capsid. The different expression levels of restriction factors between cell types may explain the varying levels of permissiveness of different cell types to HIV-1 infection.
We have recently developed a custom-made quantitative real-time PCR array to characterize the expression of different restriction factors in primary human cells10, 11 and have also correlated their expression with HIV-1 infectivity.12 A comprehensive table of host antiviral genes and their functions is depicted in Table 1.
Table 1.
NAME | GENE DESCRIPTION | KEY ANTI-HIV-1 ROLE(S) | REFS |
---|---|---|---|
APOBEC3 (A,B,C,D,F,G,H) | Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like3 | Hypermutation; lethal mutations in viral DNA Inhibition of reverse transcription Inhibition of integration |
26–34 |
BRD4 | Bromodomain-containing protein 4 | Transcriptional repression of HIV-1 | 35 |
BST-2/Tetherin | Bone marrow stromal cell antigen 2 | Blocks release of enveloped viruses | 6, 7 |
CD74 | HLA-DR antigens-associated invariant chain or CD74 (Cluster of Differentiation 74) | Inhibits viral replication in a cellular overexpression assay | 36 |
CDKN1A (P21) | Cyclin-dependent kinase inhibitor 1A (p21, Cip1) | Blocks reverse transcription Blocks RNA transcription by reducing activity of CDK9 |
37, 38 |
CH25H | Cholesterol 25-hydroxylase | Inhibits viral entry. | 39 |
CNP | 2′,3′-Cyclic Nucleotide 3′ Phosphodiesterase | Blocks HIV-1 particle assembly. | 40 |
CTR9 | Ctr9, Paf1/RNA polymerase II complex component | Inhibits early events of viral life cycle from reverse transcription to integration | 41 |
EIF2AK2 (PKR) | Eukaryotic translation initiation factor 2-alpha kinase 2 | Inhibits viral protein translation by protein phosphorylation; promotes innate immune signaling | 42 |
HERC5 | HECT domain and RLD 5 | Blocks early stage of retroviral particle assembly | 43 |
IFITM Family (3 members) | Interferon induced transmembrane protein | Inhibition of cytosolic entry | 43 |
IRF1 | Interferon regulatory factor 1 | Inhibits viral replication in a cellular overexpression assay | 36 |
IRF7 | Interferon regulatory factor 7 | Inhibits viral replication in a cellular overexpression assay | 36 |
ISG15 | ISG15 ubiquitin-like modifier | Blocks interaction between HIV-1 Gag and Tsg101 (ESCRT-I) required for efficient budding of HIV-1 | 44 |
LGALS3BP | lectin, galactoside-binding, soluble, 3 binding protein | Interferes with the maturation and incorporation of HIV-1 Env molecules into virions. | 45 |
MOV10 | Mov10, Moloney leukemia virus 10, homolog | Inhibits proteolytic processing of Gag and reverse transcription | 46 |
MX2 | Myxovirus resistance 2 | Inhibits capsid-dependent nuclear import of subviral complexes. | 47–49 |
PAF1 | Paf1, RNA polymerase II associated factor | Inhibits early events of viral life cycle from reverse transcription to integration | 41 |
RNASEL | Ribonuclease L (2′,5′-oligoisoadenylate synthetase-dependent) | Cleave single-stranded RNA in U-rich sequences; activate antiviral innate immunity | 50 |
RSAD2 (Viperin) | Radical S-adenosyl methionine domain containing 2 | Inhibits viral production | 51 |
RTF1 | Rtf1, Paf1/RNA polymerase II complex component | Inhibits early events of viral life cycle from reverse transcription to integration | 41 |
SAMHD1 | SAM domain and HD domain 1 | Inhibits HIV replication in myeloid cells, probably by regulating cellular dNTP supply | 8 |
SLFN11 | Schlafen family member 11 | Inhibits viral protein synthesis | 52 |
SUN2 | Sad1 and UNC84 domain containing 2 | Inhibits viral replication in a cellular overexpression assay | 36 |
TNFRSF10A | Tumor necrosis factor receptor superfamily, member 10a | Inhibits viral replication in a cellular overexpression assay | 36 |
TRIM family (9 members) | Tripartite motif family | Targeting of viral capsid Inhibition of viral transcription |
9, 53–55 |
In this report, we investigated the expression of a large panel of antiviral genes in psoriatic skin and blood. To determine whether psoriasis is associated with systemic antiviral immunity against HIV-1, we performed ex vivo HIV-1 replication assays in purified CD4+ T cells from individuals with psoriasis and healthy controls.
