key: cord-0702354-hi3i1mwl authors: Taylor, Jared P.; Cash, Melanie N.; Santostefano, Katherine E.; Nakanishi, Mahito; Terada, Naohiro; Wallet, Mark A. title: CRISPR/Cas9 knockout of USP18 enhances type I IFN responsiveness and restricts HIV-1 infection in macrophages date: 2018-02-13 journal: Journal of Leukocyte Biology DOI: 10.1002/jlb.3mia0917-352r sha: 4fb5e7fb5d3ec9366bfa98bb436d3b34dc4755fb doc_id: 702354 cord_uid: hi3i1mwl The IFN-stimulated gene ubiquitin specific proteinase 18 (USP18) encodes a protein that negatively regulates T1 IFN signaling via stearic inhibition of JAK1 recruitment to the IFNα receptor 2 subunit (IFNAR2). Here we demonstrate that USP18 expression is induced by HIV-1 in a T1 IFN dependent manner. Experimental depletion of USP18 by CRISPR/Cas9 gene editing results in a significant restriction of HIV-1 replication in an induced pluripotent stem cell (iPSC)-derived macrophage model. In the absence of USP18, macrophages have increased responsiveness to stimulation with T1 IFNs with prolonged phosphorylation of STAT1 and STAT2 and increased expression of interferon-stimulated genes that are key for antiviral responses. Interestingly, HIV-1 requires some signaling through the T1 IFN receptor to replicate efficiently because a neutralizing antibody that inhibits T1 IFN activity reduces HIV-1 replication rate in monocyte-derived macrophages. USP18 induction by HIV-1 tunes the IFN response to optimal levels allowing for efficient transcription from the HIV-1 LTR promoter while minimizing the T1 IFN-induced antiviral response that would otherwise restrict viral replication and spread. Finally, iPSC and CRISPR/Cas9 gene targeting offer a powerful tool to study host factors that regulate innate immune responses. It is well established that type I IFNs (T1 IFNs) can restrict acute HIV-1 infection in vitro. [1] [2] [3] [4] [5] In clinical trials treating human patients with recombinant IFN-2a, T1 IFNs can suppress viral replication in the absence of antiretroviral therapy (ART) in some patients. 6, 7 However, this approach failed in long-term treatment when study subjects became refractory to IFN treatment and viral loads returned to previous levels. T1 IFNs are important in establishing early control of HIV-1 in an in vivo SIV rhesus macaque model, 8 Signaling through IFNAR results in the phosphorylation and activation of STAT proteins including STAT1 and STAT2. The consequence of STAT1/2 phosphorylation is induced expression of hundreds of ISGs. [15] [16] [17] The protein products of ISGs then target host and viral machinery as a means to restrict viral replication. However, a small subset of the ISGs expressed are negative feedback mechanisms that turn off IFN signaling so that resolution of the immune response can occur. One of these important negative regulators is ubiquitin-specific proteinase 18 (USP18). USP18 is an IFN-inducible deISGylating enzyme that specifically removes the ubiquitin-like posttranslational modification, ISG15, from target proteins. [18] [19] [20] During an IFN response, many newly synthesized proteins are ISGylated, 21 which can have a variety of effects depending on the target. Some targets, such as IFN regulatory factor 3 (IRF3), are protected from ubiquitin-mediated proteasomal degradation. 22 It has also been shown that viral proteins, including HIV-1 Gag, can be ISGylated. 23 Durfee and colleagues 21 propose that during an infection a small subset of viral structural proteins are ISGylated to disrupt the repeating structures found in viral capsids. Influenza B has evolved a mechanism to directly neutralize ISG15 with its NS1 protein 24 and coronaviruses have a papain-like protease that has deISGylase activity as a strategy to overcome ISG15, 25, 26 indicating the importance of ISG15 in the antiviral response. In addition to its enzymatic activity, USP18 negatively regulates T1 IFN signaling. 27 USP18 is recruited by STAT2 to the type I IFN receptor subunit, IFNAR2, where it binds to IFNAR2 and prevents phosphorylation of JAK1 by blocking the interaction of JAK1 and the IFNAR2 subunit. [27] [28] [29] USP18 expression also plays a role in limiting TRAILinduced apoptosis and has also been shown to regulate the susceptibility of certain cancer cells to IFN-and drug-induced apoptosis. 