44 results on '"Rice, Charles M."'
Search Results
2. Long-Term Expansion of Functional Mouse and Human Hepatocytes as 3D Organoids
- Author
-
TN Onderwijs, Hubrecht Institute with UMC, CMM Sectie Molecular Cancer Research, Cancer, Child Health, Regenerative Medicine and Stem Cells, Brain, Hu, Huili, Gehart, Helmuth, Artegiani, Benedetta, LÖpez-Iglesias, Carmen, Dekkers, Florijn, Basak, Onur, van Es, Johan, Chuva de Sousa Lopes, Susana M., Begthel, Harry, Korving, Jeroen, van den Born, Maaike, Zou, Chenhui, Quirk, Corrine, Chiriboga, Luis, Rice, Charles M., Ma, Stephanie, Rios, Anne, Peters, Peter J., de Jong, Ype P., Clevers, Hans, TN Onderwijs, Hubrecht Institute with UMC, CMM Sectie Molecular Cancer Research, Cancer, Child Health, Regenerative Medicine and Stem Cells, Brain, Hu, Huili, Gehart, Helmuth, Artegiani, Benedetta, LÖpez-Iglesias, Carmen, Dekkers, Florijn, Basak, Onur, van Es, Johan, Chuva de Sousa Lopes, Susana M., Begthel, Harry, Korving, Jeroen, van den Born, Maaike, Zou, Chenhui, Quirk, Corrine, Chiriboga, Luis, Rice, Charles M., Ma, Stephanie, Rios, Anne, Peters, Peter J., de Jong, Ype P., and Clevers, Hans
- Published
- 2018
3. The RNA Sensor RIG-I Dually Functions as an Innate Sensor and Direct Antiviral Factor for Hepatitis B Virus
- Author
-
Sato, Seiichi, Li, Kai, Kameyama, Takeshi, Hayashi, Takaya, Ishida, Yuji, Murakami, Shuko, Watanabe, Tsunamasa, Iijima, Sayuki, Sakurai, Yu, Watashi, Koichi, Tsutsumi, Susumu, Sato, Yusuke, Akita, Hidetaka, Wakita, Takaji, Rice, Charles M., Harashima, Hideyoshi, Kohara, Michinori, Tanaka, Yasuhito, Takaoka, Akinori, Sato, Seiichi, Li, Kai, Kameyama, Takeshi, Hayashi, Takaya, Ishida, Yuji, Murakami, Shuko, Watanabe, Tsunamasa, Iijima, Sayuki, Sakurai, Yu, Watashi, Koichi, Tsutsumi, Susumu, Sato, Yusuke, Akita, Hidetaka, Wakita, Takaji, Rice, Charles M., Harashima, Hideyoshi, Kohara, Michinori, Tanaka, Yasuhito, and Takaoka, Akinori
- Abstract
Host innate recognition triggers key immune responses for viral elimination. The sensing mechanism of hepatitis B virus (HBV), a DNA virus, and the subsequent downstream signaling events remain to be fully clarified. Here we found that type III but not type I interferons are predominantly induced in human primary hepatocytes in response to HBV infection, through retinoic acid-inducible gene-I (RIG-I)-mediated sensing of the 5'-ε region of HBV pregenomic RNA. In addition, RIG-I could also counteract the interaction of HBV polymerase (P protein) with the 5'-ε region in an RNA-binding dependent manner, which consistently suppressed viral replication. Liposome-mediated delivery and vector-based expression of this ε region-derived RNA in liver abolished the HBV replication in human hepatocyte-chimeric mice. These findings identify an innate recognition mechanism by which RIG-I dually functions as an HBV sensor activating innate signaling and to counteract viral polymerase in human hepatocytes., Supplemental materials are available on the publisher's website.
- Published
- 2015
4. Identification of Interferon-Stimulated Genes with Antiretroviral Activity.
- Author
-
Kane, Melissa, Zang, Trinity M., Rihn, Suzannah J., Zhang, Fengwen, Kueck, Tonya, Alim, Mudathir, Schoggins, John, Rice, Charles M., Wilson, Sam J., and Bieniasz, Paul D.
- Abstract
Summary Interferons (IFNs) exert their anti-viral effects by inducing the expression of hundreds of IFN-stimulated genes (ISGs). The activity of known ISGs is insufficient to account for the antiretroviral effects of IFN, suggesting that ISGs with antiretroviral activity are yet to be described. We constructed an arrayed library of ISGs from rhesus macaques and tested the ability of hundreds of individual macaque and human ISGs to inhibit early and late replication steps for 11 members of the retroviridae from various host species. These screens uncovered numerous ISGs with antiretroviral activity at both the early and late stages of virus replication. Detailed analyses of two antiretroviral ISGs indicate that indoleamine 2,3-dioxygenase 1 (IDO1) can inhibit retroviral replication by metabolite depletion while tripartite motif-56 (TRIM56) accentuates ISG induction by IFNα and inhibits the expression of late HIV-1 genes. Overall, these studies reveal numerous host proteins that mediate the antiretroviral activity of IFNs. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
5. A Broad RNA Virus Survey Reveals Both miRNA Dependence and Functional Sequestration.
- Author
-
Scheel, Troels K.H., Luna, Joseph M., Liniger, Matthias, Nishiuchi, Eiko, Rozen-Gagnon, Kathryn, Shlomai, Amir, Auray, Gaël, Gerber, Markus, Fak, John, Keller, Irene, Bruggmann, Rémy, Darnell, Robert B., Ruggli, Nicolas, and Rice, Charles M.
- Abstract
Summary Small non-coding RNAs have emerged as key modulators of viral infection. However, with the exception of hepatitis C virus, which requires the liver-specific microRNA (miRNA)-122, the interactions of RNA viruses with host miRNAs remain poorly characterized. Here, we used crosslinking immunoprecipitation (CLIP) of the Argonaute (AGO) proteins to characterize strengths and specificities of miRNA interactions in the context of 15 different RNA virus infections, including several clinically relevant pathogens. Notably, replication of pestiviruses, a major threat to milk and meat industries, critically depended on the interaction of cellular miR-17 and let-7 with the viral 3′ UTR. Unlike canonical miRNA interactions, miR-17 and let-7 binding enhanced pestivirus translation and RNA stability. miR-17 sequestration by pestiviruses conferred reduced AGO binding and functional de-repression of cellular miR-17 targets, thereby altering the host transcriptome. These findings generalize the concept of RNA virus dependence on cellular miRNAs and connect virus-induced miRNA sequestration to host transcriptome regulation. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
6. Interferon Lambda Alleles Predict Innate Antiviral Immune Responses and Hepatitis C Virus Permissiveness.
- Author
-
Sheahan, Timothy, Imanaka, Naoko, Marukian, Svetlana, Dorner, Marcus, Liu, Peng, Ploss, Alexander, and Rice, Charles M.
- Abstract
Summary: Hepatitis C virus (HCV) infection can result in viral chronicity or clearance. Although host genetics and particularly genetic variation in the interferon lambda (IFNL) locus are associated with spontaneous HCV clearance and treatment success, the mechanisms guiding these clinical outcomes remain unknown. Using a laser capture microdissection-driven unbiased systems virology approach, we isolated and transcriptionally profiled HCV-infected and adjacent primary human hepatocytes (PHHs) approaching single-cell resolution. An innate antiviral immune signature dominated the transcriptional response but differed in magnitude and diversity between HCV-infected and adjacent cells. Molecular signatures associated with more effective antiviral control were determined by comparing donors with high and low infection frequencies. Cells from donors with clinically unfavorable IFNL genotypes were infected at a greater frequency and exhibited dampened antiviral and cell death responses. These data suggest that early virus-host interactions, particularly host genetics and induction of innate immunity, critically determine the outcome of HCV infection. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
7. Inhibition of HIV-1 Particle Assembly by 2′,3′-Cyclic-Nucleotide 3′-Phosphodiesterase.
- Author
-
Wilson, Sam J., Schoggins, John W., Zang, Trinity, Kutluay, Sebla B., Jouvenet, Nolwenn, Alim, Mudathir A., Bitzegeio, Julia, Rice, Charles M., and Bieniasz, Paul D.
- Subjects
CYCLIC nucleotides ,GENE expression ,PHOSPHODIESTERASE inhibitors ,ANTIVIRAL agents ,INTERFERONS ,DRUG activation ,VIRAL proteins ,THERAPEUTICS ,HIV infections - Abstract
Summary: The expression of hundreds of interferon-stimulated genes (ISGs) causes the cellular “antiviral state” in which the replication of many viruses, including HIV-1, is attenuated. We conducted a screen for ISGs that inhibit HIV-1 virion production and found that 2′,3′-cyclic-nucleotide 3′-phosphodiesterase (CNP), a membrane-associated protein with unknown function in mammals has this property. CNP binds to the structural protein Gag and blocks HIV-1 particle assembly after Gag and viral RNA have associated with the plasma membrane. Several primate lentiviruses are CNP-sensitive, and CNP sensitivity/resistance is determined by a single, naturally dimorphic, codon (E/K40) in the matrix domain of Gag. Like other antiretroviral proteins, CNP displays interspecies variation in antiviral activity. Mice encode an inactive CNP variant and a single amino acid difference in murine versus human CNP determines Gag binding and antiviral activity. Some cell types express high levels of CNP and we speculate that CNP evolved to restrict lentivirus replication therein. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
8. Antiviral innate immune memory in alveolar macrophages following SARS-CoV-2 infection ameliorates secondary influenza A virus disease.
- Author
-
Lercher, Alexander, Cheong, Jin-Gyu, Bale, Michael J., Jiang, Chenyang, Hoffmann, Hans-Heinrich, Ashbrook, Alison W., Lewy, Tyler, Yin, Yue S., Quirk, Corrine, DeGrace, Emma J., Chiriboga, Luis, Rosenberg, Brad R., Josefowicz, Steven Z., and Rice, Charles M.
