|
Post by Admin on Oct 24, 2020 22:12:35 GMT
Fig. 3 NRP mediates entry of nanoparticles coated with SARS-2 S derived CendR peptides into cultured cells, olfactory epithelium and CNS of mice. (A) Sequence of the peptides used for AgNP coating. Peptides that mimic SARS-2 S protein after furin cleavage (post) and as controls, S protein before cleavage (pre), with a point mutation of the terminal arginine (Ala) or with an amide terminus (post amide). (B and C) Representative images and quantification of the internalization of peptide-coated AgNPs in HEK-293T cells expressing NRP1. Merged images show AgNP-positive cells (magenta) and Hoechst (cyan). One-way ANOVA with Tukey’s correction for multiple comparisons. (D to G) Representative images and quantification of main olfactory epithelium (MOE) (D and E, respectively) and cortex (F and G, respectively) 6 hours after intranasal administration of AgNPs coated with SARS2-post and SARS2-post amide peptides. n = 4 replicates for C; n = 5 (E) and n = 4 (G) mice per condition. Data are means ± s.d. Two-tailed unpaired Student’s t test *p < 0.05, ***p < 0.001. AgNPs (magenta), Hoechst (cyan), NeuN (neuronal marker, green), AQP4 (yellow). Scale bars, 100 μm (B), 20 μm (D, F). Having obtained evidence for a role of NRP1 in cell entry of SARS-CoV-2, we examined whether NRP1 expression correlated with the detection of virus RNA in single cell transcriptomes. For these analyses, we used published scRNA-seq datasets of cultured experimentally infected human bronchial epithelial cell (HBECs) and cells isolated from bronchoalveolar lavage fluid (BALF) of severely affected COVID-19 patients (17). Among the proposed entry and amplification factors, NRP1, FURIN and TMPRSS11A, were enriched in SARS-CoV-2 infected cells compared to non-infected cells (fig. S6). We also detected increased expression of these proteins following infection (fig. S6). In addition, RNA expression of NRP1 and its homolog NRP2 was elevated in SARS-CoV-2-positive cells compared to adjacent cells in the BALF of severely affected COVID-19 patients (fig. S7). Because the availability of virus receptors and entry cofactors on the surface of host cells determines infectivity, we compared the expression patterns of ACE2 and NRP1 in published scRNA-seq datasets of human lung tissue (18) and human olfactory epithelium (19). While ACE2 was detected at very low levels, both NRP1 and NRP2 were abundantly expressed in almost all pulmonary and olfactory cells with the highest expression in endothelial cells (figs. S8 and S9). We confirmed these results by examining NRP1 immunoreactivity in human autopsy tissue and detected NRP1 in the epithelial surface layer of the human respiratory and olfactory epithelium (fig. S10A). ACE2 was hardly detectable in these tissues (fig. S10B). Within the olfactory epithelium, NRP1 was also observed in late olfactory neuronal progenitors and/or newly differentiated olfactory neurons (fig. S10C). In light of the widely reported disturbance of olfaction in a large fraction of COVID-19 patients (20) and the enrichment of NRPs in the olfactory epithelium, we analyzed a series of autopsies from six COVID-19 patients and eight non-infected control patients to determine whether SARS-CoV-2 could infect NRP1-positive cells (Fig. 4 and table S1). Using antibodies against the S protein, we detected infection of the olfactory epithelium in five out of six COVID-19 patients. The infected olfactory epithelial cells showed high expression of NRP1 (Fig. 4, A and B). Additional co-staining indicated infection of NRP1-positive late olfactory neuronal progenitors and/or newly differentiated olfactory neurons (Fig. 4B and fig. S11). Fig. 4 SARS-CoV-2 infects the olfactory epithelium. (A) Co-staining of S protein (brown) and NRP1 (lilac) in the apical olfactory epithelium (OE) in a COVID-19 patient and non-infected control (LP, lamina propria, HB, horizontal basal cells). (B) Co-localization of NRP1 (magenta) and S protein (yellow) in OE cells in a COVID-19 patient. Co-staining of OLIG-2 (magenta) and S protein (yellow) reveals infection of late olfactory neuronal progenitors/newly differentiated olfactory neurons. Scale bars, 10 μm. There is limited knowledge about the virus-host interactions that determine cellular entry of SARS-CoV-2. Viruses display considerable redundancy and flexibility because they can exploit weak multivalent interactions to enhance affinity. While the focus to date has been almost entirely on the role of ACE2 in SARS-CoV-2 entry, the expression pattern of ACE2 does not match tissue tropism of SARS-CoV-2 (21). This raises the possibility that co-factors are required to facilitate virus-host cell interactions in cells with low ACE2 expression. NRP1 could represent such an ACE2 potentiating factor; however, it is also possible that SARS-CoV-2 can enter cells independently of ACE2 when viral loads are high. The reason why a number of viruses (22–25) use NRPs as entry factors could be because of their high expression on epithelia facing the external environment, and their function in enabling cell, vascular, and tissue penetration (9, 13). Science 20 Oct 2020: eabd2985 DOI: 10.1126/science.abd2985