30, 31 Macrophages play an important role in HIV-1 as reservoirs and can contribute directly to HIV-1 pathogenesis. 32 HIV-1 in the ART era can be seen as a chronic disease characterized by chronic immune activation and chronic inflammation with a higher risk of non-AIDS-related morbidities and mortalities. Macrophages play an important role in this process and can act as mediators of inflammation. 33 , 34 We recently reported that HIV-1 replication in macrophages requires activity of STAT1, a protein usually associated with antiviral responses. 35 The role of STAT1 was in postintegration expression of HIV-1 mRNA from the long terminal repeat (LTR) promoter. This paradoxical mechanism where HIV-1 usurps antiviral pathways as a means of driving its own replication suggests that a complex host-pathogen interplay ultimately determines if HIV-1 can efficiently replicate in macrophages. In general, the role of macrophages in the HIV-1 life cycle is important because eliminating persistently infected macrophages in addition to latently infected T cells will be necessary for a sterilizing cure to be achieved. Thus, it is critical that we understand the host-pathogen dynamics that regu- Lenti-X 293T cells were cultured in a T75 flask and transfected with Medium was removed by aspiration and 100 L of room temperature Monocyte-derived macrophages (MDMs) were infected with 500 TCID 50 Human GIPZ lentiviral shRNA gene set for USP18-specific shRNA Differentiation was carried out as previously described 48 Fc receptors were blocked with Fc Block (Miltenyi) and stained with antibodies in PBS with 1% human serum. For intracellular staining of CD68, the cells were fixed and permeabilized with the BD Cytofix/Cytoperm TM kit (BD Biosciences). All samples were analyzed with the BD Accuri TM C6 Cytometer (BD Biosciences). The integration assay for detection of integrated HIV-1 proviral DNA was adapted from methods previously described. 49 It has been demonstrated previously in vitro that T1 IFNs restrict HIV-1 replication. [1] [2] [3] [4] [5] 51 To confirm these previous findings, we pretreated TZM-bl cells with different doses of IFN-for 24 h and infected the cells with HIV-1. As previously reported, T1 IFN restricted HIV-1 replication in a dose-dependent manner (Fig. 1A) . We also tested this in MDMs and found that HIV-1 replication is potently restricted by IFN-in a dose-dependent manner (Fig. 1B) with an IC 50 of 504.7 fg/mL (± 142.1 fg/mL). Since HIV-1 is restricted by T1 IFNs in vitro, we wanted to determine if HIV-1 was inducing an IFN response in MDMs. To test this, MDMs were infected with HIV-1 for 7 days and gene expression was analyzed by microarray ( Fig. 2A) . We observed that all genes up-regulated at least 2-fold by HIV-1 (Table 1) have been reported to be IFN-inducible in the Interferome (v2.01) database (Fig. 2B) . However, this IFN-like response is not strong enough to inhibit HIV-1 replication to the same degree as the addition of exogenous IFN (Fig. 1) . We were interested in how the IFN response was being attenuated and allowing for IFN signaling (Fig. 3) , providing evidence that HIV-1 does induce an IFN response in macrophages that has an autocrine effect. Surprisingly, HIV-1 replication was inhibited, not enhanced, in the absence of T1 IFN signaling as measured by Gag p24 ELISA on supernatants (Fig. 3) . This finding agrees with previous work where we demonstrated the importance of STAT signaling for HIV-1 replication in MDMs. 35 These data demonstrate that HIV-1 must strike a balance between some T1 IFN signaling, which is needed for producing transcription factors such as STAT1 and STAT3, and too much IFN signaling, which allows the cells to mount an effective antiviral response. Given that USP18 attenuates the IFN response, we hypothesized that induction of USP18 allowed HIV-1 to achieve the balance needed to provide the necessary STAT signaling, without too strong an antiviral response. USP18 is part of the normal negative feedback response that regulates T1 IFN signaling through IFNAR and JAK-STAT signaling. In the absence of USP18, signaling through the JAK-STAT pathway should be enhanced with increased levels of phosphorylated STAT1 and STAT2. We used siRNA to knockdown USP18 expression in MDMs and treated them with IFN-for 18 hours and found increased levels of phosphorylated STAT1 and STAT2 (Fig. 4A ). (Fig. 4B) . USP18 expression was also measured to confirm knockdown of USP18 transcripts. We next wanted to determine if USP18 expression is necessary for HIV-1 replication. Although transfecting MDMs with siRNA allowed for efficient knockdown of USP18 (Fig. 4) , it also made the cells refractory to infection even with a nontargeting control siRNA (data not shown). Instead, we utilized shRNA knockdown of USP18 in the THP-1 cell line. THP-1 cells are suspension cells that can be differentiated into adherent macrophage-like cells with PMA treatment. They are a well-established model system for studying HIV-1 infection in macrophages. 