Materials and Methods
Samples
Eighteen adult subjects with chronic, plaque psoriasis with affected body surface area > 10% and not on systemic medications were recruited from the University of California San Francisco Dermatology Department by a board-certified dermatologist. Five millimeter punch biopsies were taken from the edge of a psoriatic plaque. Sixteen normal skin samples were obtained from healthy control surgical discard specimens. All subjects provided written, informed consent for study participation under the approval of local Institutional Review Boards. Skin samples were stored in RNALater at −80°C, mechanically homogenized using a Bio-Gen Pro 200 homogenizer, and total RNA was extracted using the Qiagen RNeasy mini kit. The quality of the RNA was assessed using an Agilent 2100 Bioanalyzer. Ribosomal RNA was depleted using Ribo-Zero Gold rRNA removal kit, followed by cDNA synthesis and library preparation using ScriptSeq Complete kits (Human/Mouse/Rat) from Epicentre. The quality and quantity of library was assessed by Agilent 2100 Bioanalyzer.
Human primary peripheral blood mononuclear cells (PBMC) were isolated by the use of a Ficoll gradient from collected blood obtained from psoriasis patients (N=32) and healthy control individuals (N=25). After processing, PBMC were stored and frozen in 10% DMSO-FCS prior to subsequent analysis. Based on cell numbers and patient availability, a subset of samples was destined for ex vivo HIV-1 infectivity assays and CD4+ T cells were enriched from PBMCs using the EasySep Human CD4+ T cell enrichment magnetic kit (Stem Cell Technologies), according to manufacturer’s instructions.
RNA-sequencing gene expression in psoriatic skin and healthy control skin
RNA-seq on blinded samples was performed on an Illumina HiSeq 2500. We obtained an average of 52.3 million 101 bp paired end reads per sample. Quality was validated using FastQC. Quality and adapter trimming was performed with Trimmomatic (version 0.3). Reads were aligned to the human genome (hg19) using Tophat2 (version 2.0.9). An average of 34.8 million paired end reads were aligned for each library (minimum of 25.3 million, maximum of 61.4 million). For differential gene expression, the RefSeq gene set (UCSC genome browser, January 2013) was used as the genome annotation. Differential gene expression was calculated using EdgeR13, with counts per gene measured using htseq-count and analyzed as described.
Microarray gene expression analysis in lesional and non-lesional psoriatic skin, control skin, and atopic dermatitis skin
We validated top hits using a publically available, independent dataset (Gene Expression Omnibus dataset GSE13355) consisting of 58 lesional psoriasis skin samples, 58 non-lesional psoriasis skin samples, and 64 healthy control samples profiled using the Affymetrix HU133 Plus 2.0 Microarray containing >54,000 gene probes. The raw microarray data were processed using the Robust Multichip Average (RMA) method. Differential gene expression was calculated using the limma (Linear Models for Microarray Analysis) R package, which applies multiple-testing corrections on p-values to help correct for the occurrence of false positives.
We also examined the same set of antiviral genes in Affymetrix HU133A microarray data from atopic dermatitis skin (N=10) and healthy control skin (N=10) (Gene Expression Omnibus dataset GSE6012). Differential gene expression was calculated as above for psoriatic skin.
Real-time quantitative PCR from PBMCs
PBMC samples were quickly thawed and total RNA was extracted using Qiazol reagent from miRNeasy Mini kit (Qiagen) with the on-column DNAase treatment option using Qiagen RNase-Free DNase Set. DNase-treated clean RNA was transcribed into cDNA using random primers and the SuperScript VILO™ cDNA Synthesis Kit (Invitrogen) according to manufacturer’s instructions. Quantitative real-time PCR on blinded samples utilized custom made TaqMan Low Density Array (TLDA) from Applied Biosystems and followed the manufacturer’s instructions. Thermal cycling was performed using an ABI ViiA7 Real-Time PCR System. Up to 350ng cDNA in 200μl of Applied Biosystems TaqMan Universal PCR Master Mix, with UNG was loaded onto the designated ports of the TLDA plates. Data was analyzed using ABI ViiA7 software. A panel of 6 housekeeping genes was included in the TLDA plates (GAPDH, 18S, ACTB, PPIA, RPLP0, and UBC). RPLP0 was identified as the most stably expressed gene from those 6 housekeeping genes among the whole samples using the GeNorm algorithm.14 Therefore, raw cycle threshold numbers of amplified gene products were normalized to the housekeeping gene, RPLP0, (Ribosomal protein, large, P0) to control for cDNA input amounts. Fold induction was determined using the comparative Ct method.