- Subjects
- *
IMMUNOLOGIC memory , *VIRUS diseases , *ALVEOLAR macrophages , *TYPE I interferons , *INFLUENZA A virus - Abstract
Pathogen encounter can result in epigenetic remodeling that shapes disease caused by heterologous pathogens. Here, we examined innate immune memory in the context of commonly circulating respiratory viruses. Single-cell analyses of airway-resident immune cells in a disease-relevant murine model of SARS-CoV-2 recovery revealed epigenetic reprogramming in alveolar macrophages following infection. Post-COVID-19 human monocytes exhibited similar epigenetic signatures. In airway-resident macrophages, past SARS-CoV-2 infection increased activity of type I interferon (IFN-I)-related transcription factors and epigenetic poising of antiviral genes. Viral pattern recognition and canonical IFN-I signaling were required for the establishment of this innate immune memory and augmented secondary antiviral responses. Antiviral innate immune memory mounted by airway-resident macrophages post-SARS-CoV-2 was necessary and sufficient to ameliorate secondary disease caused by influenza A virus and curtailed hyperinflammatory dysregulation and mortality. Our findings provide insights into antiviral innate immune memory in the airway that may facilitate the development of broadly effective therapeutic strategies. [Display omitted] • Airway-resident macrophages (AMF) establish durable epigenetic memory post SARS2 • Chromatin loci of ISGs retain increased accessibility and are decorated with H3K4me1 • AMFs hyper-induce ISGs upon secondary antiviral recall responses • Innate immune memory in AMF ameliorates secondary influenza A virus disease The therapeutic potential of antigen-independent innate immune memory (IIM) is of particular relevance in the context of respiratory viruses with pandemic potential. Lercher et al. find that antiviral IIM in alveolar macrophages following SARS-CoV-2 infection ameliorates disease caused by a secondary unrelated pathogen, influenza A virus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Argonaute-CLIP delineates versatile, functional RNAi networks in Aedes aegypti, a major vector of human viruses.
- Author
-
Rozen-Gagnon, Kathryn, Gu, Meigang, Luna, Joseph M., Luo, Ji-Dung, Yi, Soon, Novack, Sasha, Jacobson, Eliana, Wang, Wei, Paul, Matthew R., Scheel, Troels K.H., Carroll, Thomas, and Rice, Charles M.
- Abstract
Argonaute (AGO) proteins bind small RNAs to silence complementary RNA transcripts, and they are central to RNA interference (RNAi). RNAi is critical for regulation of gene expression and antiviral defense in Aedes aegypti mosquitoes, which transmit Zika, chikungunya, dengue, and yellow fever viruses. In mosquitoes, AGO1 mediates miRNA interactions, while AGO2 mediates siRNA interactions. We applied AGO-crosslinking immunoprecipitation (AGO-CLIP) for both AGO1 and AGO2, and we developed a universal software package for CLIP analysis (CLIPflexR), identifying 230 small RNAs and 5,447 small RNA targets that comprise a comprehensive RNAi network map in mosquitoes. RNAi network maps predicted expression levels of small RNA targets in specific tissues. Additionally, this resource identified unexpected, context-dependent AGO2 target preferences, including endogenous viral elements and 3′UTRs. Finally, contrary to current thinking, mosquito AGO2 repressed imperfect targets. These findings expand our understanding of small RNA networks and have broad implications for the study of antiviral RNAi. [Display omitted] • Established AGO-CLIP in Ae. aegypti and CLIPflexR, a universal CLIP analysis package • AGO1 and AGO2 RNAi network maps predict tissue-specific target expression • AGO2 binding of 3′UTRs, transposons, and endogenous viral elements is context-dependent • Mosquito AGO2 can repress imperfect, 3′UTR targets in an AGO1-like fashion In the arbovirus vector Ae. aegypti , RNAi regulates gene expression and defends against viral infection. Rozen-Gagnon et al. developed CLIP and a software package, CLIPflexR, to map transcriptome-wide RNAi networks in mosquitoes. Network maps revealed that AGO2, the effector of antiviral RNAi, has flexible target preferences and represses imperfect targets. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Functional interrogation of a SARS-CoV-2 host protein interactome identifies unique and shared coronavirus host factors.
- Author
-
Hoffmann, H.-Heinrich, Sánchez-Rivera, Francisco J., Schneider, William M., Luna, Joseph M., Soto-Feliciano, Yadira M., Ashbrook, Alison W., Le Pen, Jérémie, Leal, Andrew A., Ricardo-Lax, Inna, Michailidis, Eleftherios, Hao, Yuan, Stenzel, Ansgar F., Peace, Avery, Zuber, Johannes, Allis, C. David, Lowe, Scott W., MacDonald, Margaret R., Poirier, John T., and Rice, Charles M.
- Abstract
The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has devastated the global economy and claimed more than 1.7 million lives, presenting an urgent global health crisis. To identify host factors required for infection by SARS-CoV-2 and seasonal coronaviruses, we designed a focused high-coverage CRISPR-Cas9 library targeting 332 members of a recently published SARS-CoV-2 protein interactome. We leveraged the compact nature of this library to systematically screen SARS-CoV-2 at two physiologically relevant temperatures along with three related coronaviruses (human coronavirus 229E [HCoV-229E], HCoV-NL63, and HCoV-OC43), allowing us to probe this interactome at a much higher resolution than genome-scale studies. This approach yielded several insights, including potential virus-specific differences in Rab GTPase requirements and glycosylphosphatidylinositol (GPI) anchor biosynthesis, as well as identification of multiple pan-coronavirus factors involved in cholesterol homeostasis. This coronavirus essentiality catalog could inform ongoing drug development efforts aimed at intercepting and treating coronavirus disease 2019 (COVID-19) and help prepare for future coronavirus outbreaks. • SARS-CoV-2 host protein interactome CRISPR screens for SARS-CoV-2 and three coronaviruses • Parallel CRISPR screens uncover unique and shared coronavirus host factors • Numbers of interacting host proteins and functional interactors are not proportional • Identified SARS-CoV-2 host factors are expressed in relevant cells in the human airway Building upon a published SARS-CoV-2 protein interactome, Hoffmann et al. use a custom CRISPR library to determine which of these interacting host proteins are essential for infection by SARS-CoV-2 virus as well as three seasonal coronaviruses. These factors represent potential targets to combat COVID-19 and perhaps future coronavirus outbreaks. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. RNAi Targeting an Animal Virus: News from the Front
- Author
-
Lindenbach, Brett D. and Rice, Charles M.
- Subjects
- *
RNA , *EUKARYOTIC cells - Abstract
Although many eukaryotic organisms exhibit RNA interference, its role as an antiviral pathway is established only for plants. A recent paper demonstrates that it also acts in animals, and reveals a viral suppressor of this process. [Copyright &y& Elsevier]
- Published
- 2002
- Full Text
- View/download PDF
12. Diverse Viruses Require the Calcium Transporter SPCA1 for Maturation and Spread.
- Author
-
Hoffmann, H.-Heinrich, Schneider, William M., Blomen, Vincent A., Scull, Margaret A., Hovnanian, Alain, Brummelkamp, Thijn R., and Rice, Charles M.
- Abstract
Summary Respiratory and arthropod-borne viral infections are a global threat due to the lack of effective antivirals and vaccines. A potential strategy is to target host proteins required for viruses but non-essential for the host. To identify such proteins, we performed a genome-wide knockout screen in human haploid cells and identified the calcium pump SPCA1. SPCA1 is required by viruses from the Paramyxoviridae , Flaviviridae , and Togaviridae families, including measles, dengue, West Nile, Zika, and chikungunya viruses. Calcium transport activity is required for SPCA1 to promote virus spread. SPCA1 regulates proteases within the trans- Golgi network that require calcium for their activity and are critical for virus glycoprotein maturation. Consistent with these findings, viral glycoproteins fail to mature in SPCA1-deficient cells preventing viral spread, which is evident even in cells with partial loss of SPCA1. Thus, SPCA1 is an attractive antiviral host target for a broad spectrum of established and emerging viral infections. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
13. Deep mutational scanning of hepatitis B virus reveals a mechanism for cis-preferential reverse transcription.
- Author
-
Yu, Yingpu, Kass, Maximilian A., Zhang, Mengyin, Youssef, Noor, Freije, Catherine A., Brock, Kelly P., Aguado, Lauren C., Seifert, Leon L., Venkittu, Sanjana, Hong, Xupeng, Shlomai, Amir, de Jong, Ype P., Marks, Debora S., Rice, Charles M., and Schneider, William M.
- Subjects
- *
HEPATITIS B virus , *MESSENGER RNA , *RIBOSOMES , *POLYMERASES , *DNA viruses - Abstract
Hepatitis B virus (HBV) is a small double-stranded DNA virus that chronically infects 296 million people. Over half of its compact genome encodes proteins in two overlapping reading frames, and during evolution, multiple selective pressures can act on shared nucleotides. This study combines an RNA-based HBV cell culture system with deep mutational scanning (DMS) to uncouple cis - and trans -acting sequence requirements in the HBV genome. The results support a leaky ribosome scanning model for polymerase translation, provide a fitness map of the HBV polymerase at single-nucleotide resolution, and identify conserved prolines adjacent to the HBV polymerase termination codon that stall ribosomes. Further experiments indicated that stalled ribosomes tether the nascent polymerase to its template RNA, ensuring cis -preferential RNA packaging and reverse transcription of the HBV genome. [Display omitted] • Deep mutational scanning (DMS) provides a high-resolution fitness map of HBV polymerase • Initiating HBV replication with RNA uncouples cis - and trans -acting protein functions • Ribosome pausing tethers HBV Pol to RNA, enforcing cis -preferential reverse transcription • DMS of the HBV genome supports a leaky ribosome scanning model for polymerase translation Deep mutational scanning reveals that conserved proline codons at the 3′ end of the hepatitis B virus polymerase open reading frame stall ribosomes. As a result, the ribosome physically tethers the nascent polymerase to its encoding RNA, which enforces cis -preferential genome RNA packaging and reverse transcription. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. The IFN-λ-IFN-λR1-IL-10Rβ Complex Reveals Structural Features Underlying Type III IFN Functional Plasticity.