|
|
|
Post by Admin on Nov 22, 2020 21:23:28 GMT
In order to better understand the complex relationships between host and virus genetic dependencies, the team used a broad range of analytical and experimental methods to validate their results. This integrative approach included genome editing, single-cell sequencing, confocal imaging and computational analyses of gene expression and proteomic datasets. After intensive research, the scientists and doctors claim they have found 30 genes that block the virus from infecting human cells including RAB7A, a gene that seems to regulate the ACE-2 receptor that the virus binds to and uses to enter the cell. The spike protein’s first contact with a human cell is through ACE-2 receptor. “Our findings confirmed what scientists believe to be true about ACE-2 receptor’s role in infection; it holds the key to unlocking the virus,” said Dr. tenOever. “It also revealed the virus needs a toolbox of components to infect human cells. Everything must be in alignment for the virus to enter human cells.” The team discovered that the top-ranked genes — those whose loss reduces viral infection substantially — clustered into a handful of protein complexes, including vacuolar ATPases, Retromer, Commander, Arp2/3, and PI3K. Many of these protein complexes are involved in trafficking proteins to and from the cell membrane. “We were very pleased to see multiple genes within the same family as top-ranked hits in our genome-wide screen. This gave us a high degree of confidence that these protein families were crucial to the virus lifecycle, either for getting into human cells or successful viral replication,” said Dr. Zharko Daniloski, a postdoctoral fellow in the Sanjana Lab and co-first author of the study. Using proteomic data, they found that several of the top-ranked host genes directly interact with the virus’s own proteins, highlighting their central role in the viral lifecycle. The team also analyzed common host genes required for other viral pathogens, such as Zika or H1N1 pandemic influenza. Cholesterol and the virus The research team also identified drugs that are currently on the market for different diseases that they claim block the entry of Covid-19 into human cells by increasing cellular cholesterol. In particular, they found three drugs currently on the market were more than 100-fold more effective in stopping viral entry in human lung cells: Amlodipine, brand name Norvasc, by Pfizer, to treat high blood pressure and angina. Tamoxifen, brand name Soltamox by Fortovia Therapeutics, an estrogen modulator, to treat breast cancer. Ilomastat, brand name Galardin, it’s a matrix metalloprotease inhibitor, that now being manufactured by many companies; a chemotherapy agent, with applications for skincare and anti-aging products. The other five drugs that were tested — called PIK-111, Compound 19, SAR 405, Autophinib, ALLN -- are used in research but are not yet branded and used in clinical trials for existing diseases. Their findings offer insight into novel therapies that may be effective in treating Covid-19 and reveal the underlying molecular targets of those therapies. The bioengineers in New York were working on other projects with gene-editing technology from CRISPR but quickly pivoted to studying the coronavirus when it swept through the metropolitan area last March. “Seeing the tragic impact of Covid-19 here in New York and across the world, we felt that we could use the high-throughput CRISPR gene editing tools that we have applied to other diseases to understand what are the key human genes required by the SARS-CoV-2 virus,” said Dr. Sanjana. Dr. Neville Sanjana and his team at the New York Genome Center used CRISPR to identify the genes that can protect human cells against Covid-19. As he explained, “current treatments for SARS-CoV-2 infection currently go after the virus itself, but this study offers a better understanding of how host genes influence viral entry and will enable new avenues for therapeutic discovery.” Previously, Dr. Sanjana has applied genome-wide CRISPR screens to identify the genetic drivers of diverse diseases, including drug resistance in melanoma, immunotherapy failure, lung cancer metastasis, innate immunity, inborn metabolic disorders and muscular dystrophy. “The hope is that the data from this study— which pinpoints required genes for SARS-CoV-2 infection — could in the future work be combined with human genome sequencing data to identify individuals that might be either more susceptible or more resistant to Covid-19,” Dr. Sanjana said. The New York team is not the first to use CRISPR gene editing techniques to fight Covid-19. Other bioengineering groups at MIT and Stanford have been using CRISPR to develop ways to fight the SARS-CoV-2 and develop diagnostic tools for Covid-19.