52 We generated THP-1 cell lines that constitutively express nontargeting control or USP18-specific shRNA (Supplemental Figs. S1A and S1B). After PMA differentiation, THP-1 cells expressing non-targeting control or USP18 shRNA were infected by HIV-1. We found that HIV-1 replication was significantly restricted in THP-1 cells with USP18 knockdown. There was significantly less Gag p24 detected in the supernatants and in the protein lysates of USP18 knockdown cells compared with control (Figs. 5A and 5B). While THP-1 cells and MDMs with USP18 knockdown provide useful models for investigating the effects of USP18 deficiency on IFN signaling, it may not be the best model. THP-1 cells are transformed cells with an abnormal karyotype 53 and in our hands they do not remain differentiated and adherent in culture for more than 5 days (data not shown). To test the effects of USP18 deficiency in iMacs, we utilized CRISPR/Cas9 gene editing to knockout USP18 in iPSCs before differentiation to the myeloid lineage. The advantage of knocking out USP18 in iPSCs is that once knockout is achieved, we have a self- were measured by Western blot. USP18 −/− iMacs had increased and sustained phosphorylated STAT1 and phosphorylated STAT2 (Fig. 7A ). Since USP18 −/− iMacs also had increased STAT1 and STAT2 signaling, we wanted to determine if USP18 −/− iMacs also had enhanced ISG expression after IFN-treatment. Indeed, after treatment with IFNfor 18 h, USP18 −/− iMacs had increased expression of ISGs compared with USP18 sufficient cells (Fig. 7B) . These findings are consistent with the results from siRNA knockdown in MDMs. We next determined if iMacs can support HIV-1 replication as previously reported 48 and if USP18 is induced by HIV-1 in iMacs. Indeed, USP18 +/+ iMacs support HIV-1 replication and USP18 is induced by HIV-1 in these cells (Figs. 8A and 8B). Since USP18 is induced by HIV-1 in iMacs in a similar manner as MDMs, we concluded that iMacs are a better model for studying USP18 knockout/knockdown than THP-1 cells. Next, we wanted to determine if USP18 knockout influenced HIV-1 replication. USP18 knockout results in an enhanced response to IFN stimulation in macrophages (Fig. 7) . Therefore, we hypothe- We report here that USP18 deficiency restricts HIV-1 replication in human macrophages using an iPSC model. The restriction on replication was due to an enhanced IFN response from a lack of feedback inhibition on the JAK/STAT pathway resulting in increased expression of ISGs that have antiviral effects. Previous work done by our group has shown that STAT1 and STAT3 are necessary for efficient HIV-1 replication. Blocking phosphorylation of either of these 2 molecules resulted in significant reduction in viral replication. 35 However, the current study shows that increasing the amount of phosphorylated STAT1 and STAT2 also results in a significant reduction in viral replication. Thus, we propose that USP18 acts as a tuning mechanism which prevents a robust IFN response that would restrict viral replication (Fig. 9) . Previous work has shown that Usp18 −/− mice exhibit protection from intracerebral infection by LCMV and VSV. 55 Usp18 −/− mice also The basis of antibacterial protection in USP18 deficiency can be attributed to hypersensitivity to LPS as a consequence of an overactive T1 IFN response. 