Ex vivo HIV-1 infectivity assay
pNL-LucR.T2A-Bal.ecto was a kind gift from Dr. Christina Ochsenbauer (University of Alabama at Birmingham). The plasmid encodes an infectious molecular clone that has Tat-regulated Renilla luciferase (rLuc is between Env and Nef) and it contains the entire ectodomain and a portion of the transmembrane region of Bal.15 Briefly, viral stocks were generated by transfecting proviral DNA into 293T cells using FuGENE6 according to manufacturer’s instructions. Viral supernatants were harvested 60h post-transfection, span and filtered through a 0.45μm pore size, before being frozen at −80°C.
On the day before infection, CD4+ T cells were enriched from frozen PBMCs, as described above and placed over-night in complete media (RPMI1640, 10% FCS, Pen/Strep and L-glutamine). The next day, a viable cell count was performed and 100,000 CD4+ T cells were infected in duplicate for 7 days in the presence of 50 U/ml IL-2 in a total volume of 200μl in U-bottom 96-well plates. Mock infections were also performed. After 7 days of infection, cells were lysed in 1x Renilla Luciferase assay lysis buffer and 50μl of each cell lysate was transferred to a solid-white flat bottom 96-well plate and analysed for LucR activity. Samples were analysed using a Veritas luminiometer (Promega) programed to inject 100μl of LucR assay reagent per well with an integration time of 2.5s/well and reporting relative light units (RLU).
Results
Elevated antiviral gene expression in psoriatic but not atopic dermatitis skin
We selected a panel of 42 antiviral genes for investigation in this study; the function and activity of these genes have been documented in the literature (Table 1).
To evaluate the expression of these genes in psoriasis, we obtained punch biopsies from the edge of a psoriatic plaque in subjects (N=18) with chronic psoriasis not on systemic medications. In parallel, normal skin samples (N=16) were obtained from healthy control surgical discard specimens. RNA-seq was performed using an Illumina HiSeq 2500 resulting in an average of 52 million paired-end reads per sample.
We observed statistically significant up-regulation of 16 of 42 antiviral genes (Table 2), including ISG15 (9.93 fold increase, p=1.05E-25), RSAD2/Viperin (9.95 fold increase, p = 2.11E-20), TRIM21 (2.21 fold increase, p = 8.68E-17), IRF7 (3.40-fold increase, p = 3.60E-14) and MX2 (3.52-fold increase, p = 9.25E-12). In contrast, SLFN11 (0.64-fold decrease, p=2.44E-04), TRIM32 (0.75-fold decrease, p = 9.97E-04), SUN2 (0.76-fold decrease, p=0.001), and BRD4 (0.72-fold decrease, p = 0.020) were among the genes that were down-regulated in psoriatic skin. Several members of the APOBEC3-family (A3A, A3B, A3D, A3G and A3H) were not differentially expressed between psoriatic and healthy skin.
Table 2. Antiviral restriction factor expression in psoriatic versus healthy skin.
RNA-Seq | Microarray | |||||
---|---|---|---|---|---|---|
Gene | Fold change | P-Value | Fold change | P-Value | Confirm | Description |
ISG15 | 9.93 | 1.05E-25 | 5.43 | 6.10E-30 | Yes | ubiquitin-like protein ISG15 |
RSAD2 | 9.95 | 2.11E-20 | 5.28 | 1.10E-24 | Yes* | radical S-adenosyl methionine domain-containing protein 2 |
TRIM21 | 2.21 | 8.68E-17 | 1.38 | 6.73E-15 | Yes | tripartite motif containing 21 |
IRF7 | 3.40 | 3.60E-14 | 4.56 | 8.16E-47 | Yes | interferon regulatory factor 7 |
MX2 | 3.52 | 9.25E-12 | 2.25 | 6.24E-16 | Yes | interferon-induced GTP-binding protein Mx2 |
CDKN1A | 0.43 | 9.72E-10 | 1.04 | 3.72E-02 | No* | cyclin-dependent kinase inhibitor 1 |
TRIM22 | 2.10 | 5.69E-08 | 2.30 | 3.36E-29 | Yes | tripartite motif containing 22 |
BST2 | 2.09 | 9.38E-08 | 1.58 | 1.64E-08 | Yes | bone marrow stromal cell antigen 2 |
EIF2AK2 | 1.63 | 1.72E-07 | 1.19 | 6.84E-07 | Yes* | eukaryotic translation initiation factor 2-alpha kinase 2 |