- Author
-
Mendoza, Juan L., Schneider, William M., Hoffmann, Hans-Heinrich, Vercauteren, Koen, Jude, Kevin M., Xiong, Anming, Moraga, Ignacio, Horton, Tim M., Glenn, Jeffrey S., de Jong, Ype P., Rice, Charles M., and Garcia, K. Christopher
- Subjects
- *
INTERFERONS , *INTERLEUKIN-18 , *CYTOKINE receptors , *CELLULAR signal transduction , *TYROSINE - Abstract
Summary Type III interferons (IFN-λs) signal through a heterodimeric receptor complex composed of the IFN-λR1 subunit, specific for IFN-λs, and interleukin-10Rβ (IL-10Rβ), which is shared by multiple cytokines in the IL-10 superfamily. Low affinity of IL-10Rβ for cytokines has impeded efforts aimed at crystallizing cytokine-receptor complexes. We used yeast surface display to engineer a higher-affinity IFN-λ variant, H11, which enabled crystallization of the ternary complex. The structure revealed that IL-10Rβ uses a network of tyrosine residues as hydrophobic anchor points to engage IL-10 family cytokines that present complementary hydrophobic binding patches, explaining its role as both a cross-reactive but cytokine-specific receptor. H11 elicited increased anti-proliferative and antiviral activities in vitro and in vivo. In contrast, engineered higher-affinity type I IFNs did not increase antiviral potency over wild-type type I IFNs. Our findings provide insight into cytokine recognition by the IL-10R family and highlight the plasticity of type III interferon signaling and its therapeutic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
15. Engineering a Therapeutic Lectin by Uncoupling Mitogenicity from Antiviral Activity.
- Author
-
Swanson, Michael D., Boudreaux, Daniel M., Salmon, Loïc, Chugh, Jeetender, Winter, Harry C., Meagher, Jennifer L., André, Sabine, Murphy, Paul V., Oscarson, Stefan, Roy, René, King, Steven, Kaplan, Mark H., Goldstein, Irwin J., Tarbet, E. Bart, Hurst, Brett L., Smee, Donald F., de la Fuente, Cynthia, Hoffmann, Hans-Heinrich, Xue, Yi, and Rice, Charles M.
- Subjects
- *
LECTINS , *ANTIVIRAL agents , *MITOGENS , *SUBSTITUTION reactions , *BIOENGINEERING , *TARGETED drug delivery , *THERAPEUTICS - Abstract
Summary A key effector route of the Sugar Code involves lectins that exert crucial regulatory controls by targeting distinct cellular glycans. We demonstrate that a single amino-acid substitution in a banana lectin, replacing histidine 84 with a threonine, significantly reduces its mitogenicity, while preserving its broad-spectrum antiviral potency. X-ray crystallography, NMR spectroscopy, and glycocluster assays reveal that loss of mitogenicity is strongly correlated with loss of pi-pi stacking between aromatic amino acids H84 and Y83, which removes a wall separating two carbohydrate binding sites, thus diminishing multivalent interactions. On the other hand, monovalent interactions and antiviral activity are preserved by retaining other wild-type conformational features and possibly through unique contacts involving the T84 side chain. Through such fine-tuning, target selection and downstream effects of a lectin can be modulated so as to knock down one activity, while preserving another, thus providing tools for therapeutics and for understanding the Sugar Code. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
16. Hepatitis C Virus RNA Functionally Sequesters miR-122.
- Author
-
Luna, Joseph M., Scheel, Troels K.H., Danino, Tal, Shaw, Katharina S., Mele, Aldo, Fak, John J., Nishiuchi, Eiko, Takacs, Constantin N., Catanese, Maria Teresa, de Jong, Ype P., Jacobson, Ira M., Rice, Charles M., and Darnell, Robert B.
- Subjects
- *
HEPATITIS C , *VIRAL genetics , *MICRORNA , *VIRAL replication , *IMMUNOPRECIPITATION , *GENE targeting , *GENETICS - Abstract
Summary Hepatitis C virus (HCV) uniquely requires the liver-specific microRNA-122 for replication, yet global effects on endogenous miRNA targets during infection are unexplored. Here, high-throughput sequencing and crosslinking immunoprecipitation (HITS-CLIP) experiments of human Argonaute (AGO) during HCV infection showed robust AGO binding on the HCV 5′UTR at known and predicted miR-122 sites. On the human transcriptome, we observed reduced AGO binding and functional mRNA de-repression of miR-122 targets during virus infection. This miR-122 “sponge” effect was relieved and redirected to miR-15 targets by swapping the miRNA tropism of the virus. Single-cell expression data from reporters containing miR-122 sites showed significant de-repression during HCV infection depending on expression level and site number. We describe a quantitative mathematical model of HCV-induced miR-122 sequestration and propose that such miR-122 inhibition by HCV RNA may result in global de-repression of host miR-122 targets, providing an environment fertile for the long-term oncogenic potential of HCV. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
17. A Serpin Shapes the Extracellular Environment to Prevent Influenza A Virus Maturation.
- Author
-
Dittmann, Meike, Hoffmann, Hans-Heinrich, Scull, Margaret A., Gilmore, Rachel H., Bell, Kierstin L., Ciancanelli, Michael, Wilson, Sam J., Crotta, Stefania, Yu, Yingpu, Flatley, Brenna, Xiao, Jing W., Casanova, Jean-Laurent, Wack, Andreas, Bieniasz, Paul D., and Rice, Charles M.
- Subjects
- *
SERPINS , *INFLUENZA A virus , *INTERFERONS , *VIRAL genetics , *PLASMINOGEN activator inhibitors , *ANTIVIRAL agents , *BIOCHEMICAL mechanism of action - Abstract
Summary Interferon-stimulated genes (ISGs) act in concert to provide a tight barrier against viruses. Recent studies have shed light on the contribution of individual ISG effectors to the antiviral state, but most have examined those acting on early, intracellular stages of the viral life cycle. Here, we applied an image-based screen to identify ISGs inhibiting late stages of influenza A virus (IAV) infection. We unraveled a directly antiviral function for the gene SERPINE1 , encoding plasminogen activator inhibitor 1 (PAI-1). By targeting extracellular airway proteases, PAI-1 inhibits IAV glycoprotein cleavage, thereby reducing infectivity of progeny viruses. This was biologically relevant for IAV restriction in vivo. Further, partial PAI-1 deficiency, attributable to a polymorphism in human SERPINE1 , conferred increased susceptibility to IAV in vitro. Together, our findings reveal that manipulating the extracellular environment to inhibit the last step in a virus life cycle is an important mechanism of the antiviral response. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. TMEM41B Is a Pan-flavivirus Host Factor.
- Author
-
Hoffmann, H.-Heinrich, Schneider, William M., Rozen-Gagnon, Kathryn, Miles, Linde A., Schuster, Felix, Razooky, Brandon, Jacobson, Eliana, Wu, Xianfang, Yi, Soon, Rudin, Charles M., MacDonald, Margaret R., McMullan, Laura K., Poirier, John T., and Rice, Charles M.
- Subjects
- *
FLAVIVIRUSES , *FLAVIVIRAL diseases , *EAST Asians , *VIRAL genomes , *MEMBRANE proteins , *RNA viruses - Abstract
Flaviviruses pose a constant threat to human health. These RNA viruses are transmitted by the bite of infected mosquitoes and ticks and regularly cause outbreaks. To identify host factors required for flavivirus infection, we performed full-genome loss of function CRISPR-Cas9 screens. Based on these results, we focused our efforts on characterizing the roles that TMEM41B and VMP1 play in the virus replication cycle. Our mechanistic studies on TMEM41B revealed that all members of the Flaviviridae family that we tested require TMEM41B. We tested 12 additional virus families and found that SARS-CoV-2 of the Coronaviridae also required TMEM41B for infection. Remarkably, single nucleotide polymorphisms present at nearly 20% in East Asian populations reduce flavivirus infection. Based on our mechanistic studies, we propose that TMEM41B is recruited to flavivirus RNA replication complexes to facilitate membrane curvature, which creates a protected environment for viral genome replication. • TMEM41B is required for flavivirus infection, but autophagy is not required • TMEM41B associates with flavivirus proteins and may facilitate cell membrane remodeling • TMEM41B single nucleotide polymorphisms reduce flavivirus infection • TMEM41B-deficiency confers a heightened innate immune response to flavivirus infection Hoffmann et al. determine that the transmembrane protein, TMEM41B, is required for infection by members of the Flaviviridae family of viruses. Loss of TMEM41B reduces viral RNA replication and increases innate immune activation in response to flavivirus infection. Thus, TMEM41B is a potential host target for antiviral therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. Genome-Scale Identification of SARS-CoV-2 and Pan-coronavirus Host Factor Networks.
- Author
-
Schneider, William M., Luna, Joseph M., Hoffmann, H.-Heinrich, Sánchez-Rivera, Francisco J., Leal, Andrew A., Ashbrook, Alison W., Le Pen, Jérémie, Ricardo-Lax, Inna, Michailidis, Eleftherios, Peace, Avery, Stenzel, Ansgar F., Lowe, Scott W., MacDonald, Margaret R., Rice, Charles M., and Poirier, John T.
- Subjects
- *
SARS-CoV-2 , *COVID-19 , *STEROL regulatory element-binding proteins , *PANDEMICS , *BONE morphogenetic proteins , *RESPIRATORY infections , *GLYCOSAMINOGLYCANS - Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed the lives of over one million people worldwide. The causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a member of the Coronaviridae family of viruses that can cause respiratory infections of varying severity. The cellular host factors and pathways co-opted during SARS-CoV-2 and related coronavirus life cycles remain ill defined. To address this gap, we performed genome-scale CRISPR knockout screens during infection by SARS-CoV-2 and three seasonal coronaviruses (HCoV-OC43, HCoV-NL63, and HCoV-229E). These screens uncovered host factors and pathways with pan-coronavirus and virus-specific functional roles, including major dependency on glycosaminoglycan biosynthesis, sterol regulatory element-binding protein (SREBP) signaling, bone morphogenetic protein (BMP) signaling, and glycosylphosphatidylinositol biosynthesis, as well as a requirement for several poorly characterized proteins. We identified an absolute requirement for the VMP1, TMEM41, and TMEM64 (VTT) domain-containing protein transmembrane protein 41B (TMEM41B) for infection by SARS-CoV-2 and three seasonal coronaviruses. This human coronavirus host factor compendium represents a rich resource to develop new therapeutic strategies for acute COVID-19 and potential future coronavirus pandemics. • Genome-wide CRISPR screens for SARS-CoV-2 and seasonal coronavirus host factors • Identification of host factors and pathways with pan-coronavirus and discrete roles • Coronaviruses co-opt multiple biological pathways • TMEM41B is a critical pan-coronavirus host factor Schneider et al. conducted parallel genome-wide CRISPR knockout screens with SARS-CoV-2 and three seasonal coronaviruses to identify pan-coronavirus and virus-specific host factor requirements. They identified an interconnected network of host factors required by these four viruses and validated TMEM41B as a pan-coronavirus host factor required for a post-entry step in the coronavirus life cycle. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
20. Human ADAR1 Prevents Endogenous RNA from Triggering Translational Shutdown.
- Author
-
Chung, Hachung, Wu, Xianfang, Sun, Tony, Yu, Yingpu, Sarbanes, Stephanie L., Dao Thi, Viet Loan, Shilvock, Abigail R., Hoffmann, H.-Heinrich, Rice, Charles M., Calis, Jorg J.A., and Rosenberg, Brad R.