56 Mechanistically, these studies showed that USP18 deficiency resulted in increased ISGylation of proteins and increased phosphorylated STAT1 levels in response to LPS treatment or viral infection. It was based on these properties that we sought to determine if human USP18 regulates HIV-1 replication. supports HIV-1 replication is a window of low level immune activation that is due, at least in part, to balanced positive and negative regulators of antiviral immunity (Fig. 9 ). Infected cells eventually become refractory to T1 IFN signaling due to induction of negative regulators such as USP18. Other negative regulators are also induced by T1 IFNs including SOCS1, SOCS3, and PIAS. 75 We focused on USP18 for this study due to its additional function as a deISGylating enzyme and its appearance in gene expression profiling experiments following HIV-1 infection. The role of interferons in the control of HIV replication in macrophages Restriction of HIV replication in infected T cells and monocytes by interferon-alpha Mechanisms for the inhibition of HIV replication by interferons-alpha, -beta, and -gamma in primary human macrophages Interferon alpha-mediated inhibition of human immunodeficiency virus type 1 provirus synthesis in T-cells Recombinant human interferon alfa-A suppresses HTLV-III replication in vitro AIDS Clinical Trials Group A5192 Team. Safety, tolerability, and mechanisms of antiretroviral activity of pegylated interferon Alfa-2a in HIV-1-monoinfected participants: a phase II clinical trial Pegylated Interferon alfa-2a monotherapy results in suppression of HIV type 1 replication and decreased cell-associated HIV DNA integration Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression The interferon paradox: can inhibiting an antiviral mechanism advance an HIV cure Interferon-is the primary plasma type-I IFN in HIV-1 infection and correlates with immune activation and disease markers Targeting type I interferon-mediated activation restores immune function in chronic HIV infection Type I interferons suppress viral replication but contribute to T cell depletion and dysfunction during chronic HIV-1 infection Blockade of chronic type I interferon signaling to control persistent LCMV infection Persistent LCMV infection is controlled by blockade of type I interferon signaling Interferon-induced nuclear factors that bind a shared promoter element correlate with positive and negative transcriptional control Interferoninduced transcription of a gene encoding a 15-kDa protein depends on an upstream enhancer element Interferonstimulated transcription: isolation of an inducible gene and identification of its regulatory region UBP43 (USP18) specifically removes ISG15 from conjugated proteins Structural basis of the specificity of USP18 toward ISG15 Molecular characterization of ubiquitin-specific protease 18 reveals substrate specificity for interferon-stimulated gene 15 The ISG15 conjugation system broadly targets newly synthesized proteins: implications for the antiviral function of ISG15 ISG15 enhances the innate antiviral response by inhibition of IRF-3 degradation Human HERC5 restricts an early stage of HIV-1 assembly by a mechanism correlating with the ISGylation of Gag Influenza B virus NS1 protein inhibits conjugation of the interferon (IFN)-induced ubiquitin-like ISG15 protein MERS-CoV papain-like protease has deISGylating and deubiquitinating activities Structural Basis for the Ubiquitin-Linkage Specificity and deISGylating activity of SARS-CoV papain-like protease UBP43 is a novel regulator of interferon signaling independent of its ISG15 isopeptidase activity Multiple functions of USP18 STAT2 is an essential adaptor in USP18-mediated suppression of type I interferon signaling The DeIS-Gylase USP18 limits TRAIL-induced apoptosis through the regulation of TRAIL levels: cellular levels of TRAIL influences responsiveness to TRAIL-induced apoptosis Identification of USP18 as an important regulator of the susceptibility to IFN-alpha and drug-induced apoptosis Macrophages and HIV-1: an Unhealthy Constellation Immune activation and inflammation in HIV-1 infection: causes and consequences Systemic effects of inflammation on health during chronic HIV infection HIV-1 Infection Primes Macrophages through STAT Signaling to Promote Enhanced Inflammation and Viral Replication Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1 Identification of gammaretroviruses constitutively released from cell lines used for human immunodeficiency virus research Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is modulated by coreceptor specificity defined by the V3 loop of gp120 Evidence that ecotropic murine leukemia virus contamination in TZM-bl cells does not affect the outcome of neutralizing