|
|
|
Post by Admin on Nov 23, 2020 5:02:33 GMT
Identification of Required Host Factors for SARS-CoV-2 Infection in Human Cells Summary To better understand host-virus genetic dependencies and find potential therapeutic targets for COVID-19, we performed a genome-scale CRISPR loss-of-function screen to identify host factors required for SARS-CoV-2 viral infection of human alveolar epithelial cells. Top-ranked genes cluster into distinct pathways, including the vacuolar ATPase proton pump, Retromer, and Commander complexes. We validate these gene targets using several orthogonal methods such as CRISPR knockout, RNA interference knockdown, and small-molecule inhibitors. Using single-cell RNA-sequencing, we identify shared transcriptional changes in cholesterol biosynthesis upon loss of top-ranked genes. In addition, given the key role of the ACE2 receptor in the early stages of viral entry, we show that loss of RAB7A reduces viral entry by sequestering the ACE2 receptor inside cells. Overall, this work provides a genome-scale, quantitative resource of the impact of the loss of each host gene on fitness/response to viral infection. Published:October 24, 2020 DOI:https://doi.org/10.1016/j.cell.2020.10.030 Introduction As of October 2020, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, has infected 40 million people worldwide and led to the deaths of more than 1 million people, according to the John Hopkins Research Center (Gardner, 2020). SARS-CoV-2 belongs to the family of enveloped viruses known as Coronaviridae and was first reported in late 2019 in China. Over the past two decades, it is the third zoonotic coronavirus to emerge: compared to the other two coronaviruses, SARS-CoV (2002) and Middle East respiratory syndrome (MERS)-CoV (2012), SARS-CoV-2 shows an increased infectivity and lower case-fatality rate, contributing to its wide-spread transmission and resulting in a pandemic (Gates, 2020; Liu et al., 2020). Given that SARS-CoV-2 has already taken a major toll on human life and livelihoods worldwide, many research institutions, governmental organizations, and pharmaceutical companies are working to identify antiviral drugs and develop vaccines. Currently, there are nearly 30 vaccines against SARS-CoV-2 in clinical trials and a Food and Drug Administration (FDA)-approved antiviral drug (remdesivir) that acts as an inhibitor of the SARS-CoV-2 viral RNA-dependent RNA polymerase (Beigel et al., 2020; Funk et al., 2020). A recent study identified small molecules that antagonize SARS-CoV-2 replication and infection by testing ∼12,000 clinical-stage and FDA-approved inhibitors (Riva et al., 2020). Here, we utilize an alternative approach—a genome-scale loss-of-function screen—to identify targets among host genes that are required for SARS-CoV-2 infection. These gene targets (and inhibitors of these genes) may aid in the development of new therapies for COVID-19. SARS-CoV-2 is an enveloped positive-sense RNA virus that relies on host factors for all stages of its life cycle (Kim et al., 2020; Zhou et al., 2020). The viral envelope is coated by Spike protein trimers that bind to angiotensin converting-enzyme 2 (ACE2) receptor, which is required for SARS-CoV-2 infection (Hoffmann et al., 2020a; Zhou et al., 2020). The Spike protein undergoes proteolytic cleavage that is catalyzed by several host proteases, such as furin, TMPRSS2, and cathepsin L, and can occur in the secretory pathway of the host cell or during viral entry in the target cell. Proteolytic cleavage is considered to be required for activation of Spike that in turn allows for viral-host membrane fusion and release of the viral RNA into the host cytoplasm (Hoffmann et al., 2020b). Once in the cytoplasm, the virus utilizes the host and its own machinery to replicate its genetic material and assemble new viral particles. Recent proteomic studies have identified hundreds