151
|
Phosphoproteomic-based kinase profiling early in influenza virus infection identifies GRK2 as antiviral drug target. Nat Commun 2018; 9:3679. [PMID: 30206219 PMCID: PMC6133941 DOI: 10.1038/s41467-018-06119-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 08/16/2018] [Indexed: 01/23/2023] Open
Abstract
Although annual influenza epidemics affect around 10% of the global population, current treatment options are limited and development of new antivirals is needed. Here, using quantitative phosphoproteomics, we reveal the unique phosphoproteome dynamics that occur in the host cell within minutes of influenza A virus (IAV) infection. We uncover cellular kinases required for the observed signaling pattern and find that inhibition of selected candidates, such as the G protein-coupled receptor kinase 2 (GRK2), leads to decreased IAV replication. As GRK2 has emerged as drug target in heart disease, we focus on its role in IAV infection and show that it is required for viral uncoating. Replication of seasonal and pandemic IAVs is severely decreased by specific GRK2 inhibitors in primary human airway cultures and in mice. Our study reveals the IAV-induced changes to the cellular phosphoproteome and identifies GRK2 as crucial node of the kinase network that enables IAV replication. Influenza A virus (IAV) causes annual epidemics and development of antivirals is needed. Here, the authors perform phosphoproteomics during IAV entry and identify GRK2 as drug target, inhibition of which decreases replication of seasonal and pandemic IAV in primary human cells and animal models.
Collapse
|
152
|
Hepatitis C virus enters liver cells using the CD81 receptor complex proteins calpain-5 and CBLB. PLoS Pathog 2018; 14:e1007111. [PMID: 30024968 PMCID: PMC6053247 DOI: 10.1371/journal.ppat.1007111] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/18/2018] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) and the malaria parasite Plasmodium use the membrane protein CD81 to invade human liver cells. Here we mapped 33 host protein interactions of CD81 in primary human liver and hepatoma cells using high-resolution quantitative proteomics. In the CD81 protein network, we identified five proteins which are HCV entry factors or facilitators including epidermal growth factor receptor (EGFR). Notably, we discovered calpain-5 (CAPN5) and the ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene B (CBLB) to form a complex with CD81 and support HCV entry. CAPN5 and CBLB were required for a post-binding and pre-replication step in the HCV life cycle. Knockout of CAPN5 and CBLB reduced susceptibility to all tested HCV genotypes, but not to other enveloped viruses such as vesicular stomatitis virus and human coronavirus. Furthermore, Plasmodium sporozoites relied on a distinct set of CD81 interaction partners for liver cell entry. Our findings reveal a comprehensive CD81 network in human liver cells and show that HCV and Plasmodium highjack selective CD81 interactions, including CAPN5 and CBLB for HCV, to invade cells. CD81 is a cell membrane protein, which functions as entry factor for hepatitis C virus (HCV) and malaria sporozoites in the human liver. Currently, it remains enigmatic how CD81 guides the entry process of both pathogens and whether it functions in a similar way during liver cell invasion of HCV and malaria parasites. Here, we use high resolution quantitative proteomics to identify CD81 associated host proteins in liver cells. We found that at least 33 proteins form a complex with CD81, 23 of which were not reported as interaction partners before. We further determined that at least five CD81 interactors are HCV host factors, among them calpain-5 (CAPN5) and the ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene B (CBLB). All tested HCV genotypes require CAPN5 and CBLB for full infection, but neither malaria parasites nor other tested enveloped virus rely on CAPN5 or CBLB. Our study maps the liver cell interactome of CD81 and provides new insight into the distinct cell invasion mechanisms of HCV and malaria parasites.
Collapse
|
153
|
Hu W, Zhang S, Shen Y, Yang Q. Epidermal growth factor receptor is a co-factor for transmissible gastroenteritis virus entry. Virology 2018; 521:33-43. [PMID: 29879540 PMCID: PMC7112045 DOI: 10.1016/j.virol.2018.05.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 01/06/2023]
Abstract
Transmissible gastroenteritis virus (TGEV) causes severe diarrhea and high mortality in newborn piglets. It is well established that porcine intestinal epithelium is the target of the TGEV infection, however the mechanism that TGEV invades the host epithelium remains largely unknown. Aminopeptidase N (APN) is a known receptor of TGEV. This study discovered that the extracellular receptor binding domain 1 pertaining to epidermal growth receptor (EGFR) interact with TGEV spike protein. APN and EGFR synergistically promote TGEV invasion. TGEV promotes APN and EGFR clustering early in infection. Furthermore APN and EGFR synergistically stimulate PI3K/AKT as well as MEK/ERK1/2 endocytosis signaling pathways. TGEV entry is via clathrin and caveolin mediated endocytosis in IPEC-J2 cells. TGEV binds with EGFR, and subsequently promotes EGFR internalization by a clathrin-mediated endocytosis pathway. These results show that EGFR is a co-factor of TGEV, and that it plays a synergistic role with APN early in TGEV infection.