TRIM14 | 1.78 | 3.11E-07 | 2.95 | 4.32E-48 | Yes* | tripartite motif-containing protein 14 |
HERC5 | 1.84 | 3.75E-07 | 1.47 | 5.34E-10 | Yes | E3 ISG15--protein ligase HERC5 |
CH25H | 0.38 | 5.26E-07 | 2.16 | 6.61E-25 | No | cholesterol 25-hydroxylase |
SAMHD1 | 1.58 | 7.08E-06 | 1.49 | 8.01E-10 | Yes* | SAM domain and HD domain 1 |
PAF1 | 1.54 | 2.31E-05 | 1.15 | 3.58E-10 | Yes | Paf1, RNA Polymerase II Associated Factor, Homolog |
IFITM1 | 1.53 | 9.27E-05 | 1.55 | 8.92E-22 | Yes* | interferon-induced transmembrane protein 1 (9–27) isoform 1 |
TRIM5 | 1.46 | 1.87E-04 | 1.38 | 7.61E-09 | Yes* | tripartite motif-containing protein 5 |
IFITM3 | 1.61 | 2.05E-04 | 1.35 | 3.48E-18 | Yes | interferon induced transmembrane protein 3 |
SLFN11 | 0.64 | 2.44E-04 | 0.84 | 2.60E-03 | Yes | schlafen family member 11 |
TRIM32 | 0.75 | 9.97E-04 | 0.82 | 6.00E-07 | Yes* | E3 ubiquitin-protein ligase TRIM32 |
SUN2 | 0.76 | 0.001 | 0.87 | 1.38E-09 | Yes | SAD1/UNC-84 domain-containing protein 2 |
IRF1 | 0.68 | 0.002 | 1.91 | 2.91E-27 | No* | interferon regulatory factor 1 |
APOBEC3C | 0.75 | 0.015 | 1.17 | 1.18E-05 | No | probable DNA dC->dU-editing enzyme APOBEC-3C |
BRD4 | 0.72 | 0.020 | 0.79 | 4.67E-19 | Yes* | bromodomain-containing protein 4 |
APOBEC3F | 0.76 | 0.023 | 1.18 | 1.73E-05 | No* | DNA dC->dU-editing enzyme APOBEC-3F |
TRIM26 | 1.20 | 0.033 | 1.04 | 1.25E-01 | No | tripartite motif-containing protein 26 |
Indicates more than one microarray probe present per gene. Data are from most significant probe displayed.
Genes not differentially expressed (p > 0.05) by RNA-Seq: APOBEC3A, APOBEC3B, APOBEC3D, APOBEC3G, APOBEC3H, CD74, CNP, CTR9, IFITM2, LGALS3BP, MOV10, PML, RNASEL, RTF1, TNFRSF10A, TRIM11, TRIM28
To confirm our RNA-seq results, we validated top hits using an independent dataset of 58 lesional psoriasis skin samples and 64 healthy control samples profiled using the Affymetrix HU133 Plus 2.0 Microarray containing over 54,000 gene probes. We found that 18 out of our 20 most significant hits were replicated (Table 2).
To determine whether the differential antiviral gene expression observed in psoriatic skin was due to generalized skin inflammation, we examined the same set of antiviral genes in atopic dermatitis skin (N=10) and healthy control skin (N=10) profiled on the Affymetrix HU133A microarray data. Atopic dermatitis is an inflammatory skin disorder characterized clinically by erythema and itch and histologically by a lymphocytic infiltrate and the presence of intercellular edema. Among the 36 target genes for which data were available in the atopic dermatitis dataset, only 6 genes were differentially expressed (p < 0.05) between atopic dermatitis skin and healthy control skin (APOBEC3A, CH25H, IFITM1, IRF7, ISG15 and LGALS3BP). All six genes showed significant under-expression in atopic dermatitis skin (Supplementary Table 1). This contrasted with psoriasis in which a significant number of antiviral genes were elevated compared to control skin (16 of 42 in psoriasis vs 0 of 36 in atopic dermatitis, p < 0.0001, Fisher’s exact test). Furthermore, we examined antiviral gene expression in non-lesional psoriatic skin and found none to be significantly elevated (Supplementary Table 1). These data suggest that lesional psoriatic skin exhibits a selective overexpression of antiviral genes that is not a general feature of inflammatory skin disease or of non-lesional psoriatic skin.
No antiviral signature in PBMC from psoriatic patients
Since psoriatic skin was enriched for antiviral genes, we investigated whether peripheral blood mononuclear cells (PBMC) from psoriatic patients might also possess increased expression of host restriction factors. We obtained PBMC from psoriatic patients (N=32) and healthy control individuals (N=25) and quantified the expression of 42 different antiviral genes using quantitative real-time PCR.