of host proteins that directly bind to SARS-CoV-2 viral proteins and have mapped changes in the global protein phosphorylation landscape in response to viral infection, highlighting the interest in better understanding of host-virus genetic dependencies (Bouhaddou et al., 2020; Gordon et al., 2020). To date, there are no genome-wide studies that directly identify human genes required for viral infection, which will be of great interest and utility for the broader scientific community. Here, we perform a genome-scale CRISPR loss-of-function screen in human alveolar basal epithelial carcinoma cells to identify genes whose loss confers resistance to SARS-CoV-2 viral infection. We validate that these genes reduce SARS-CoV-2 infection using multiple orthogonal cell perturbations (CRISPR knockout, RNA interference knockdown, and small-molecule inhibitors). For the top gene hits, we explore potential mechanisms of their antiviral activity using single-cell transcriptomics, flow cytometry, and immunofluorescence. Using single-cell transcriptomics, we identified a group of genes (ATP6AP1, ATP6V1A, NPC1, RAB7A, CCDC22, and PIK3C3) whose knockout induced shared transcriptional changes in cholesterol biosynthesis pathway. Perturbation of the cholesterol biosynthesis pathway with the small molecule amlodipine reduced viral infection. In addition, we show that loss of RAB7A reduces viral entry by sequestering ACE2 receptors inside cells through altered endosomal trafficking. Prior to this study, our knowledge of essential host genes for SARS-CoV-2 has been limited to only a handful of genes, such as ACE2 and cathepsin L: this work provides a quantitative resource of the impact of each gene’s loss on response to viral infection for every protein-coding gene in the human genome.
|
|
|
Post by Admin on Nov 23, 2020 19:46:52 GMT
Results A High-Throughput Screen to Identify Genes Required for SARS-CoV-2 Infection To identify key genes required for SARS-CoV-2 infection, we performed a genome-scale loss-of-function screen targeting 19,050 genes in the human genome using the GeCKOv2 CRISPR-Cas9 library (Sanjana et al., 2014). The GeCKOv2 library contains 122,411 CRISPR single-guide RNAs (sgRNAs) (6 guide RNAs per gene) and has previously been used in CRISPR screens for drug resistance, immunotherapy, synthetic lethality, mitochondrial disease, and therapeutic discovery for muscular dystrophy (Erb et al., 2017; Jain et al., 2016; Lek et al., 2020; Patel et al., 2017; Shalem et al., 2014). First, we transduced a human alveolar basal epithelial carcinoma cell line (A549) that constitutively expresses ACE2 (referred to as A549ACE2) with an all-in-one lentiviral vector containing Cas9, guide RNAs from the GeCKOv2 human library, and a puromycin resistance gene. The transduction was performed at a low multiplicity of infection (MOI ∼0.2) to ensure that most cells would receive only one guide RNA construct (Figure 1A). We then selected with puromycin so that only library-transduced cells remained. We also measured the survival rate after puromycin selection was complete (3 days) to ensure high coverage of the 122,411 guide RNAs (∼1,000 cells per guide RNA). After puromycin selection was complete, we cultured the cells for 9 days to ensure protein depletion after CRISPR gene targeting. Figure 1 A Genome-Scale CRISPR Loss-of-Function Screen to Identify Genes that Prevent SARS-CoV-2 Infection of Human Alveolar Epithelial Carcinoma Cells Next, we infected the GeCKOv2 pool of A549ACE2 cells with SARS-CoV-2 virus (Isolate USA-WA1/2020 NR-52281) at either a high (0.3) or a low (0.01) MOI. We verified that SARS-CoV-2 infects A549ACE2 cells by staining for the nucleocapsid (N) protein at 24 h post-infection (Figure 1B), and at day 6 post-infection, we measured cell survival for both the high and low MOI conditions (Figure 1C). As expected, the higher MOI infection resulted