Collapse
Affiliation(s)
- Weiwei Hu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Shuai Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Yumeng Shen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu 210095, PR China.
| |
Collapse
|
154
|
Shin JS, Jung E, Kim M, Baric RS, Go YY. Saracatinib Inhibits Middle East Respiratory Syndrome-Coronavirus Replication In Vitro. Viruses 2018; 10:v10060283. [PMID: 29795047 PMCID: PMC6024778 DOI: 10.3390/v10060283] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 11/16/2022] Open
Abstract
The Middle East respiratory syndrome-coronavirus (MERS-CoV), first identified in Saudi Arabia, is an emerging zoonotic pathogen that causes severe acute respiratory illness in humans with a high fatality rate. Since its emergence, MERS-CoV continues to spread to countries outside of the Arabian Peninsula and gives rise to sporadic human infections following the entry of infected individuals to other countries, which can precipitate outbreaks similar to the one that occurred in South Korea in 2015. Current therapeutics against MERS-CoV infection have primarily been adapted from previous drugs used for the treatment of severe acute respiratory syndrome. In search of new potential drug candidates, we screened a library composed of 2334 clinically approved drugs and pharmacologically active compounds. The drug saracatinib, a potent inhibitor of Src-family of tyrosine kinases (SFK), was identified as an inhibitor of MERS-CoV replication in vitro. Our results suggest that saracatinib potently inhibits MERS-CoV at the early stages of the viral life cycle in Huh-7 cells, possibly through the suppression of SFK signaling pathways. Furthermore, saracatinib exhibited a synergistic effect with gemcitabine, an anticancer drug with antiviral activity against several RNA viruses. These data indicate that saracatinib alone or in combination with gemcitabine can provide a new therapeutic option for the treatment of MERS-CoV infection.
Collapse
Affiliation(s)
- Jin Soo Shin
- Virus Research Group, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea.
| | - Eunhye Jung
- Virus Research Group, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
| | - Meehyein Kim
- Virus Research Group, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34114, Korea.
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Yun Young Go
- Virus Research Group, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34114, Korea.
| |
Collapse
|
155
|
A Sialylated Voltage-Dependent Ca 2+ Channel Binds Hemagglutinin and Mediates Influenza A Virus Entry into Mammalian Cells. Cell Host Microbe 2018; 23:809-818.e5. [PMID: 29779930 DOI: 10.1016/j.chom.2018.04.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/04/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022]
Abstract
Influenza A virus (IAV) infection is initiated by the attachment of the viral glycoprotein hemagglutinin (HA) to sialic acid on the host cell surface. However, the sialic acid-containing receptor crucial for IAV infection has remained unidentified. Here, we show that HA binds to the voltage-dependent Ca2+ channel Cav1.2 to trigger intracellular Ca2+ oscillations and subsequent IAV entry and replication. IAV entry was inhibited by Ca2+ channel blockers (CCBs) or by knockdown of Cav1.2. The CCB diltiazem also inhibited virus replication in vivo. Reintroduction of wild-type but not the glycosylation-deficient mutants of Cav1.2 restored Ca2+ oscillations and virus infection in Cav1.2-depleted cells, demonstrating the significance of Cav1.2 sialylation. Taken together, we identify Cav1.2 as a sialylated host cell surface receptor that binds HA and is critical for IAV entry.
Collapse
|
156
|
Helenius A. Virus Entry: Looking Back and Moving Forward. J Mol Biol 2018; 430:1853-1862. [PMID: 29709571 PMCID: PMC7094621 DOI: 10.1016/j.jmb.2018.03.034] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/15/2018] [Accepted: 03/16/2018] [Indexed: 12/29/2022]
Abstract
Research over a period of more than half a century has provided a reasonably accurate picture of mechanisms involved in animal virus entry into their host cells. Successive steps in entry include binding to receptors, endocytosis, passage through one or more membranes, targeting to specific sites within the cell, and uncoating of the genome. For some viruses, the molecular interactions are known in great detail. However, as more viruses are analyzed, and as the focus shifts from tissue culture to in vivo experiments, it is evident that viruses display considerable redundancy and flexibility in receptor usage, endocytic mechanism, location of penetration, and uncoating mechanism. For many viruses, the picture is still elusive because the interactions that they engage in rely on sophisticated adaptation to complex cellular functions and defense mechanisms. Studies using a broad combination of technologies have provided detailed information on the entry and uncoating of many animal viruses. Not only the identity of cell surface receptors but their distribution in plasma membrane and in microdomains defines cell tropism and infection efficiency. The majority of viruses enter by endocytic mechanisms and penetrate into the cytosol intracellularly from a variety of different organelles. The picture is often elusive because many viruses display redundancy in receptor choice and entry strategy.