- Subjects
- *
ADENOSINES , *DOUBLE-stranded RNA , *AICARDI-Goutieres syndrome , *GENETIC mutation , *TYPE I interferons , *RNA polymerase II , *GENETICS - Abstract
Summary Type I interferon (IFN) is produced when host sensors detect foreign nucleic acids, but how sensors differentiate self from nonself nucleic acids, such as double-stranded RNA (dsRNA), is incompletely understood. Mutations in ADAR1, an adenosine-to-inosine editing enzyme of dsRNA, cause Aicardi-Goutières syndrome, an autoinflammatory disorder associated with spontaneous interferon production and neurologic sequelae. We generated ADAR1 knockout human cells to explore ADAR1 substrates and function. ADAR1 primarily edited Alu elements in RNA polymerase II (pol II)-transcribed mRNAs, but not putative pol III-transcribed Alus. During the IFN response, ADAR1 blocked translational shutdown by inhibiting hyperactivation of PKR, a dsRNA sensor. ADAR1 dsRNA binding and catalytic activities were required to fully prevent endogenous RNA from activating PKR. Remarkably, ADAR1 knockout neuronal progenitor cells exhibited MDA5 (dsRNA sensor)-dependent spontaneous interferon production, PKR activation, and cell death. Thus, human ADAR1 regulates sensing of self versus nonself RNA, allowing pathogen detection while avoiding autoinflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
21. Intrinsic Immunity Shapes Viral Resistance of Stem Cells.
- Author
-
Wu, Xianfang, Dao Thi, Viet Loan, Huang, Yumin, Billerbeck, Eva, Saha, Debjani, Hoffmann, Hans-Heinrich, Wang, Yaomei, Silva, Luis A. Vale, Sarbanes, Stephanie, Sun, Tony, Andrus, Linda, Yu, Yingpu, Quirk, Corrine, Li, Melody, MacDonald, Margaret R., Schneider, William M., An, Xiuli, Rosenberg, Brad R., and Rice, Charles M.
- Subjects
- *
STEM cells , *CELLULAR immunity , *VIRAL disease treatment , *ANTIVIRAL agents , *GENE expression , *INTERFERONS - Abstract
Summary Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
22. Argonaute CLIP Defines a Deregulated miR-122-Bound Transcriptome that Correlates with Patient Survival in Human Liver Cancer.
- Author
-
Luna, Joseph M., Barajas, Juan M., Teng, Kun-yu, Sun, Hui-Lung, Moore, Michael J., Rice, Charles M., Darnell, Robert B., and Ghoshal, Kalpana
- Subjects
- *
MICRORNA genetics , *LIVER cancer , *LIVER cancer patients , *BIOCHEMICAL engineering , *LABORATORY mice - Abstract
Summary MicroRNA-122, an abundant and conserved liver-specific miRNA, regulates hepatic metabolism and functions as a tumor suppressor, yet systematic and direct biochemical elucidation of the miR-122 target network remains incomplete. To this end, we performed Argonaute crosslinking immunoprecipitation (Argonaute [Ago]-CLIP) sequencing in miR-122 knockout and control mouse livers, as well as in matched human hepatocellular carcinoma (HCC) and benign liver tissue to identify miRNA target sites transcriptome-wide in two species. We observed a majority of miR-122 binding on 3′ UTRs and coding exons followed by extensive binding to other genic and non-genic sites. Motif analysis of miR-122-dependent binding revealed a G-bulged motif in addition to canonical motifs. A large number of miR-122 targets were found to be species specific. Upregulation of several common mouse and human targets, most notably BCL9, predicted survival in HCC patients. These results broadly define the molecular consequences of miR-122 downregulation in hepatocellular carcinoma. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Human antibodies in Mexico and Brazil neutralizing tick-borne flaviviruses.
- Author
-
Cervantes Rincón T, Kapoor T, Keeffe JR, Simonelli L, Hoffmann HH, Agudelo M, Jurado A, Peace A, Lee YE, Gazumyan A, Guidetti F, Cantergiani J, Cena B, Bianchini F, Tamagnini E, Moro SG, Svoboda P, Costa F, Reis MG, Ko AI, Fallon BA, Avila-Rios S, Reyes-Téran G, Rice CM, Nussenzweig MC, Bjorkman PJ, Ruzek D, Varani L, MacDonald MR, and Robbiani DF
- Subjects
- Humans, Brazil, Mexico, Antibodies, Viral immunology, Animals, Encephalitis Viruses, Tick-Borne immunology, Flavivirus immunology, Epitopes immunology, Antibodies, Monoclonal immunology, Ticks virology, Ticks immunology, Female, Male, Antibodies, Neutralizing immunology
- Abstract
Flaviviruses such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV) are spread by mosquitoes and cause human disease and mortality in tropical areas. In contrast, Powassan virus (POWV), which causes severe neurologic illness, is a flavivirus transmitted by ticks in temperate regions of the Northern hemisphere. We find serologic neutralizing activity against POWV in individuals living in Mexico and Brazil. Monoclonal antibodies P002 and P003, which were derived from a resident of Mexico (where POWV is not reported), neutralize POWV lineage I by recognizing an epitope on the virus envelope domain III (EDIII) that is shared with a broad range of tick- and mosquito-borne flaviviruses. Our findings raise the possibility that POWV, or a flavivirus closely related to it, infects humans in the tropics., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. The HLA-II immunopeptidome of SARS-CoV-2.
- Author
-
Weingarten-Gabbay S, Chen DY, Sarkizova S, Taylor HB, Gentili M, Hernandez GM, Pearlman LR, Bauer MR, Rice CM, Clauser KR, Hacohen N, Carr SA, Abelin JG, Saeed M, and Sabeti PC
- Subjects
- Humans, Epitopes, T-Lymphocyte, Histocompatibility Antigens Class I, HLA Antigens, Histocompatibility Antigens, CD8-Positive T-Lymphocytes, Peptides, SARS-CoV-2, COVID-19
- Abstract
Targeted synthetic vaccines have the potential to transform our response to viral outbreaks, yet the design of these vaccines requires a comprehensive knowledge of viral immunogens. Here, we report severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) peptides that are naturally processed and loaded onto human leukocyte antigen-II (HLA-II) complexes in infected cells. We identify over 500 unique viral peptides from canonical proteins as well as from overlapping internal open reading frames. Most HLA-II peptides colocalize with known CD4
+ T cell epitopes in coronavirus disease 2019 patients, including 2 reported immunodominant regions in the SARS-CoV-2 membrane protein. Overall, our analyses show that HLA-I and HLA-II pathways target distinct viral proteins, with the structural proteins accounting for most of the HLA-II peptidome and nonstructural and noncanonical proteins accounting for the majority of the HLA-I peptidome. These findings highlight the need for a vaccine design that incorporates multiple viral elements harboring CD4+ and CD8+ T cell epitopes to maximize vaccine effectiveness., Competing Interests: Declaration of interests S.W.-G., D.-Y.C., S.S., K.R.C., N.H., S.A.C., J.G.A., M.S., and P.C.S. are named co-inventors on a patent application related to this work, filed by The Broad Institute, that is being made available in accordance with the COVID-19 technology licensing framework to maximize access to university innovations. N.H. is a founder of Neon Therapeutics (now BioNTech US), was a member of its scientific advisory board, and holds shares. N.H. is also an advisor for IFM Therapeutics. S.A.C. is a member of the scientific advisory boards of Kymera, PTM BioLabs, Seer, and PrognomIQ. J.G.A. is a past employee of Neon Therapeutics (now BioNTech US). P.C.S. is a cofounder of and consultant to Sherlock Biosciences and Delve Biosciences and a board member of Danaher Corporation and holds equity in the companies., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
25. Epigenetic memory of coronavirus infection in innate immune cells and their progenitors.
- Author
-
Cheong JG, Ravishankar A, Sharma S, Parkhurst CN, Grassmann SA, Wingert CK, Laurent P, Ma S, Paddock L, Miranda IC, Karakaslar EO, Nehar-Belaid D, Thibodeau A, Bale MJ, Kartha VK, Yee JK, Mays MY, Jiang C, Daman AW, Martinez de Paz A, Ahimovic D, Ramos V, Lercher A, Nielsen E, Alvarez-Mulett S, Zheng L, Earl A, Yallowitz A, Robbins L, LaFond E, Weidman KL, Racine-Brzostek S, Yang HS, Price DR, Leyre L, Rendeiro AF, Ravichandran H, Kim J, Borczuk AC, Rice CM, Jones RB, Schenck EJ, Kaner RJ, Chadburn A, Zhao Z, Pascual V, Elemento O, Schwartz RE, Buenrostro JD, Niec RE, Barrat FJ, Lief L, Sun JC, Ucar D, and Josefowicz SZ
- Subjects
- Animals, Humans, Mice, Cell Differentiation, Disease Models, Animal, Hematopoietic Stem Cells, Inflammation genetics, Trained Immunity, Monocytes immunology, COVID-19 immunology, Epigenetic Memory, Post-Acute COVID-19 Syndrome genetics, Post-Acute COVID-19 Syndrome immunology, Post-Acute COVID-19 Syndrome pathology
- Abstract
Inflammation can trigger lasting phenotypes in immune and non-immune cells. Whether and how human infections and associated inflammation can form innate immune memory in hematopoietic stem and progenitor cells (HSPC) has remained unclear. We found that circulating HSPC, enriched from peripheral blood, captured the diversity of bone marrow HSPC, enabling investigation of their epigenomic reprogramming following coronavirus disease 2019 (COVID-19). Alterations in innate immune phenotypes and epigenetic programs of HSPC persisted for months to 1 year following severe COVID-19 and were associated with distinct transcription factor (TF) activities, altered regulation of inflammatory programs, and durable increases in myelopoiesis. HSPC epigenomic alterations were conveyed, through differentiation, to progeny innate immune cells. Early activity of IL-6 contributed to these persistent phenotypes in human COVID-19 and a mouse coronavirus infection model. Epigenetic reprogramming of HSPC may underlie altered immune function following infection and be broadly relevant, especially for millions of COVID-19 survivors., Competing Interests: Declaration of interests J.D.B. holds patents related to ATAC-seq and scATAC-seq and serves on the Scientific Advisory Board of CAMP4 Therapeutics, seqWell, and CelSee. S.Z.J. and F.J.B. declare a related patent application: 10203-02-PC; EFS ID: 44924864 Enrichment and Characterization of Rare Circulating Cells, including Progenitor Cells from Peripheral Blood and Uses Thereof. F.J.B. is a co-founder and scientific advisor of IpiNovyx Bio. E.J.S. reports personal fees from NIAID through Axle Informatics for the subject matter expert program for the COVID-19 vaccine clinical trials. R.E.S. is on the scientific advisory board of Miromatrix Inc. and Lime Therapeutics and is a paid consultant and speaker for Alnylam Inc., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Isogenic human trophectoderm cells demonstrate the role of NDUFA4 and associated variants in ZIKV infection.