In contrast with the increased gene expression levels detected in the skin of psoriatic patients, PBMC from psoriatic patients did not exhibit statistically significant increase in the expression of antiviral genes compared to healthy controls (Supplementary Table 2). These data suggest a compartmentalization of the antiviral gene signature in psoriasis whereby expression of antiviral genes is up-regulated in skin but not in peripheral blood.
Ex vivo HIV infectivity assay
We then investigated whether peripheral blood CD4+ T cells from psoriatic patients would support HIV-1 replication at the same rate as control CD4+ T cells. Based on cell availability, we infected CD4+ T cells from psoriatic patients (N=7) and healthy control individuals (N=10) with HIV-1BaL encoding Renilla Luciferase (LucR) reporter gene. After 7 days of infection we quantified LucR activity using a luminometer and detected no statistically significant differences in HIV-1 infectivity in CD4+ T cells from psoriatic and healthy individuals (p > 0.05, Mann-Whitney) (Supplementary Figure 2).
Discussion
Here we have examined a large panel of antiviral genes in psoriasis and identified overexpression of innate antiviral genes in psoriatic skin. Our findings were determined using two independent cohorts and two technical approaches: RNA sequencing and microarrays.
Our findings build on a previous observation that four antiviral proteins, MX1, BST2, ISG15, and OAS2, are increased in psoriatic skin and induced by IL-29.1 The type I interferon signaling pathway is activated in psoriatic lesional skin16 and the blocking of IFN-alpha signaling or blocking the ability of plasmacytoid dendritic cells to produce IFN-alpha prevented the development of disease in a xenograft model of human psoriasis.17 The significant up-regulation of antiviral genes in psoriatic skin is in agreement with the role of interferon-alpha in inducing antiviral gene expression. Moreover, the two most up-regulated genes in our study, ISG15 and RSAD2/Viperin, are strongly induced by exogenous interferon-alpha in vivo.18
We did not observe the same differential expression of antiviral genes in atopic dermatitis skin. Psoriasis, a Th1 and Th17 driven disease, activates distinct immune pathways compared to atopic dermatitis, a Th2 driven disease. Furthermore, psoriasis more than atopic dermatitis is associated with expansion of keratinocytes and it is possible that antiviral genes are expressed robustly in keratinocytes. However, prior studies in HIV (Table 1) have shown innate antiviral genes to be highly expressed in immune cells; thus our results may reflect a combination of antiviral activity in both immunocytes and keratinocytes.
In chronic HIV-1 infection, CD4+ T cell activation is elevated in viremic non-controller patients and it gradually decreases from ART-suppressed to elite controllers; it is low in HIV-1 seronegative individuals.19 We have previously reported a strong correlation between the levels of CD4+ T cell activation and the expression of antiviral genes.10, 20 Curiously, we did not observe elevated antiviral gene expression in psoriatic peripheral blood, despite the increased activation levels previously shown in psoriatic PBMCs.21 It is possible that psoriatic autoantigens are expressed primarily in the skin and thus the observed antiviral immune responses localize preferentially in skin.
An important clinical question is whether psoriasis patients have enhanced immunity to systemic viral infections. Here, we specifically examined the effect of psoriasis on infection by HIV-1. Addition of HIV-1 reporter virus to psoriatic and non-psoriatic peripheral CD4+ T cells did not reveal any differences in viral replication. This suggests the antiviral phenotype of psoriatic skin does not impact viral replication within the blood. In agreement, we observed no significant difference in the viral loads of HIV positive patients with psoriasis and without psoriasis (see companion manuscript, Wu et al).
A limitation of the present study is that psoriasis is a genetically heterogeneous disease and with our sample size we were not able to stratify psoriasis patients by genetic subtype. For example, only about 25% of psoriasis patients in the North American population are positive for the psoriasis susceptibility allele HLA-B*57, which is also associated with control of both HIV-122 and hepatitis C virus.23 We have previously shown that the presence of the HLA-B*57 allele is associated with increased expression of antiviral restriction factors in the PBMCs of healthy individuals.11 A second limitation to this study is that we did not perform HIV-1 infectivity assays on CD4+ T cells derived from psoriatic skin, as it is technically difficult to obtain a requisite numbers of cells from skin. Thus, we were not able to directly ascertain whether the high antiviral gene expression in psoriatic skin could impact HIV-1 replication. However, it has been clinically observed that psoriasis patients have decreased occurrence of cutaneous bacterial and viral skin infections compared to atopic dermatitis patients24 and have significant overproduction of antimicrobial peptides with both antibacterial and antiviral activity.25
Our findings highlight psoriasis as an immune-mediated, inflammatory disease with compartmentalized immune activation against viral pathogens.