Collapse
Affiliation(s)
- Ari Helenius
- ETH Zurich, Institute of Biochemistry, Otto-Stern-Weg 3, Zurich 8093, Switzerland.
| |
Collapse
|
157
|
Duy J, Honko AN, Altamura LA, Bixler SL, Wollen-Roberts S, Wauquier N, O'Hearn A, Mucker EM, Johnson JC, Shamblin JD, Zelko J, Botto MA, Bangura J, Coomber M, Pitt ML, Gonzalez JP, Schoepp RJ, Goff AJ, Minogue TD. Virus-encoded miRNAs in Ebola virus disease. Sci Rep 2018; 8:6480. [PMID: 29691416 PMCID: PMC5915558 DOI: 10.1038/s41598-018-23916-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
Ebola virus (EBOV) is a negative-strand RNA virus that replicates in the cytoplasm and causes an often-fatal hemorrhagic fever. EBOV, like other viruses, can reportedly encode its own microRNAs (miRNAs) to subvert host immune defenses. miRNAs are short noncoding RNAs that can regulate gene expression by hybridizing to multiple mRNAs, and viral miRNAs can enhance viral replication and infectivity by regulating host or viral genes. To date, only one EBOV miRNA has been examined in human infection. Here, we assayed mouse, rhesus macaque, cynomolgus macaque, and human samples infected with three EBOV variants for twelve computationally predicted viral miRNAs using RT-qPCR. Ten miRNAs aligned to EBOV variants and were detectable in the four species during disease with several viral miRNAs showing presymptomatic amplification in animal models. miRNA abundances in both the mouse and nonhuman primate models mirrored the human cohort, with miR-1-5p, miR-1-3p, and miR-T3-3p consistently at the highest levels. These striking similarities in the most abundant miRNAs during infection with different EBOV variants and hosts indicate that these miRNAs are potential valuable diagnostic markers and key effectors of EBOV pathogenesis.
Collapse
Affiliation(s)
- Janice Duy
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Anna N Honko
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Louis A Altamura
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Sandra L Bixler
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Suzanne Wollen-Roberts
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Nadia Wauquier
- Metabiota, Kenema, Sierra Leone.,MRIGlobal - Global Health Surveillance and Diagnostics, Gaithersburg, MD, USA
| | - Aileen O'Hearn
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Eric M Mucker
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Joshua C Johnson
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Joshua D Shamblin
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Justine Zelko
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Miriam A Botto
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | | | | | - M Louise Pitt
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jean-Paul Gonzalez
- Metabiota, Washington, DC, USA.,Center of Excellence for Emerging & Zoonotic Animal Disease, Kansas State University, Manhattan, KS, USA
| | - Randal J Schoepp
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Arthur J Goff
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Timothy D Minogue
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
158
|
Hsu ACY. Influenza Virus: A Master Tactician in Innate Immune Evasion and Novel Therapeutic Interventions. Front Immunol 2018; 9:743. [PMID: 29755452 PMCID: PMC5932403 DOI: 10.3389/fimmu.2018.00743] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022] Open
Abstract
Influenza is a contagion that has plagued mankind for many decades, and continues to pose concerns every year, with millions of infections globally. The frequent mutations and recombination of the influenza A virus (IAV) cast a looming threat that antigenically novel strains/subtypes will rise with unpredictable pathogenicity and fear of it evolving into a pandemic strain. There have been four major influenza pandemics, since the beginning of twentieth century, with the great 1918 pandemic being the most severe, killing more than 50 million people worldwide. The mechanisms of IAV infection, host immune responses, and how viruses evade from such defensive responses at the molecular and structural levels have been greatly investigated in the past 30 years. While this has advanced our understanding of virus–host interactions and human immunology, and has led to the development of several antiviral drugs, they have minimal impact on the clinical outcomes of infection. The heavy use of these drugs has also imposed selective pressure on IAV to evolve and develop resistance. Vaccination remains the cornerstone of public health efforts to protect against influenza; however, rapid mass-production of sufficient vaccines is unlikely to occur immediately after the beginning of a pandemic. This, therefore, requires novel therapeutic strategies against this continually emerging infectious virus with higher specificity and cross-reactivity against multiple strains/subtypes of IAVs. This review discusses essential virulence factors of IAVs that determine sustainable human-to-human transmission, the mechanisms of viral hijacking of host cells and subversion of host innate immune responses, and novel therapeutic interventions that demonstrate promising antiviral properties against IAV. This hopefully will promote discussions and investigations on novel avenues of prevention and treatment strategies of influenza, that are effective and cross-protective against multiple strains/subtypes of IAV, in preparation for the advent of future IAVs and pandemics.