- Author
-
Yang L, Han Y, Zhou T, Lacko LA, Saeed M, Tan C, Danziger R, Zhu J, Zhao Z, Cahir C, Giani AM, Li Y, Dong X, Moroziewicz D, Paull D, Chen Z, Zhong A, Noggle SA, Rice CM, Qi Q, Evans T, and Chen S
- Abstract
Population-based genome-wide association studies (GWAS) normally require a large sample size, which can be labor intensive and costly. Recently, we reported a human induced pluripotent stem cell (hiPSC) array-based GWAS method, identifying NDUFA4 as a host factor for Zika virus (ZIKV) infection. In this study, we extended our analysis to trophectoderm cells, which constitute one of the major routes of mother-to-fetus transmission of ZIKV during pregnancy. We differentiated hiPSCs from various donors into trophectoderm cells. We then infected cells carrying loss of function mutations in NDUFA4 , harboring risk versus non-risk alleles of SNPs (rs917172 and rs12386620) or having deletions in the NDUFA4 cis -regulatory region with ZIKV. We found that loss/reduction of NDUFA4 suppressed ZIKV infection in trophectoderm cells. This study validated our published hiPSC array-based system as a useful platform for GWAS and confirmed the role of NDUFA4 as a susceptibility locus for ZIKV in disease-relevant trophectoderm cells., Competing Interests: S.C. and T.E. are the co-founders of OncoBeat, LLC. S.C. is a consultant of Vesalius Therapeutics., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
27. Human IRF1 governs macrophagic IFN-γ immunity to mycobacteria.
- Author
-
Rosain J, Neehus AL, Manry J, Yang R, Le Pen J, Daher W, Liu Z, Chan YH, Tahuil N, Türel Ö, Bourgey M, Ogishi M, Doisne JM, Izquierdo HM, Shirasaki T, Le Voyer T, Guérin A, Bastard P, Moncada-Vélez M, Han JE, Khan T, Rapaport F, Hong SH, Cheung A, Haake K, Mindt BC, Pérez L, Philippot Q, Lee D, Zhang P, Rinchai D, Al Ali F, Ahmad Ata MM, Rahman M, Peel JN, Heissel S, Molina H, Kendir-Demirkol Y, Bailey R, Zhao S, Bohlen J, Mancini M, Seeleuthner Y, Roelens M, Lorenzo L, Soudée C, Paz MEJ, González ML, Jeljeli M, Soulier J, Romana S, L'Honneur AS, Materna M, Martínez-Barricarte R, Pochon M, Oleaga-Quintas C, Michev A, Migaud M, Lévy R, Alyanakian MA, Rozenberg F, Croft CA, Vogt G, Emile JF, Kremer L, Ma CS, Fritz JH, Lemon SM, Spaan AN, Manel N, Abel L, MacDonald MR, Boisson-Dupuis S, Marr N, Tangye SG, Di Santo JP, Zhang Q, Zhang SY, Rice CM, Béziat V, Lachmann N, Langlais D, Casanova JL, Gros P, and Bustamante J
- Subjects
- Child, Humans, Interferon-gamma, SARS-CoV-2, Interferon-alpha, Interferon Regulatory Factor-1, COVID-19, Mycobacterium
- Abstract
Inborn errors of human IFN-γ-dependent macrophagic immunity underlie mycobacterial diseases, whereas inborn errors of IFN-α/β-dependent intrinsic immunity underlie viral diseases. Both types of IFNs induce the transcription factor IRF1. We describe unrelated children with inherited complete IRF1 deficiency and early-onset, multiple, life-threatening diseases caused by weakly virulent mycobacteria and related intramacrophagic pathogens. These children have no history of severe viral disease, despite exposure to many viruses, including SARS-CoV-2, which is life-threatening in individuals with impaired IFN-α/β immunity. In leukocytes or fibroblasts stimulated in vitro, IRF1-dependent responses to IFN-γ are, both quantitatively and qualitatively, much stronger than those to IFN-α/β. Moreover, IRF1-deficient mononuclear phagocytes do not control mycobacteria and related pathogens normally when stimulated with IFN-γ. By contrast, IFN-α/β-dependent intrinsic immunity to nine viruses, including SARS-CoV-2, is almost normal in IRF1-deficient fibroblasts. Human IRF1 is essential for IFN-γ-dependent macrophagic immunity to mycobacteria, but largely redundant for IFN-α/β-dependent antiviral immunity., Competing Interests: Declaration of interests J.-L.C. serves on the scientific advisory boards of ADMA Biologics Inc., Kymera Therapeutics, and Elixiron Immunotherapeutics., (Crown Copyright © 2022. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
28. A human iPSC-array-based GWAS identifies a virus susceptibility locus in the NDUFA4 gene and functional variants.
- Author
-
Han Y, Tan L, Zhou T, Yang L, Carrau L, Lacko LA, Saeed M, Zhu J, Zhao Z, Nilsson-Payant BE, Lira Neto FT, Cahir C, Giani AM, Chai JC, Li Y, Dong X, Moroziewicz D, Paull D, Zhang T, Koo S, Tan C, Danziger R, Ba Q, Feng L, Chen Z, Zhong A, Wise GJ, Xiang JZ, Wang H, Schwartz RE, tenOever BR, Noggle SA, Rice CM, Qi Q, Evans T, and Chen S
- Subjects
- Humans, Alleles, DNA, Mitochondrial metabolism, Genetic Predisposition to Disease, Genome-Wide Association Study, Induced Pluripotent Stem Cells metabolism, Interferon Type I metabolism, Polymorphism, Single Nucleotide, SARS-CoV-2, Zika Virus, COVID-19 genetics, Electron Transport Complex IV genetics, Zika Virus Infection genetics, Dengue genetics
- Abstract
Population-based studies to identify disease-associated risk alleles typically require samples from a large number of individuals. Here, we report a human-induced pluripotent stem cell (hiPSC)-based screening strategy to link human genetics with viral infectivity. A genome-wide association study (GWAS) identified a cluster of single-nucleotide polymorphisms (SNPs) in a cis-regulatory region of the NDUFA4 gene, which was associated with susceptibility to Zika virus (ZIKV) infection. Loss of NDUFA4 led to decreased sensitivity to ZIKV, dengue virus, and SARS-CoV-2 infection. Isogenic hiPSC lines carrying non-risk alleles of SNPs or deletion of the cis-regulatory region lower sensitivity to viral infection. Mechanistic studies indicated that loss/reduction of NDUFA4 causes mitochondrial stress, which leads to the leakage of mtDNA and thereby upregulation of type I interferon signaling. This study provides proof-of-principle for the application of iPSC arrays in GWAS and identifies NDUFA4 as a previously unknown susceptibility locus for viral infection., Competing Interests: Declaration of interests R.E.S. is on the scientific advisory board of Miromatrix, Inc and Lime Therapeutics and is a paid consultant and speaker for Alnylam, Inc. S.C. and T.E. are the co-founders of OncoBeat, LLC. S.C. is a consultant of Vesaliustx Therapeutics., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Human hepatocyte PNPLA3-148M exacerbates rapid non-alcoholic fatty liver disease development in chimeric mice.
- Author
-
Kabbani M, Michailidis E, Steensels S, Fulmer CG, Luna JM, Le Pen J, Tardelli M, Razooky B, Ricardo-Lax I, Zou C, Zeck B, Stenzel AF, Quirk C, Foquet L, Ashbrook AW, Schneider WM, Belkaya S, Lalazar G, Liang Y, Pittman M, Devisscher L, Suemizu H, Theise ND, Chiriboga L, Cohen DE, Copenhaver R, Grompe M, Meuleman P, Ersoy BA, Rice CM, and de Jong YP
- Subjects
- Acyltransferases, Animals, Hepatocytes metabolism, Humans, Lipase genetics, Lipase metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Phospholipases A2, Calcium-Independent, Non-alcoholic Fatty Liver Disease genetics
- Abstract
Advanced non-alcoholic fatty liver disease (NAFLD) is a rapidly emerging global health problem associated with pre-disposing genetic polymorphisms, most strikingly an isoleucine to methionine substitution in patatin-like phospholipase domain-containing protein 3 (PNPLA3-I148M). Here, we study how human hepatocytes with PNPLA3 148I and 148M variants engrafted in the livers of broadly immunodeficient chimeric mice respond to hypercaloric diets. As early as four weeks, mice developed dyslipidemia, impaired glucose tolerance, and steatosis with ballooning degeneration selectively in the human graft, followed by pericellular fibrosis after eight weeks of hypercaloric feeding. Hepatocytes with the PNPLA3-148M variant, either from a homozygous 148M donor or overexpressed in a 148I donor background, developed microvesicular and severe steatosis with frequent ballooning degeneration, resulting in more active steatohepatitis than 148I hepatocytes. We conclude that PNPLA3-148M in human hepatocytes exacerbates NAFLD. These models will facilitate mechanistic studies into human genetic variant contributions to advanced fatty liver diseases., Competing Interests: Declaration of interests L.F., R.C., and M.G. have financial interest in Yecuris Corporation. All others declare no competing interests., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
30. Analysis of memory B cells identifies conserved neutralizing epitopes on the N-terminal domain of variant SARS-Cov-2 spike proteins.