Supplementary Material
Acknowledgments
Funding sources: This work was supported by grants from the National Institutes of Health (R01AR065174 and K08AR057763 to W.L.; AI093179 to D.F.N.; and 1K01DA024654 to S.K.P.), the Peter and Shelagh Godsoe Family Foundation thorough the AIDS Research Institute at UCSF (to D.F.N. and P.J.K.), the UCSF-Gladstone Institute of Virology & Immunology Center for AIDS Research (P30-AI027763 to R.A.R.), and in part from grants provided by the Academic Senate under UCSF RAP awards (to D.F.N. and to P.J.K.). Further funding was from the Creative and Novel Ideas in HIV Research Program (to W.L.) through a supplement to the University of Alabama at the Birmingham Center For AIDS Research funding (P30 AI027767-24), made possible by collaborative efforts of the Office of AIDS Research, NIAID, and the International AIDS Society.
Footnotes
Conflict of Interest: The authors state no conflict of interest.
References
- 1.Wolk K, Witte K, Witte E, et al. IL-29 is produced by T(H)17 cells and mediates the cutaneous antiviral competence in psoriasis. Science translational medicine. 2013;5:204ra129. doi: 10.1126/scitranslmed.3006245. [DOI] [PubMed] [Google Scholar]
- 2.Chen H, Hayashi G, Lai OY, et al. Psoriasis patients are enriched for genetic variants that protect against HIV-1 disease. PLoS Genet. 2012;8:e1002514. doi: 10.1371/journal.pgen.1002514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Tsoi LC, Spain SL, Knight J, et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nature genetics. 2012;44:1341–1348. doi: 10.1038/ng.2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bijlmakers MJ, Kanneganti SK, Barker JN, Trembath RC, Capon F. Functional analysis of the RNF114 psoriasis susceptibility gene implicates innate immune responses to double-stranded RNA in disease pathogenesis. Hum Mol Genet. 2011;20:3129–3137. doi: 10.1093/hmg/ddr215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Harris RS, Hultquist JF, Evans DT. The restriction factors of human immunodeficiency virus. The Journal of biological chemistry. 2012;287:40875–40883. doi: 10.1074/jbc.R112.416925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Neil SJ, Zang T, Bieniasz PD. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature. 2008;451:425–430. doi: 10.1038/nature06553. [DOI] [PubMed] [Google Scholar]
- 7.Van Damme N, Goff D, Katsura C, et al. The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein. Cell Host Microbe. 2008;3:245–252. doi: 10.1016/j.chom.2008.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hrecka K, Hao C, Gierszewska M, et al. Vpx relieves inhibition of HIV-1 infection of macrophages mediated by the SAMHD1 protein. Nature. 2011;474:658–661. doi: 10.1038/nature10195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J. The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature. 2004;427:848–853. doi: 10.1038/nature02343. [DOI] [PubMed] [Google Scholar]
- 10.Abdel-Mohsen M, Raposo RA, Deng X, et al. Expression profile of host restriction factors in HIV-1 elite controllers. Retrovirology. 2013;10:106. doi: 10.1186/1742-4690-10-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Raposo RA, Abdel-Mohsen M, Holditch SJ, et al. Increased expression of intrinsic antiviral genes in HLA-B*57-positive individuals. Journal of leukocyte biology. 2013;94:1051–1059. doi: 10.1189/jlb.0313150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Raposo RA, Abdel-Mohsen M, Bilska M, Montefiori DC, Nixon DF, Pillai SK. Effects of cellular activation on anti-HIV-1 restriction factor expression profile in primary cells. Journal of virology. 2013;87:11924–11929. doi: 10.1128/JVI.02128-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Anders S, McCarthy DJ, Chen Y, et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat Protoc. 2013;8:1765–1786. doi: 10.1038/nprot.2013.099. [DOI] [PubMed] [Google Scholar]
- 14.Vandesompele J, De Preter K, Pattyn F, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome biology. 2002;3:RESEARCH0034. doi: 10.1186/gb-2002-3-7-research0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Edmonds TG, Ding H, Yuan X, et al. Replication competent molecular clones of HIV-1 expressing Renilla luciferase facilitate the analysis of antibody inhibition in PBMC. Virology. 2010;408:1–13. doi: 10.1016/j.virol.2010.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.van der Fits L, van der Wel LI, Laman JD, Prens EP, Verschuren MC. Psoriatic lesional skin exhibits an aberrant expression pattern of interferon regulatory factor-2 (IRF-2) The Journal of pathology. 2003;199:107–114. doi: 10.1002/path.1263. [DOI] [PubMed] [Google Scholar]