Collapse
Affiliation(s)
- Alan Chen-Yu Hsu
- Viruses, Infections/Immunity, Vaccines & Asthma, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
159
|
Choi AH, O’Leary MP, Lu J, Kim SI, Fong Y, Chen NG. Endogenous Akt Activity Promotes Virus Entry and Predicts Efficacy of Novel Chimeric Orthopoxvirus in Triple-Negative Breast Cancer. MOLECULAR THERAPY-ONCOLYTICS 2018; 9:22-29. [PMID: 29988465 PMCID: PMC6026447 DOI: 10.1016/j.omto.2018.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/01/2018] [Indexed: 12/04/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with high recurrence rate and poor prognosis. Here, we describe a novel, chimeric orthopoxvirus (CF33) that efficiently kills TNBC. Cytotoxicity was assayed in vitro in four TNBC cell lines. Viral replication was examined through standard plaque assay. Two orthotopic TNBC xenograft models were generated in athymic nude mice and were injected with CF33 intratumorally. CF33 was effective in vitro with potent cytotoxicity and efficient intracellular replication observed in TNBC lines with phosphatidylinositol 3-kinase (PI3K)/Akt pathway mutations that resulted in endogenous phospho-Akt (p-Akt) activity (BT549, Hs578T, and MDA-MB-468). Relative resistance to CF33 by wild-type PI3K/Akt pathway cell line MDA-MB-231 was overcome using higher MOI. The virus was effective in vivo with significant tumor size reduction in both xenograft models. Mechanistically, CF33 appears to share similar properties to vaccinia virus with respect to Akt-mediated and low-pH-mediated viral entry. In summary, CF33 demonstrated potent antitumoral effect in vitro and in vivo, with the most potent effect predicted by the presence of endogenous Akt activity in the TNBC cell line. Further investigation of its mechanism of action as well as genetic modifications to enhance its natural viral tropism are warranted for preclinical development.
Collapse
Affiliation(s)
- Audrey H. Choi
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Michael P. O’Leary
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Nanhai G. Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Gene Editing and Viral Vector Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Corresponding author: Nanhai G. Chen, PhD, Department of Surgery, City of Hope National Medical Center, 1500 E. Duarte Road, Rm 1102, Familian Science Building, Duarte, CA 91010, USA.
| |
Collapse
|
160
|
Abstract
Protein-protein interaction networks (PPIs) collect information on physical-and in some cases-functional interactions between proteins. Most PPIs are annotated with confidence scores, which reflect the probability that a reported interaction is a true interaction. These scores, however, do not allow users to isolate interactions relevant in a particular biological context. Here, we describe solutions for performing context filtering on PPIs to allow biological data interpretation and functional inference in two publicly available PPIs resources (HIPPIE and STRING) and in the proprietary pathway analysis tool and knowledge base Ingenuity Pathway Analysis.
Collapse
|
161
|
Kersh AE, Ng S, Chang YM, Sasaki M, Thomas SN, Kissick HT, Lesinski GB, Kudchadkar RR, Waller EK, Pollack BP. Targeted Therapies: Immunologic Effects and Potential Applications Outside of Cancer. J Clin Pharmacol 2018; 58:7-24. [PMID: 29136276 PMCID: PMC5972536 DOI: 10.1002/jcph.1028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
Two pharmacologic approaches that are currently at the forefront of treating advanced cancer are those that center on disrupting critical growth/survival signaling pathways within tumor cells (commonly referred to as "targeted therapies") and those that center on enhancing the capacity of a patient's immune system to mount an antitumor response (immunotherapy). Maximizing responses to both of these approaches requires an understanding of the oncogenic events present in a given patient's tumor and the nature of the tumor-immune microenvironment. Although these 2 modalities were developed and initially used independently, combination regimens are now being tested in clinical trials, underscoring the need to understand how targeted therapies influence immunologic events. Translational studies and preclinical models have demonstrated that targeted therapies can influence immune cell trafficking, the production of and response to chemokines and cytokines, antigen presentation, and other processes relevant to antitumor immunity and immune homeostasis. Moreover, because these and other effects of targeted therapies occur in nonmalignant cells, targeted therapies are being evaluated for use in applications outside of oncology.