in fewer surviving cells at day 6 post-infection. Next, we extracted genomic DNA, and via amplicon sequencing, we quantified guide abundance in each biological condition (Figure 1A). To confirm that library representation was properly maintained, we computed the correlation between the guide representation in the plasmid library and after puromycin selection (r = 0.84) (Figure S1A). In contrast, after SARS-CoV-2 infection, there was a much greater degree of guide dropout, as expected given that SARS-CoV-2 rapidly kills A549ACE2 cells without CRISPR perturbations (Figures 1C and 1D). Figure S1 Genome-wide Loss-of-Function CRISPR Screen Enriched Gene Identification, Related to Figure 1 Show full caption Using robust-rank aggregation (RRA) on the guide relative enrichments, we computed gene-level scores to identify genes where loss-of-function mutations led to enrichment within the pool (Figure 1E; Kolde et al., 2012). We identified ∼1,000 genes with significant RRA enrichment (p < 0.05) (Figure S1B). We also used two other previously published methods to compute gene enrichments (RIGER weighted-sum and second-best rank) and found a high degree of overlap between enriched genes identified by all three methods (Figure S1C; Chen et al., 2015; Luo et al., 2008). We also found a high degree of shared genes across both the low and high SARS-CoV-2 MOI conditions: when examining the top 50 most enriched genes, we found that 27 of them were shared between the low and high MOI conditions (Figure 1F; Table S1), suggesting that several host genes involved in viral pathogenesis function independently of viral dose. An independent genome-scale CRISPR screen for SARS-CoV-2 infection also performed in A549 that overexpress ACE2 but with a different CRISPR library identified similar top-ranked genes (Zhu et al., 2020), highlighting the robustness of our results (Figure S1D). Enriched Genes Are Involved in Multiple Aspects of the Viral Life Cycle and Are Broadly Expressed Upon close examination of the most enriched genes, we found genes involved in key aspects of viral entry and replication (Figure 2; Du et al., 2009). For example, the well-established entry receptor angiotensin-converting enzyme 2 (ACE2) receptor was ranked as the 8th most-enriched gene in the low MOI screen and 12th in the high MOI screen (Table S1; Hoffmann et al., 2020a; Zhou et al., 2020). Among the top 50 enriched genes, we identified several sets of related genes that function together in complexes, giving us further confidence in the genome-scale screen (Figures 2 and 3A ). We found genes essential for initial attachment and endocytosis (ACE2, RAB7A, and 4 members of the ARP2/3 complex: ACTR2, ACTR3, ARPC3, and ARPC4), Spike protein cleavage and viral membrane fusion (CTSL and 13 members of the vacuolar-ATPase proton pump: ATP6AP1, ATP6AP2, ATP6V0B ATP6V0C, ATP6V0D1, ATP6V1A, ATP6V1B2, ATP6V1C1, ATP6V1E1, ATP6V1G1, ATP6V1H, TMEM199, and TOR1AIP1), endosome recycling (4 members of the endosomal protein sorting Retromer complex: VPS26A, VPS29, VPS35, and SNX27; 4 members of the endosomal trafficking Commander complex: COMMD2, COMMD3, COMMD3-BMI1, and COMMD4; and 3 members of the PI3K pathway: PIK3C3/VPS34, WDR81, and ACP5), ER-Golgi trafficking (DPM3, ERMP1, PPID, and CHST14), and transcriptional modulators (SLTM and SPEN). A consistent theme among the enriched complexes is endosome function and regulation (V-ATPase proton pump, Retromer, Commander, class 3 PI3Ks) (Banerjee and Kane, 2020; Mallam and Marcotte, 2017; McNally and Cullen, 2018). Gene set enrichment analysis on the full ranked list of genes identified significantly enriched Gene Ontology (GO) categories for endosome processing, transport, and acidification and categories related to cytokinesis and virion attachment (false discovery rate [FDR] q < 0.1) (Figures 3B and S2A–S2D ; Table S2; Subramanian et al., 2005).