- Author
-
Wang Z, Muecksch F, Cho A, Gaebler C, Hoffmann HH, Ramos V, Zong S, Cipolla M, Johnson B, Schmidt F, DaSilva J, Bednarski E, Ben Tanfous T, Raspe R, Yao K, Lee YE, Chen T, Turroja M, Milard KG, Dizon J, Kaczynska A, Gazumyan A, Oliveira TY, Rice CM, Caskey M, Bieniasz PD, Hatziioannou T, Barnes CO, and Nussenzweig MC
- Subjects
- Antibodies, Monoclonal, Antibodies, Neutralizing, Antibodies, Viral, Epitopes, Humans, Memory B Cells, SARS-CoV-2, COVID-19, Spike Glycoprotein, Coronavirus
- Abstract
SARS-CoV-2 infection or vaccination produces neutralizing antibody responses that contribute to better clinical outcomes. The receptor-binding domain (RBD) and the N-terminal domain (NTD) of the spike trimer (S) constitute the two major neutralizing targets for antibodies. Here, we use NTD-specific probes to capture anti-NTD memory B cells in a longitudinal cohort of infected individuals, some of whom were vaccinated. We found 6 complementation groups of neutralizing antibodies. 58% targeted epitopes outside the NTD supersite, 58% neutralized either Gamma or Omicron, and 14% were broad neutralizers that also neutralized Omicron. Structural characterization revealed that broadly active antibodies targeted three epitopes outside the NTD supersite including a class that recognized both the NTD and SD2 domain. Rapid recruitment of memory B cells producing these antibodies into the plasma cell compartment upon re-infection likely contributes to the relatively benign course of subsequent infections with SARS-CoV-2 variants, including Omicron., Competing Interests: Declaration of interests The Rockefeller University has filed a provisional patent application in connection with this work on which M.C.N. and Z.W. are inventors (US patent 17/575,246)., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. System-wide transcriptome damage and tissue identity loss in COVID-19 patients.
- Author
-
Park J, Foox J, Hether T, Danko DC, Warren S, Kim Y, Reeves J, Butler DJ, Mozsary C, Rosiene J, Shaiber A, Afshin EE, MacKay M, Rendeiro AF, Bram Y, Chandar V, Geiger H, Craney A, Velu P, Melnick AM, Hajirasouliha I, Beheshti A, Taylor D, Saravia-Butler A, Singh U, Wurtele ES, Schisler J, Fennessey S, Corvelo A, Zody MC, Germer S, Salvatore S, Levy S, Wu S, Tatonetti NP, Shapira S, Salvatore M, Westblade LF, Cushing M, Rennert H, Kriegel AJ, Elemento O, Imielinski M, Rice CM, Borczuk AC, Meydan C, Schwartz RE, and Mason CE
- Subjects
- Adult, Aged, Aged, 80 and over, COVID-19 metabolism, COVID-19 virology, Case-Control Studies, Cohort Studies, Female, Gene Expression Regulation, Humans, Influenza, Human genetics, Influenza, Human pathology, Influenza, Human virology, Lung metabolism, Male, Middle Aged, Orthomyxoviridae, RNA-Seq methods, Respiratory Distress Syndrome genetics, Respiratory Distress Syndrome microbiology, Respiratory Distress Syndrome pathology, Viral Load, COVID-19 genetics, COVID-19 pathology, Lung pathology, SARS-CoV-2, Transcriptome genetics
- Abstract
The molecular mechanisms underlying the clinical manifestations of coronavirus disease 2019 (COVID-19), and what distinguishes them from common seasonal influenza virus and other lung injury states such as acute respiratory distress syndrome, remain poorly understood. To address these challenges, we combine transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues to define body-wide transcriptome changes in response to COVID-19. We then match these data with spatial protein and expression profiling across 357 tissue sections from 16 representative patient lung samples and identify tissue-compartment-specific damage wrought by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, evident as a function of varying viral loads during the clinical course of infection and tissue-type-specific expression states. Overall, our findings reveal a systemic disruption of canonical cellular and transcriptional pathways across all tissues, which can inform subsequent studies to combat the mortality of COVID-19 and to better understand the molecular dynamics of lethal SARS-CoV-2 and other respiratory infections., Competing Interests: O.E. is scientific adviser and equity holder in Freenome, Owkin, Volastra Therapeutics, and OneThree Biotech. R.E.S. is on the scientific advisory board of Miromatrix, Inc., and is a consultant and speaker for Alnylam, Inc. L.S. is a scientific co-founder and paid consultant. C.M. and E.E.A. are consultants for Onegevity Health. C.E.M. is a co-founder of Biotia and Onegevity Health and an advisor to Nanostring. T.H., S.W., Y.K., and J.R. are employees of Nanostring, Inc. All other authors declare no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
32. Profiling SARS-CoV-2 HLA-I peptidome reveals T cell epitopes from out-of-frame ORFs.
- Author
-
Weingarten-Gabbay S, Klaeger S, Sarkizova S, Pearlman LR, Chen DY, Gallagher KME, Bauer MR, Taylor HB, Dunn WA, Tarr C, Sidney J, Rachimi S, Conway HL, Katsis K, Wang Y, Leistritz-Edwards D, Durkin MR, Tomkins-Tinch CH, Finkel Y, Nachshon A, Gentili M, Rivera KD, Carulli IP, Chea VA, Chandrashekar A, Bozkus CC, Carrington M, Bhardwaj N, Barouch DH, Sette A, Maus MV, Rice CM, Clauser KR, Keskin DB, Pregibon DC, Hacohen N, Carr SA, Abelin JG, Saeed M, and Sabeti PC
- Subjects
- A549 Cells, Alleles, Amino Acid Sequence, Animals, Antigen Presentation immunology, COVID-19 immunology, COVID-19 virology, Female, HEK293 Cells, Humans, Kinetics, Male, Mice, Peptides chemistry, T-Lymphocytes immunology, Epitopes, T-Lymphocyte immunology, Histocompatibility Antigens Class I immunology, Open Reading Frames genetics, Peptides immunology, Proteome immunology, SARS-CoV-2 immunology
- Abstract
T cell-mediated immunity plays an important role in controlling SARS-CoV-2 infection, but the repertoire of naturally processed and presented viral epitopes on class I human leukocyte antigen (HLA-I) remains uncharacterized. Here, we report the first HLA-I immunopeptidome of SARS-CoV-2 in two cell lines at different times post infection using mass spectrometry. We found HLA-I peptides derived not only from canonical open reading frames (ORFs) but also from internal out-of-frame ORFs in spike and nucleocapsid not captured by current vaccines. Some peptides from out-of-frame ORFs elicited T cell responses in a humanized mouse model and individuals with COVID-19 that exceeded responses to canonical peptides, including some of the strongest epitopes reported to date. Whole-proteome analysis of infected cells revealed that early expressed viral proteins contribute more to HLA-I presentation and immunogenicity. These biological insights, as well as the discovery of out-of-frame ORF epitopes, will facilitate selection of peptides for immune monitoring and vaccine development., Competing Interests: Declaration of interests S.W.-G., S.K., S.S., K.R.C, N.H., S.A.C., J.G.A., M.S., and P.C.S. are named co-inventors on a patent application related to immunogenic compositions of this manuscript filed by The Broad Institute that is being made available in accordance with the COVID-19 technology licensing framework to maximize access to university innovations. D.L.-E., C.T., Y.W., M.R.D., W.A.D., and D.C.P. are employees and stockholders of Repertoire Immune Medicines. N.B. is an extramural member of the Parker Institute for Cancer Immunotherapy; receives research funds from Regeneron, Harbor Biomedical, DC Prime, and Dragonfly Therapeutics; and is on the advisory boards of Neon Therapeutics, Novartis, Avidea, Boehringer Ingelheim, Rome Therapeutics, Roswell Park Comprehensive Cancer Center, BreakBio, Carisma Therapeutics, CureVac, Genotwin, BioNTech, Gilead Therapeutics, Tempest Therapeutics, and the Cancer Research Institute. A.S. is a consultant for Gritstone, Flow Pharma, CellCarta, OxfordImmunotech, Immunoscape, and Avalia. La Jolla Institute for Immunology has filed for patent protection for various aspects of T cell epitope and vaccine design work. D.B.K. has previously advised Neon Therapeutics and has received consulting fees from Neon Therapeutics. D.B.K. owns equity in AduroBiotech, Agenus Inc., Armata Pharmaceuticals, Breakbio Corp., Biomarin Pharmaceutical Inc., Bristol Myers Squibb Com., Celldex Therapeutics Inc., Editas Medicine Inc., Exelixis Inc., Gilead Sciences Inc., IMV Inc., Lexicon Pharmaceuticals Inc., Moderna Inc., and Regeneron Pharmaceuticals. D.B.K. receives SARS-CoV-2 research support from BeiGene for a project unrelated to this publication. N.H. is a founder of Neon Therapeutics, Inc. (now BioNTech US), was a member of its scientific advisory board, and holds shares. N.H. is also an advisor for IFM Therapeutics. S.A.C. is a member of the scientific advisory boards of Kymera, PTM BioLabs, and Seer and a scientific advisor to Pfizer and Biogen. J.G.A. is a past employee and shareholder of Neon Therapeutics, Inc. (now BioNTech US). P.C.S. is a co-founder and shareholder of Sherlock Biosciences and a non-executive board member and shareholder of Danaher Corporation., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
33. Quantitative measurements of early alphaviral replication dynamics in single cells reveals the basis for superinfection exclusion.
- Author
-
Singer ZS, Ambrose PM, Danino T, and Rice CM
- Subjects
- Evaluation Studies as Topic, Humans, Alphavirus genetics, Single-Cell Analysis methods, Superinfection virology
- Abstract
While decades of research have elucidated many steps of the alphavirus lifecycle, the earliest replication dynamics have remained unclear. This missing time window has obscured early replicase strand-synthesis behavior and prevented elucidation of how the first events of infection might influence subsequent viral competition. Using quantitative live-cell and single-molecule imaging, we observed the initial replicase activity and its strand preferences in situ and measured the trajectory of replication over time. Under this quantitative framework, we investigated viral competition, where one alphavirus is able to exclude superinfection by a second homologous virus. We show that this appears as an indirect phenotypic consequence of a bidirectional competition between the two species, coupled with the rapid onset of viral replication and a limited total cellular carrying capacity. Together, these results emphasize the utility of analyzing viral kinetics within single cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. A Combination of Human Broadly Neutralizing Antibodies against Hepatitis B Virus HBsAg with Distinct Epitopes Suppresses Escape Mutations.