Collapse
Affiliation(s)
- Anna E. Kersh
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Spencer Ng
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yun Min Chang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Atlanta, GA
| | | | - Susan N. Thomas
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haydn T. Kissick
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gregory B. Lesinski
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ragini R. Kudchadkar
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Edmund K. Waller
- Emory University Winship Cancer Institute, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Brian P. Pollack
- Atlanta VA Medical Center, Atlanta, GA, USA
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA
- Emory University Winship Cancer Institute, Atlanta, GA, USA
| |
Collapse
|
162
|
Hu W, Zhu L, Yang X, Lin J, Yang Q. The epidermal growth factor receptor regulates cofilin activity and promotes transmissible gastroenteritis virus entry into intestinal epithelial cells. Oncotarget 2017; 7:12206-21. [PMID: 26933809 PMCID: PMC4914279 DOI: 10.18632/oncotarget.7723] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 01/29/2016] [Indexed: 01/01/2023] Open
Abstract
Transmissible gastroenteritis virus (TGEV), a coronavirus, causes severe diarrhea and high mortality in newborn piglets. The porcine intestinal epithelium is the target of TGEV infection, but the mechanisms that TGEV disrupts the actin cytoskeleton and invades the host epithelium remain largely unknown. We not only found that TGEV infection stimulates F-actin to gather at the cell membrane but the disruption of F-actin inhibits TGEV entry as well. Cofilin is involved in F-actin reorganization and TGEV entry. The TGEV spike protein is capable of binding with EGFR, activating the downstream phosphoinositide-3 kinase (PI3K), then causing the phosphorylation of cofilin and F-actin polymerization via Rac1/Cdc42 GTPases. Inhibition of EGFR and PI3K decreases the entry of TGEV. EGFR is also the upstream activator of mitogen-activated protein kinase (MAPK) signaling pathways that is involved in F-actin reorganization. Additionally, lipid rafts act as signal platforms for the EGFR-associated signaling cascade and correlate with the adhesion of TGEV. In conlusion, these results provide valuable data of the mechanisms which are responsible for the TGEV pathogenesis and may lead to the development of new methods about controlling TGEV.
Collapse
Affiliation(s)
- Weiwei Hu
- Veterinary College, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Liqi Zhu
- Veterinary College, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xing Yang
- Veterinary College, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Jian Lin
- Life Science College, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Qian Yang
- Veterinary College, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| |
Collapse
|
163
|
Schor S, Einav S. Repurposing of Kinase Inhibitors as Broad-Spectrum Antiviral Drugs. DNA Cell Biol 2017; 37:63-69. [PMID: 29148875 DOI: 10.1089/dna.2017.4033] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The high cost of drug development and the narrow spectrum of coverage typically provided by direct-acting antivirals limit the scalability of this antiviral approach. This review summarizes progress and challenges in the repurposing of approved kinase inhibitors as host-targeted broad-spectrum antiviral therapies.
Collapse
Affiliation(s)
- Stanford Schor
- 1 Division of Infectious Diseases and Geographic Medicine , Department of Medicine, Stanford, California.,2 Department of Microbiology and Immunology, Stanford University School of Medicine , Stanford, California
| | - Shirit Einav
- 1 Division of Infectious Diseases and Geographic Medicine , Department of Medicine, Stanford, California.,2 Department of Microbiology and Immunology, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
164
|
Sutherland DM, Aravamudhan P, Dermody TS. An Orchestra of Reovirus Receptors: Still Searching for the Conductor. Adv Virus Res 2017; 100:223-246. [PMID: 29551138 DOI: 10.1016/bs.aivir.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Viruses are constantly engaged in a molecular arms race with the host, where efficient and tactical use of cellular receptors benefits critical steps in infection. Receptor use dictates initiation, establishment, and spread of viral infection to new tissues and hosts. Mammalian orthoreoviruses (reoviruses) are pervasive pathogens that use multiple receptors to overcome protective host barriers to disseminate from sites of initial infection and cause disease in young mammals. In particular, reovirus invades the central nervous system (CNS) with serotype-dependent tropism and disease. A single viral gene, encoding the attachment protein σ1, segregates with distinct patterns of CNS injury. Despite the identification and characterization of several reovirus receptors, host factors that dictate tropism via interaction with σ1 remain undefined. Here, we summarize the state of the reovirus receptor field and discuss open questions toward understanding how the reovirus attachment protein dictates CNS tropism.
Collapse
Affiliation(s)
| | | | - Terence S Dermody
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
165
|
Markov AV, Sen'kova AV, Warszycki D, Salomatina OV, Salakhutdinov NF, Zenkova MA, Logashenko EB. Soloxolone methyl inhibits influenza virus replication and reduces virus-induced lung inflammation. Sci Rep 2017; 7:13968. [PMID: 29070858 PMCID: PMC5656677 DOI: 10.1038/s41598-017-14029-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022] Open
Abstract
Highly pathogenic influenza viruses pose a serious public health threat to humans. Although vaccines are available, new antivirals are needed to efficiently control disease progression and virus transmission due to the emergence of drug-resistant viral strains. In this study, we describe the anti-viral properties of Soloxolone methyl (SM) (methyl 2-cyano-3,12-dioxo-18βH-olean-9(11),1(2)-dien-30-oate, a chemical derivative of glycyrrhetinic acid) against the flu virus. Anti-flu efficacy studies revealed that SM exhibits antiviral activity against the H1N1 influenza A virus in a dose-dependent manner causing a more than 10-fold decrease in virus titer and a reduction in the expression of NP and M2 viral proteins. In a time-of-addition study, SM was found to act at an early stage of infection to exhibit an inhibitory effect on both the attachment step and virus uptake into cells. Also, in infected cells SM downregulates the expression of the inflammatory cytokines IL-6 and TNF-α. In infected mice, SM administered intranasally prior to and after infection significantly decreases virus titers in the lung and prevents post-challenge pneumonia. Together, these results suggest that Soloxolone methyl might serve as an effective therapeutic agent to manage influenza outbreaks and virus-associated complications, and further preclinical and clinical investigation may be warranted.