- 17.Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. The Journal of experimental medicine. 2005;202:135–143. doi: 10.1084/jem.20050500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Abdel-Mohsen M, Deng X, Liegler T, et al. Effects of alpha interferon treatment on intrinsic anti-HIV-1 immunity in vivo. Journal of virology. 2014;88:763–767. doi: 10.1128/JVI.02687-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hunt PW, Brenchley J, Sinclair E, et al. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. The Journal of infectious diseases. 2008;197:126–133. doi: 10.1086/524143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Raposo RA, Abdel-Mohsen M, Deng X, et al. Dynamic regulation of host restriction factor expression over the course of HIV-1 infection in vivo. Journal of virology. 2014;88:11624–11629. doi: 10.1128/JVI.01771-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ferenczi K, Burack L, Pope M, Krueger JG, Austin LM. CD69, HLA-DR and the IL-2R identify persistently activated T cells in psoriasis vulgaris lesional skin: blood and skin comparisons by flow cytometry. Journal of autoimmunity. 2000;14:63–78. doi: 10.1006/jaut.1999.0343. [DOI] [PubMed] [Google Scholar]
- 22.International HIVCS. Pereyra F, Jia X, et al. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science. 2010;330:1551–1557. doi: 10.1126/science.1195271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kim AY, Kuntzen T, Timm J, et al. Spontaneous control of HCV is associated with expression of HLA-B 57 and preservation of targeted epitopes. Gastroenterology. 2011;140:686–696e681. doi: 10.1053/j.gastro.2010.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Christophers E, Henseler T. Contrasting disease patterns in psoriasis and atopic dermatitis. Archives of dermatological research. 1987;279 (Suppl):S48–51. doi: 10.1007/BF00585919. [DOI] [PubMed] [Google Scholar]
- 25.Morizane S, Gallo RL. Antimicrobial peptides in the pathogenesis of psoriasis. The Journal of dermatology. 2012;39:225–230. doi: 10.1111/j.1346-8138.2011.01483.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sheehy AM, Gaddis NC, Choi JD, Malim MH. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature. 2002;418:646–650. doi: 10.1038/nature00939. [DOI] [PubMed] [Google Scholar]
- 27.Bishop KN, Holmes RK, Sheehy AM, Davidson NO, Cho SJ, Malim MH. Cytidine deamination of retroviral DNA by diverse APOBEC proteins. Curr Biol. 2004;14:1392–1396. doi: 10.1016/j.cub.2004.06.057. [DOI] [PubMed] [Google Scholar]
- 28.Zheng YH, Irwin D, Kurosu T, Tokunaga K, Sata T, Peterlin BM. Human APOBEC3F is another host factor that blocks human immunodeficiency virus type 1 replication. Journal of virology. 2004;78:6073–6076. doi: 10.1128/JVI.78.11.6073-6076.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yu Q, Chen D, Konig R, Mariani R, Unutmaz D, Landau NR. APOBEC3B and APOBEC3C are potent inhibitors of simian immunodeficiency virus replication. The Journal of biological chemistry. 2004;279:53379–53386. doi: 10.1074/jbc.M408802200. [DOI] [PubMed] [Google Scholar]
- 30.Yu Q, Konig R, Pillai S, et al. Single-strand specificity of APOBEC3G accounts for minus-strand deamination of the HIV genome. Nat Struct Mol Biol. 2004;11:435–442. doi: 10.1038/nsmb758. [DOI] [PubMed] [Google Scholar]
- 31.Bourara K, Liegler TJ, Grant RM. Target cell APOBEC3C can induce limited G-to-A mutation in HIV-1. PLoS Pathog. 2007;3:1477–1485. doi: 10.1371/journal.ppat.0030153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dang Y, Wang X, Esselman WJ, Zheng YH. Identification of APOBEC3DE as another antiretroviral factor from the human APOBEC family. Journal of virology. 2006;80:10522–10533. doi: 10.1128/JVI.01123-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Dang Y, Siew LM, Wang X, Han Y, Lampen R, Zheng YH. Human cytidine deaminase APOBEC3H restricts HIV-1 replication. The Journal of biological chemistry. 2008;283:11606–11614. doi: 10.1074/jbc.M707586200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Berger G, Durand S, Fargier G, et al. APOBEC3A is a specific inhibitor of the early phases of HIV-1 infection in myeloid cells. PLoS Pathog. 2011;7:e1002221. doi: 10.1371/journal.ppat.1002221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhu J, Gaiha GD, John SP, et al. Reactivation of Latent HIV-1 by Inhibition of BRD4. Cell Rep. 2012;2:807–816. doi: 10.1016/j.celrep.