- Author
-
Wang Q, Michailidis E, Yu Y, Wang Z, Hurley AM, Oren DA, Mayer CT, Gazumyan A, Liu Z, Zhou Y, Schoofs T, Yao KH, Nieke JP, Wu J, Jiang Q, Zou C, Kabbani M, Quirk C, Oliveira T, Chhosphel K, Zhang Q, Schneider WM, Jahan C, Ying T, Horowitz J, Caskey M, Jankovic M, Robbiani DF, Wen Y, de Jong YP, Rice CM, and Nussenzweig MC
- Subjects
- Animals, Antibodies, Monoclonal immunology, Cell Line, Tumor, Child, Preschool, Disease Models, Animal, Epitopes immunology, Female, HEK293 Cells, Hep G2 Cells, Hepatitis B, Chronic drug therapy, Hepatitis B, Chronic immunology, Humans, Infant, Mice, Mice, Knockout, Protein Conformation, Broadly Neutralizing Antibodies immunology, Hepatitis B Antibodies immunology, Hepatitis B Surface Antigens immunology, Hepatitis B virus immunology
- Abstract
Although there is no effective cure for chronic hepatitis B virus (HBV) infection, antibodies are protective and correlate with recovery from infection. To examine the human antibody response to HBV, we screened 124 vaccinated and 20 infected, spontaneously recovered individuals. The selected individuals produced shared clones of broadly neutralizing antibodies (bNAbs) that targeted 3 non-overlapping epitopes on the HBV S antigen (HBsAg). Single bNAbs protected humanized mice against infection but selected for resistance mutations in mice with prior established infection. In contrast, infection was controlled by a combination of bNAbs targeting non-overlapping epitopes with complementary sensitivity to mutations that commonly emerge during human infection. The co-crystal structure of one of the bNAbs with an HBsAg peptide epitope revealed a stabilized hairpin loop. This structure, which contains residues frequently mutated in clinical immune escape variants, provides a molecular explanation for why immunotherapy for HBV infection may require combinations of complementary bNAbs., Competing Interests: Declaration of Interests Q.W. and M.C.N. have a provisional patent application with the U.S. Patent and Trademark Office (62898735). Other authors have no conflicts of interest to declare., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. Long-Term Expansion of Functional Mouse and Human Hepatocytes as 3D Organoids.
- Author
-
Hu H, Gehart H, Artegiani B, LÖpez-Iglesias C, Dekkers F, Basak O, van Es J, Chuva de Sousa Lopes SM, Begthel H, Korving J, van den Born M, Zou C, Quirk C, Chiriboga L, Rice CM, Ma S, Rios A, Peters PJ, de Jong YP, and Clevers H
- Subjects
- Animals, Cell Culture Techniques, Cells, Cultured, Hepatocytes cytology, Humans, Mice, Mice, Inbred BALB C, Mice, Knockout, Organoids cytology, Stem Cells cytology, Stem Cells metabolism, Time Factors, Cell Proliferation, Hepatocytes metabolism, Organoids metabolism
- Abstract
The mammalian liver possesses a remarkable regenerative ability. Two modes of damage response have been described: (1) The "oval cell" response emanates from the biliary tree when all hepatocytes are affected by chronic liver disease. (2) A massive, proliferative response of mature hepatocytes occurs upon acute liver damage such as partial hepatectomy (PHx). While the oval cell response has been captured in vitro by growing organoids from cholangiocytes, the hepatocyte proliferative response has not been recapitulated in culture. Here, we describe the establishment of a long-term 3D organoid culture system for mouse and human primary hepatocytes. Organoids can be established from single hepatocytes and grown for multiple months, while retaining key morphological, functional and gene expression features. Transcriptional profiles of the organoids resemble those of proliferating hepatocytes after PHx. Human hepatocyte organoids proliferate extensively after engraftment into mice and thus recapitulate the proliferative damage-response of hepatocytes., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
36. A Combination of Two Human Monoclonal Antibodies Prevents Zika Virus Escape Mutations in Non-human Primates.
- Author
-
Keeffe JR, Van Rompay KKA, Olsen PC, Wang Q, Gazumyan A, Azzopardi SA, Schaefer-Babajew D, Lee YE, Stuart JB, Singapuri A, Watanabe J, Usachenko J, Ardeshir A, Saeed M, Agudelo M, Eisenreich T, Bournazos S, Oliveira TY, Rice CM, Coffey LL, MacDonald MR, Bjorkman PJ, Nussenzweig MC, and Robbiani DF
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal chemistry, Antibodies, Neutralizing immunology, Dengue Virus immunology, Epitopes immunology, HEK293 Cells, Humans, Macaca, Mice, Knockout, Protein Domains, Antibodies, Monoclonal immunology, Mutation genetics, Zika Virus immunology
- Abstract
Zika virus (ZIKV) causes severe neurologic complications and fetal aberrations. Vaccine development is hindered by potential safety concerns due to antibody cross-reactivity with dengue virus and the possibility of disease enhancement. In contrast, passive administration of anti-ZIKV antibodies engineered to prevent enhancement may be safe and effective. Here, we report on human monoclonal antibody Z021, a potent neutralizer that recognizes an epitope on the lateral ridge of the envelope domain III (EDIII) of ZIKV and is protective against ZIKV in mice. When administered to macaques undergoing a high-dose ZIKV challenge, a single anti-EDIII antibody selected for resistant variants. Co-administration of two antibodies, Z004 and Z021, which target distinct sites on EDIII, was associated with a delay and a 3- to 4-log decrease in peak viremia. Moreover, the combination of these antibodies engineered to avoid enhancement prevented viral escape due to mutation in macaques, a natural host for ZIKV., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
37. NS5A Promotes Constitutive Degradation of IP3R3 to Counteract Apoptosis Induced by Hepatitis C Virus.
- Author
-
Kuchay S, Saeed M, Giorgi C, Li J, Hoffmann HH, Pinton P, Rice CM, and Pagano M
- Subjects
- Animals, Carbamates, Cell Line, F-Box Proteins metabolism, Hepacivirus drug effects, Hepatitis C metabolism, Hepatitis C pathology, Hepatitis C virology, Humans, Imidazoles pharmacology, Leucine analogs & derivatives, Leucine pharmacology, Protein Multimerization drug effects, Pyrrolidines, Valine analogs & derivatives, Virus Replication drug effects, Apoptosis drug effects, Hepacivirus physiology, Inositol 1,4,5-Trisphosphate Receptors metabolism, Proteolysis drug effects, Viral Nonstructural Proteins metabolism
- Abstract
FBXL2 targets IP3R3 for ubiquitin-mediated degradation to limit Ca
2+ flux to mitochondria and, consequently, apoptosis. Efficient replication of hepatitis C virus (HCV) requires geranylgeranylation of FBXL2. Here, we show that the viral protein NS5A forms a trimeric complex with IP3R3 and FBXL2, unmasking IP3R3's degron in the absence of inositol 1,4,5-trisphosphate (IP3 ) stimulation. FBXL2 knockdown or expression of a stable IP3R3 mutant causes persistent Ca2+ flux and sensitizes cells to apoptosis, resulting in the inhibition of viral replication. Importantly, the effect of FBXL2 silencing is rescued by depleting IP3R3, but not p85β, another established FBXL2 substrate, indicating that the anti-HCV effect of FBXL2 knockdown is largely due to IP3R3 stabilization. Finally, disruption of the FBXL2-NS5A-IP3R3 complex using somatic cell genetics or pharmacologic inhibition results in IP3R3 stabilization and suppression of HCV replication. This study reveals an IP3 -independent molecular mechanism through which HCV promotes IP3R3 degradation, thereby inhibiting virus-induced apoptosis and establishing chronic infection., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
38. Differential Regulation of Lipoprotein and Hepatitis C Virus Secretion by Rab1b.
- Author
-
Takacs CN, Andreo U, Dao Thi VL, Wu X, Gleason CE, Itano MS, Spitz-Becker GS, Belote RL, Hedin BR, Scull MA, Rice CM, and Simon SM
- Subjects
- Cell Line, Tumor, Exocytosis, HEK293 Cells, Hepacivirus metabolism, Humans, Mutation, rab1 GTP-Binding Proteins genetics, Apolipoproteins metabolism, Secretory Pathway, rab1 GTP-Binding Proteins metabolism
- Abstract
Secretory cells produce diverse cargoes, yet how they regulate concomitant secretory traffic remains insufficiently explored. Rab GTPases control intracellular vesicular transport. To map secretion pathways, we generated a library of lentivirus-expressed dominant-negative Rab mutants and used it in a large-scale screen to identify regulators of hepatic lipoprotein secretion. We identified several candidate pathways, including those mediated by Rab11 and Rab8. Surprisingly, inhibition of Rab1b, the major regulator of transport from the endoplasmic reticulum to the Golgi, differently affected the secretion of the very-low-density lipoprotein components ApoE and ApoB100, despite their final association on mature secreted lipoprotein particles. Since hepatitis C virus (HCV) incorporates ApoE and ApoB100 into its virus particle, we also investigated infectious HCV secretion and show that its regulation by Rab1b mirrors that of ApoB100. These observations reveal differential regulation of hepatocyte secretion by Rab1b and advance our understanding of lipoprotein assembly and lipoprotein and HCV secretion., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