Collapse
Affiliation(s)
- Andrey V Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, 8, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation
| | - Alexandra V Sen'kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, 8, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation
| | - Dawid Warszycki
- Institute of Pharmacology, Polish Academy of Sciences, 12, Smętna street, Kraków, 31-343, Poland
| | - Oksana V Salomatina
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch Russian Academy of Sciences, 9, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation
| | - Nariman F Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch Russian Academy of Sciences, 9, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation
| | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, 8, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation
| | - Evgeniya B Logashenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch Russian Academy of Sciences, 8, Lavrent'ev ave., Novosibirsk, 630090, Russian Federation.
| |
Collapse
|
166
|
Sogi KM, Lien KA, Johnson JR, Krogan NJ, Stanley SA. The Tyrosine Kinase Inhibitor Gefitinib Restricts Mycobacterium tuberculosis Growth through Increased Lysosomal Biogenesis and Modulation of Cytokine Signaling. ACS Infect Dis 2017; 3:564-574. [PMID: 28537707 PMCID: PMC5728371 DOI: 10.1021/acsinfecdis.7b00046] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Host-directed therapeutics have the potential to combat the global tuberculosis pandemic. We previously identified gefitinib, an inhibitor of EGFR, as a potential host-targeted therapeutic effective against Mycobacterium tuberculosis infection of macrophages and mice. Here we examine the functional consequences of gefitinib treatment on M. tuberculosis infected macrophages. Using phosphoproteomic and transcriptional profiling, we identify two mechanisms by which gefitinib influences macrophage responses to infection to affect cytokine responses and limit replication of M. tuberculosis in macrophages. First, we find that gefitinib treatment of M. tuberculosis infected macrophages inhibits STAT3, a transcription factor known to repress effective immune responses to M. tuberculosis in vivo. Second, we find that gefitinib treatment of M. tuberculosis infected macrophages leads to increased expression of genes involved in lysosomal biogenesis and function and an increase of functional lysosomes in gefitinib treated cells. Furthermore, we show that gefitinib treatment increases the targeting of bacteria to lysosomes, providing an explanation for the cell intrinsic effects of gefitinib treatment on M. tuberculosis infection. Our data provide novel insights into the effects of gefitinib on mammalian cells and into the possible roles for EGFR signaling in macrophages.
Collapse
Affiliation(s)
- Kimberly M. Sogi
- School of Public Health, Division of Infectious Diseases and Vaccinology, Li Ka Shing Center, University of California, Berkeley, California 94720, United States
| | - Katie A. Lien
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| | - Jeffrey R. Johnson
- Department of Cellular and Molecular Pharmacology, University of California—San Francisco, San Francisco, California 94158, United States
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California—San Francisco, San Francisco, California 94158, United States
| | - Sarah A. Stanley
- School of Public Health, Division of Infectious Diseases and Vaccinology, Li Ka Shing Center, University of California, Berkeley, California 94720, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, United States
| |
Collapse
|
167
|
van de Wakker SI, Fischer MJ, Oosting RS. New drug-strategies to tackle viral-host interactions for the treatment of influenza virus infections. Eur J Pharmacol 2017; 809:178-190. [DOI: 10.1016/j.ejphar.2017.05.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 04/24/2017] [Accepted: 05/19/2017] [Indexed: 12/13/2022]
|
168
|
Mitogenic stimulation accelerates influenza-induced mortality by increasing susceptibility of alveolar type II cells to infection. Proc Natl Acad Sci U S A 2017; 114:E6613-E6622. [PMID: 28739896 DOI: 10.1073/pnas.1621172114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Development of pneumonia is the most lethal consequence of influenza, increasing mortality more than 50-fold compared with uncomplicated infection. The spread of viral infection from conducting airways to the alveolar epithelium is therefore a pivotal event in influenza pathogenesis. We found that mitogenic stimulation with keratinocyte growth factor (KGF) markedly accelerated mortality after infectious challenge with influenza A virus (IAV). Coadministration of KGF with IAV markedly accelerated the spread of viral infection from the airways to alveoli compared with challenge with IAV alone, based on spatial and temporal analyses of viral nucleoprotein staining of lung tissue sections and dissociated lung cells. To better define the temporal relationship between KGF administration and susceptibility to IAV infection in vivo, we administered KGF 120, 48, 24, and 0 h before intrapulmonary IAV challenge and assessed the percentages of proliferating and IAV-infected, alveolar type II (AECII) cells in dispersed lung cell populations. Peak AECII infectivity coincided with the timing of KGF administration that also induced peak AECII proliferation. AECII from mice that were given intrapulmonary KGF before isolation and then infected with IAV ex vivo exhibited the same temporal pattern of proliferation and infectious susceptibility. KGF-induced increases in mortality, AECII proliferation, and enhanced IAV susceptibility were all reversed by pretreatment of the animals with the mTOR inhibitor rapamycin before mitogenic stimulation. Taken together, these data suggest mTOR signaling-dependent, mitogenic conditioning of AECII is a determinant of host susceptibility to infection with IAV.