|
|
|
Post by Admin on Nov 24, 2020 4:40:33 GMT
Figure 2 Top-Ranked Genes from the CRISPR Screen Are Involved in Key Elements of the SARS-CoV-2 Viral Life Cycle Figure 3 Enriched Genes Cluster into Related Pathways, Are Expressed Broadly, Interact Directly with Viral Proteins, and Are Also Involved in Viral Pathogenesis of Pandemic Flu and Zika Virus Figure S2 Gene Set Enrichment and Overlap of Top-Ranked Genes with Other Viral Infections, Related to Figures 2 and 3 Although we performed our CRISPR screen in human lung cells, we explored whether the expression of host genes whose loss reduces SARS-CoV-2 infection were lung-specific or more broadly expressed. To answer this question, we took the top-ranked genes and examined their expression across 12 tissues using 4,790 RNA-sequencing datasets from the Genotype-Tissue Expression (GTEx) v8 database (Figure 3C; Aguet et al., 2019). Virtually all of the top gene hits were broadly expressed across all tissues, implying that these mechanisms may function independent of cell or tissue type. Among the top-ranked genes, only ACE2 showed tissue-specific expression with a particular enrichment in testis, small intestine, kidney, and heart (Figure 3C). Enriched Genes Have Been Suggested to Interact with Viral Proteins and Are Also Essential for Other Viral Pathogens Recently, Gordon et al. (2020) performed an in-depth study of SARS-CoV-2 protein-protein interaction networks by overexpressing affinity-tagged versions of each protein encoded in the viral genome followed by tandem mass spectrometry after pull-down. Their study identified 332 high-confidence SARS-CoV-2-human protein-protein interactions (PPIs). We found that some of the highly ranked genes from our loss-of-function screen were reported to have direct PPI with different viral proteins (Figure 3D; Table S2). For example, two highly ranked subunits of the vacuolar-ATPase proton pump, ATP6AP1 and ATP6V1A, interact with SARS-CoV-2 non-structural protein 6 (nsp6) and membrane (M) protein, respectively. ATP6AP1, which was ranked 2nd in the low MOI CRISPR screen and 4th in the high MOI CRISPR screen, has a very strong PPI interaction with nsp6 (mass spectrometry interaction statistics [MIST] score = 0.99) (Verschueren et al., 2015). Another key endocytosis protein, RAB7A, is ranked in the top 50 genes in both CRISPR screens and interacts strongly with non-structural protein 7 (nsp7) (MIST score = 0.97). We also compared the top-ranked genes with another proteomic study that used proximity labeling in A549 cells overexpressing BioID-tagged viral proteins and found that 22 out of the top 50 low MOI CRISPR screen genes had direct interactions with viral genes—a significant enrichment over randomly chosen genes (p = 2 × 10−4) (Samavarchi-Tehrani et al., 2020). Because similar loss-of-function CRISPR screens have been performed to identify host genes required for other viral pathogens, we next sought to understand whether the hits identified in our SARS-CoV-2 screen were shared with those identified in prior screens for Zika virus (ZIKV) and pandemic H1N1 avian influenza (IAV) (Li et al., 2019, 2020). We examined whether top-ranked hits from the ZIKV and IAV screens shared similar genes and similar functional categories. Overall, there was greater similarity between GO categories of enriched genes for SARS-CoV-2 and ZIKV (Figure 3E; Table S2). When examining the top 50 genes from the SARS-CoV-2 screen, we found several genes that were highly enriched in all 3 viral pathogen screens (Figure S2E). This group included subunits of the vacuolar-ATPase proton pump, a well-known family of genes essential for acidification and endosomal processing (Banerjee and Kane, 2020).
|
|