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Schoggins JW, Wilson SJ, Panis M, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485. doi: 10.1038/nature09907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen H, Li C, Huang J, et al. CD4+ T cells from elite controllers resist HIV-1 infection by selective upregulation of p21. J Clin Invest. 2011;121:1549–1560. doi: 10.1172/JCI44539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fellay J, Shianna KV, Ge D, et al. A whole-genome association study of major determinants for host control of HIV-1. Science. 2007;317:944–947. doi: 10.1126/science.1143767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Liu SY, Aliyari R, Chikere K, et al. Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol. Immunity. 2013;38:92–105. doi: 10.1016/j.immuni.2012.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wilson SJ, Schoggins JW, Zang T, et al. Inhibition of HIV-1 particle assembly by 2′,3′-cyclic-nucleotide 3′-phosphodiesterase. Cell Host Microbe. 2012;12:585–597. doi: 10.1016/j.chom.2012.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Liu L, Oliveira NM, Cheney KM, et al. A whole genome screen for HIV restriction factors. Retrovirology. 2011;8:94. doi: 10.1186/1742-4690-8-94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Muto NF, Martinand-Mari C, Adelson ME, Suhadolnik RJ. Inhibition of replication of reactivated human immunodeficiency virus type 1 (HIV-1) in latently infected U1 cells transduced with an HIV-1 long terminal repeat-driven PKR cDNA construct. Journal of virology. 1999;73:9021–9028. doi: 10.1128/jvi.73.11.9021-9028.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Woods MW, Kelly JN, Hattlmann CJ, et al. Human HERC5 restricts an early stage of HIV-1 assembly by a mechanism correlating with the ISGylation of Gag. Retrovirology. 2011;8:95. doi: 10.1186/1742-4690-8-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Pincetic A, Kuang Z, Seo EJ, Leis J. The interferon-induced gene ISG15 blocks retrovirus release from cells late in the budding process. Journal of virology. 2010;84:4725–4736. doi: 10.1128/JVI.02478-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lodermeyer V, Suhr K, Schrott N, et al. 90K, an interferon-stimulated gene product, reduces the infectivity of HIV-1. Retrovirology. 2013;10:111. doi: 10.1186/1742-4690-10-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Furtak V, Mulky A, Rawlings SA, et al. Perturbation of the P-body component Mov10 inhibits HIV-1 infectivity. PLoS One. 2010;5:e9081. doi: 10.1371/journal.pone.0009081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kane M, Yadav SS, Bitzegeio J, et al. MX2 is an interferon-induced inhibitor of HIV-1 infection. Nature. 2013;502:563–566. doi: 10.1038/nature12653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Goujon C, Moncorge O, Bauby H, et al. Human MX2 is an interferon-induced post-entry inhibitor of HIV-1 infection. Nature. 2013;502:559–562. doi: 10.1038/nature12542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Liu Z, Pan Q, Ding S, et al. The Interferon-Inducible MxB Protein Inhibits HIV-1 Infection. Cell Host Microbe. 2013;14:398–410. doi: 10.1016/j.chom.2013.08.015. [DOI] [PubMed] [Google Scholar]
- 50.Malathi K, Dong B, Gale M, Jr, Silverman RH. Small self-RNA generated by RNase L amplifies antiviral innate immunity. Nature. 2007;448:816–819. doi: 10.1038/nature06042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nasr N, Maddocks S, Turville SG, et al. HIV-1 infection of human macrophages directly induces viperin which inhibits viral production. Blood. 2012;120:778–788. doi: 10.1182/blood-2012-01-407395. [DOI] [PubMed] [Google Scholar]
- 52.Li M, Kao E, Gao X, et al. Codon-usage-based inhibition of HIV protein synthesis by human schlafen 11. Nature. 2012;491:125–128. doi: 10.1038/nature11433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Carthagena L, Bergamaschi A, Luna JM, et al. Human TRIM gene expression in response to interferons. PLoS One. 2009;4:e4894. doi: 10.1371/journal.pone.0004894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Uchil PD, Quinlan BD, Chan WT, Luna JM, Mothes W. TRIM E3 ligases interfere with early and late stages of the retroviral life cycle. PLoS Pathog. 2008;4:e16. doi: 10.1371/journal.ppat.0040016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Barr SD, Smiley JR, Bushman FD. The interferon response inhibits HIV particle production by induction of TRIM22. PLoS Pathog. 2008;4:e1000007. doi: 10.1371/journal.ppat.1000007. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.