Collapse
|
169
|
Ho J, Moyes DL, Tavassoli M, Naglik JR. The Role of ErbB Receptors in Infection. Trends Microbiol 2017; 25:942-952. [PMID: 28522156 PMCID: PMC7126822 DOI: 10.1016/j.tim.2017.04.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 04/11/2017] [Accepted: 04/24/2017] [Indexed: 01/18/2023]
Abstract
Members of the epidermal growth factor receptor family (ErbB family) possess a wide distribution and diverse functions ranging from cellular growth to migration and apoptosis. Though highly implicated in a variety of cancers, their involvement in infectious disease is less recognised. A growing body of evidence now highlights the importance of the ErbB family in a variety of infections. Their role as growth factor receptors, along with other characteristics, such as surface expression and continuous intracellular trafficking, make this receptor family ideally placed for exploitation by pathogens. Herein, we review our current understanding of the role of the ErbB family in the context of infectious disease, exploring the mechanisms that govern pathogen exploitation of this system. A wide and diverse variety of microbes have each evolved distinct mechanisms to exploit ErbB receptors, highlighting this receptor kinase family as a critical factor in initiation and maintenance of pathogen infections. ErbB family members are utilised by pathogens attempting to gain cellular entry, subvert immune responses, and manipulate the cell cycle of infected host cells. These events support and are necessary for pathogen persistence. Pathogen-mediated ErbB-exploitation may contribute to cellular transformation and oncogenesis in a variety of cancers. The use of existing FDA-approved drugs that target ErbB receptors and associated signalling components may offer potential future therapies against infection.
Collapse
Affiliation(s)
- Jemima Ho
- Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK.
| | - David L Moyes
- Centre for Host Microbiome interactions, Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Mahvash Tavassoli
- Department of Molecular Oncology, Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| | - Julian R Naglik
- Mucosal & Salivary Biology Division, Dental Institute, King's College London SE1 1UL, UK
| |
Collapse
|
170
|
Wang M, Wang S, Wang W, Wang Y, Wang H, Zhu W. Inhibition effects of novel polyketide compound PPQ-B against influenza A virus replication by interfering with the cellular EGFR pathway. Antiviral Res 2017; 143:74-84. [PMID: 28414053 DOI: 10.1016/j.antiviral.2017.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/12/2017] [Indexed: 12/13/2022]
Abstract
Development of anti-influenza A virus (IAV) drugs with novel targets and low toxicity is critical for preparedness against influenza outbreaks. In the current study, our results indicated that the novel polyketide compound purpurquinone B (PPQ-B) derived from acid-tolerant fungus Penicillium purpurogenum strain JS03-21 suppressed the replication of IAV in vitro with low toxicity, and may block some stages after virus adsorption. PPQ-B could inhibit H1N1 (A/Puerto Rico/8/34; PR8), H1N1 (A/California/04/2009; Cal09) and H3N2 (A/swine/Minnesota/02719/2009) virus replication in vitro, suggesting that PPQ-B possesses broad-spectrum anti-IAV activities. PPQ-B's antiviral activity may be largely related to its inhibition of some steps that occur 0-4 h after adsorption. Oral administration of PPQ-B could decrease pulmonary viral titers and improve survival rate in IAV infected mice. PPQ-B also significantly decreased the production of inflammatory factors TNF-α, IL-6, RANTES and KC in IAV infected lungs and A549 cells, suggesting that PPQ-B may also attenuate the inflammatory responses caused by IAV infection. PPQ-B may down-regulate the NF-κB and MAPK pathways to inhibit both virus replication and inflammatory responses. In summary, PPQ-B has the potential to be developed into a novel anti-IAV drug targeting host EGFR pathway in the future.
Collapse
Affiliation(s)
- Miaomiao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Shuyao Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China; Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, PR China.
| | - Yi Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Hui Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Weiming Zhu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| |
Collapse
|
171
|
Wang W, Yin R, Zhang M, Yu R, Hao C, Zhang L, Jiang T. Boronic Acid Modifications Enhance the Anti-Influenza A Virus Activities of Novel Quindoline Derivatives. J Med Chem 2017; 60:2840-2852. [PMID: 28267329 DOI: 10.1021/acs.jmedchem.6b00326] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The unique glycan-binding ability of chemically synthesized boronic acid derivatives makes them emerging candidates for developing anti-influenza A virus (IAV) drugs. Herein we report the synthesis and the anti-IAV activities of three series of novel boronic acid-modified quindoline derivatives both in vitro and in vivo. Boronic acid-modified compounds 6a and 7a effectively prevented the entry of virus RNP into the nucleus, reduced virus titers in IAV infected cells, and also inhibited the activity of viral neuraminidase. Compound 7a possessed broad antiviral spectrum and was able to inhibit cellular NF-κB and MAPK signaling pathways to block IAV infection. More importantly, IAV infected mice treated with compound 7a showed better survival rates than mice treated with oseltamivir, a popular anti-IAV drug. Thus, our study provides not only an antiviral preclinical candidate but also useful information for further research and development of boronic acid-modified anti-IAV drugs.