antibody assays with human immunodeficiency virus type 1 Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone Monokine regulation of human immunodeficiency virus-1 expression in a chronically infected human T cell clone Efficient magnetic bead-based separation of HIV-1-infected cells using an improved reporter virus system reveals that p53 up-regulation occurs exclusively in the virus-expressing cell population Role of the basic domain of human immunodeficiency virus type 1 matrix in macrophage infection The method of "right and wrong cases" ("constant stimuli") without Gauss's formulae Beitrag zur kollektiven Behandlung pharmakologischer Reihenversuche Development of defective and persistent Sendai virus vector: a unique gene delivery/expression system ideal for cell reprogramming Efficient, long term production of monocyte-derived macrophages from human pluripotent stem cells under partly-defined and fully-defined conditions Cross-clade ultrasensitive PCR-based assays to measure HIV persistence in large-cohort studies Analysis of early human immunodeficiency virus type 1 DNA synthesis by use of a new sensitive assay for quantifying integrated provirus analysis of early human immunodeficiency virus type 1 DNA synthesis by use of a new sensitive assay for quantifying integrated provirus Regulation of HIV replication in infected monocytes by IFN-alpha. Mechanisms for viral restriction Monocyte-derived macrophages and myeloid cell lines as targets of HIV-1 replication and persistence Cytogenetic and molecular analysis of the acute monocytic leukemia cell line THP-1 with an MLL-AF9 translocation Development of Sendai virus vectors and their potential applications in gene therapy and regenerative medicine Role of ISG15 protease UBP43 (USP18) in innate immunity to viral infection Enhanced antibacterial potential in UBP43-deficient mice against Salmonella typhimurium infection by up-regulating type I IFN signaling ISG15 deficiency and increased viral resistance in humans but not mice Human intracellular ISG15 prevents interferon-/ over-amplification and autoinflammation Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome Microarray analysis reveals that Type I interferon strongly increases the expression of immune-response related genes in Ubp43 (Usp18) deficient macrophages Enforced viral replication activates adaptive immunity and is essential for the control of a cytopathic virus The antiviral activities of ISG15 Role of interferonstimulated gene ISG-15 in the interferon-omega-mediated inhibition of human immunodeficiency virus replication Innate antiviral response targets HIV-1 release by the induction of ubiquitin-like protein ISG15 The interferon-induced gene ISG15 blocks retrovirus release from cells late in the budding process A diverse range of gene products are effectors of the type I interferon antiviral response The broad-spectrum antiviral functions of IFIT and IFITM proteins IFITM proteins restrict HIV-1 infection by antagonizing the envelope glycoprotein IFITM proteins incorporated into HIV-1 virions impair viral fusion and spread IFITM proteins are incorporated onto HIV-1 virion particles and negatively imprint their infectivity MX2 is an interferoninduced inhibitor of HIV-1 infection Human MX2 is an interferon-induced post-entry inhibitor of HIV-1 infection Transcriptional and posttranscriptional regulation of HIV-1 gene expression. Cold Spring Harb Perspect Med HIV-1 latency: an update of molecular mechanisms and therapeutic strategies Interferon-Stimulated Genes: a Complex Web of Host Defenses Vascular smooth muscle cells from hypertensive patient-derived induced pluripotent stem cells to advance hypertension pharmacogenomics Genome therapy of myotonic dystrophy type 1 iPS cells for development of autologous stem cell therapy Disulfide bond disrupting agents activate the unfolded protein response in EGFRand HER2-positive breast tumor cells Directed differentiation of embryonic stem cells into cardiomyocytes by bacterial injection of defined transcription factors CRISPR/Cas9 knockout of USP18 enhances type I IFN responsiveness and restricts HIV-1 infection in macrophages Armitage for his assistance with CRISPR plasmid transfections and Sneha Patel for her assistance with Western blots. The authors declare no conflicts of interest. http://orcid.org/0000-0001-7700-530X