39. Recurrent Potent Human Neutralizing Antibodies to Zika Virus in Brazil and Mexico.
- Author
-
Robbiani DF, Bozzacco L, Keeffe JR, Khouri R, Olsen PC, Gazumyan A, Schaefer-Babajew D, Avila-Rios S, Nogueira L, Patel R, Azzopardi SA, Uhl LFK, Saeed M, Sevilla-Reyes EE, Agudelo M, Yao KH, Golijanin J, Gristick HB, Lee YE, Hurley A, Caskey M, Pai J, Oliveira T, Wunder EA Jr, Sacramento G, Nery N Jr, Orge C, Costa F, Reis MG, Thomas NM, Eisenreich T, Weinberger DM, de Almeida ARP, West AP Jr, Rice CM, Bjorkman PJ, Reyes-Teran G, Ko AI, MacDonald MR, and Nussenzweig MC
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, Antibodies, Viral blood, Antibodies, Viral immunology, B-Lymphocytes immunology, Brazil, Female, Humans, Immunologic Memory, Leukocytes, Mononuclear immunology, Male, Mexico, Mice, Zika Virus Infection blood, Antibodies, Neutralizing chemistry, Antibodies, Viral chemistry, Zika Virus Infection immunology
- Abstract
Antibodies to Zika virus (ZIKV) can be protective. To examine the antibody response in individuals who develop high titers of anti-ZIKV antibodies, we screened cohorts in Brazil and Mexico for ZIKV envelope domain III (ZEDIII) binding and neutralization. We find that serologic reactivity to dengue 1 virus (DENV1) EDIII before ZIKV exposure is associated with increased ZIKV neutralizing titers after exposure. Antibody cloning shows that donors with high ZIKV neutralizing antibody titers have expanded clones of memory B cells that express the same immunoglobulin VH3-23/VK1-5 genes. These recurring antibodies cross-react with DENV1, but not other flaviviruses, neutralize both DENV1 and ZIKV, and protect mice against ZIKV challenge. Structural analyses reveal the mechanism of recognition of the ZEDIII lateral ridge by VH3-23/VK1-5 antibodies. Serologic testing shows that antibodies to this region correlate with serum neutralizing activity to ZIKV. Thus, high neutralizing responses to ZIKV are associated with pre-existing reactivity to DENV1 in humans., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. Superior In vivo Transduction of Human Hepatocytes Using Engineered AAV3 Capsid.
- Author
-
Vercauteren K, Hoffman BE, Zolotukhin I, Keeler GD, Xiao JW, Basner-Tschakarjan E, High KA, Ertl HC, Rice CM, Srivastava A, de Jong YP, and Herzog RW
- Subjects
- Animals, Cells, Cultured, Dependovirus metabolism, Hepatocytes metabolism, Humans, Mice, Organ Specificity, Protein Engineering, Transduction, Genetic, Capsid Proteins genetics, Dependovirus genetics, Genetic Vectors administration & dosage, Hepatocytes ultrastructure
- Abstract
Adeno-associated viral (AAV) vectors are currently being tested in multiple clinical trials for liver-directed gene transfer to treat the bleeding disorders hemophilia A and B and metabolic disorders. The optimal viral capsid for transduction of human hepatocytes has been under active investigation, but results across various models are inconsistent. We tested in vivo transduction in "humanized" mice. Methods to quantitate percent AAV transduced human and murine hepatocytes in chimeric livers were optimized using flow cytometry and confocal microscopy with image analysis. Distinct transduction efficiencies were noted following peripheral vein administration of a self-complementary vector expressing a gfp reporter gene. An engineered AAV3 capsid with two amino acid changes, S663V+T492V (AAV3-ST), showed best efficiency for human hepatocytes (~3-times, ~8-times, and ~80-times higher than for AAV9, AAV8, and AAV5, respectively). AAV5, 8, and 9 were more efficient in transducing murine than human hepatocytes. AAV8 yielded the highest transduction rate of murine hepatocytes, which was 19-times higher than that for human hepatocytes. In summary, our data show substantial differences among AAV serotypes in transduction of human and mouse hepatocytes, are the first to report on AAV5 in humanized mice, and support the use of AAV3-based vectors for human liver gene transfer.
- Published
- 2016
- Full Text
- View/download PDF
41. Quantitative Proteomics Identifies Serum Response Factor Binding Protein 1 as a Host Factor for Hepatitis C Virus Entry.
- Author
-
Gerold G, Meissner F, Bruening J, Welsch K, Perin PM, Baumert TF, Vondran FW, Kaderali L, Marcotrigiano J, Khan AG, Mann M, Rice CM, and Pietschmann T
- Subjects
- Cell Line, Tumor, Humans, Hepacivirus physiology, Proteomics, Transcription Factors metabolism, Virus Internalization
- Abstract
Hepatitis C virus (HCV) enters human hepatocytes through a multistep mechanism involving, among other host proteins, the virus receptor CD81. How CD81 governs HCV entry is poorly characterized, and CD81 protein interactions after virus binding remain elusive. We have developed a quantitative proteomics protocol to identify HCV-triggered CD81 interactions and found 26 dynamic binding partners. At least six of these proteins promote HCV infection, as indicated by RNAi. We further characterized serum response factor binding protein 1 (SRFBP1), which is recruited to CD81 during HCV uptake and supports HCV infection in hepatoma cells and primary human hepatocytes. SRFBP1 facilitates host cell penetration by all seven HCV genotypes, but not of vesicular stomatitis virus and human coronavirus. Thus, SRFBP1 is an HCV-specific, pan-genotypic host entry factor. These results demonstrate the use of quantitative proteomics to elucidate pathogen entry and underscore the importance of host protein-protein interactions during HCV invasion., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. ATP-dependent effector-like functions of RIG-I-like receptors.
- Author
-
Yao H, Dittmann M, Peisley A, Hoffmann HH, Gilmore RH, Schmidt T, Schmidt-Burgk J, Hornung V, Rice CM, and Hur S
- Subjects
- Antiviral Agents metabolism, Base Sequence, Blotting, Western, Cell Line, Tumor, DEAD Box Protein 58, DEAD-box RNA Helicases genetics, HEK293 Cells, Humans, Interferon-Induced Helicase, IFIH1, Interferon-beta genetics, Interferon-beta metabolism, Models, Molecular, Mutation, Nucleic Acid Conformation, Phosphorylation, RNA Interference, RNA, Double-Stranded chemistry, RNA, Double-Stranded genetics, RNA, Double-Stranded metabolism, RNA, Viral chemistry, RNA, Viral genetics, RNA, Viral metabolism, Receptors, Immunologic, Receptors, Pattern Recognition genetics, Reverse Transcriptase Polymerase Chain Reaction, Viral Proteins genetics, Viral Proteins metabolism, Virus Diseases genetics, Virus Diseases metabolism, eIF-2 Kinase genetics, eIF-2 Kinase metabolism, Adenosine Triphosphate metabolism, DEAD-box RNA Helicases metabolism, Receptors, Pattern Recognition metabolism
- Abstract
The vertebrate antiviral innate immune system is often considered to consist of two distinct groups of proteins: pattern recognition receptors (PRRs) that detect viral infection and induce the interferon (IFN) signaling, and effectors that directly act against viral replication. Accordingly, previous studies on PRRs, such as RIG-I and MDA5, have primarily focused on their functions in viral double-stranded RNA (dsRNA) detection and consequent antiviral signaling. We report here that both RIG-I and MDA5 efficiently displace viral proteins pre-bound to dsRNA in a manner dependent on their ATP hydrolysis, and that this activity assists a dsRNA-dependent antiviral effector protein, PKR, and allows RIG-I to promote MDA5 signaling. Furthermore, truncated RIG-I/MDA5 lacking the signaling domain, and hence the IFN stimulatory activity, displaces viral proteins and suppresses replication of certain viruses in an ATP-dependent manner. Thus, this study reveals novel "effector-like" functions of RIG-I and MDA5 that challenge the conventional view of PRRs., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
43. The RNA sensor RIG-I dually functions as an innate sensor and direct antiviral factor for hepatitis B virus.
- Author
-
Sato S, Li K, Kameyama T, Hayashi T, Ishida Y, Murakami S, Watanabe T, Iijima S, Sakurai Y, Watashi K, Tsutsumi S, Sato Y, Akita H, Wakita T, Rice CM, Harashima H, Kohara M, Tanaka Y, and Takaoka A
- Subjects
- Animals, Child, Preschool, Female, Hep G2 Cells, Hepatocytes transplantation, Hepatocytes virology, Humans, Immunity, Innate, Interferons metabolism, Liver virology, Membrane Proteins immunology, Mice, Mice, SCID, Nerve Tissue Proteins immunology, RNA, Viral genetics, Receptors, Cell Surface, Transgenes genetics, Transplantation Chimera, Virus Replication genetics, Gene Products, pol antagonists & inhibitors, Hepatitis B virus physiology, Hepatitis B, Chronic immunology, Hepatocytes physiology, Liver physiology, Membrane Proteins metabolism, Nerve Tissue Proteins metabolism, RNA, Viral immunology
- Abstract
Host innate recognition triggers key immune responses for viral elimination. The sensing mechanism of hepatitis B virus (HBV), a DNA virus, and the subsequent downstream signaling events remain to be fully clarified. Here we found that type III but not type I interferons are predominantly induced in human primary hepatocytes in response to HBV infection, through retinoic acid-inducible gene-I (RIG-I)-mediated sensing of the 5'-ε region of HBV pregenomic RNA. In addition, RIG-I could also counteract the interaction of HBV polymerase (P protein) with the 5'-ε region in an RNA-binding dependent manner, which consistently suppressed viral replication. Liposome-mediated delivery and vector-based expression of this ε region-derived RNA in liver abolished the HBV replication in human hepatocyte-chimeric mice. These findings identify an innate-recognition mechanism by which RIG-I dually functions as an HBV sensor activating innate signaling and to counteract viral polymerase in human hepatocytes., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
44. Humanized mice for modeling human infectious disease: challenges, progress, and outlook.
- Author
-
Legrand N, Ploss A, Balling R, Becker PD, Borsotti C, Brezillon N, Debarry J, de Jong Y, Deng H, Di Santo JP, Eisenbarth S, Eynon E, Flavell RA, Guzman CA, Huntington ND, Kremsdorf D, Manns MP, Manz MG, Mention JJ, Ott M, Rathinam C, Rice CM, Rongvaux A, Stevens S, Spits H, Strick-Marchand H, Takizawa H, van Lent AU, Wang C, Weijer K, Willinger T, and Ziegler P
- Subjects
- Animals, Humans, Mice, Biomedical Research trends, Communicable Diseases, Disease Models, Animal
- Abstract
Over 800 million people worldwide are infected with hepatitis viruses, human immunodeficiency virus (HIV), and malaria, resulting in more than 5 million deaths annually. Here we discuss the potential and challenges of humanized mouse models for developing effective and affordable therapies and vaccines, which are desperately needed to combat these diseases.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.