Collapse
Affiliation(s)
- Wei Wang
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China , Qingdao 266003, P. R. China.,Marine Biomedical Research Institute of Qingdao , Qingdao 266003, P. R. China
| | - Ruijuan Yin
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China , Qingdao 266003, P. R. China.,Marine Biomedical Research Institute of Qingdao , Qingdao 266003, P. R. China
| | - Meng Zhang
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China , Qingdao 266003, P. R. China
| | - Rilei Yu
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China , Qingdao 266003, P. R. China.,Marine Biomedical Research Institute of Qingdao , Qingdao 266003, P. R. China
| | - Cui Hao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University Medical College , Qingdao, 266003, P. R. China
| | - Lijuan Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University Medical College , Qingdao, 266003, P. R. China
| | - Tao Jiang
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China , Qingdao 266003, P. R. China.,Marine Biomedical Research Institute of Qingdao , Qingdao 266003, P. R. China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology , Qingdao, 266237, P. R. China
| |
Collapse
|
172
|
Kedzierski L, Tate MD, Hsu AC, Kolesnik TB, Linossi EM, Dagley L, Dong Z, Freeman S, Infusini G, Starkey MR, Bird NL, Chatfield SM, Babon JJ, Huntington N, Belz G, Webb A, Wark PA, Nicola NA, Xu J, Kedzierska K, Hansbro PM, Nicholson SE. Suppressor of cytokine signaling (SOCS)5 ameliorates influenza infection via inhibition of EGFR signaling. eLife 2017; 6. [PMID: 28195529 PMCID: PMC5354519 DOI: 10.7554/elife.20444] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/06/2017] [Indexed: 12/11/2022] Open
Abstract
Influenza virus infections have a significant impact on global human health. Individuals with suppressed immunity, or suffering from chronic inflammatory conditions such as COPD, are particularly susceptible to influenza. Here we show that suppressor of cytokine signaling (SOCS) five has a pivotal role in restricting influenza A virus in the airway epithelium, through the regulation of epidermal growth factor receptor (EGFR). Socs5-deficient mice exhibit heightened disease severity, with increased viral titres and weight loss. Socs5 levels were differentially regulated in response to distinct influenza viruses (H1N1, H3N2, H5N1 and H11N9) and were reduced in primary epithelial cells from COPD patients, again correlating with increased susceptibility to influenza. Importantly, restoration of SOCS5 levels restricted influenza virus infection, suggesting that manipulating SOCS5 expression and/or SOCS5 targets might be a novel therapeutic approach to influenza.
Collapse
Affiliation(s)
- Lukasz Kedzierski
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia.,Monash University, Clayton, Australia
| | - Alan C Hsu
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Tatiana B Kolesnik
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Edmond M Linossi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Laura Dagley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Zhaoguang Dong
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sarah Freeman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Giuseppe Infusini
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Nicola L Bird
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| | - Simon M Chatfield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Nicholas Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Gabrielle Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Andrew Webb
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Peter Ab Wark
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Nicos A Nicola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Australia
| | - Philip M Hansbro
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute and The University of Newcastle, Newcastle, Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
173
|
Hazra B, Kumawat KL, Basu A. The host microRNA miR-301a blocks the IRF1-mediated neuronal innate immune response to Japanese encephalitis virus infection. Sci Signal 2017; 10:eaaf5185. [PMID: 28196914 DOI: 10.1126/scisignal.aaf5185] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Effective recognition of viral components and the subsequent stimulation of the production of type I interferons (IFNs) is crucial for the induction of host antiviral immunity. The failure of the host to efficiently produce type I IFNs in response to infection by the Japanese encephalitis virus (JEV) is linked with an increased probability for the disease to become lethal. JEV is a neurotropic virus of the Flaviviridae family that causes encephalitis in humans. JEV infection is regulated by several host factors, including microRNAs, which are conserved noncoding RNAs that participate in various physiological and pathological processes. We showed that the JEV-induced expression of miR-301a led to inhibition of the production of type I IFN by reducing the abundances of the transcription factor IFN regulatory factor 1 (IRF1) and the signaling protein suppressor of cytokine signaling 5 (SOCS5). Mechanistically, induction of miR-301a expression during JEV infection required the transcription factor nuclear factor κB. In mouse neurons, neutralization of miR-301a restored the host innate immune response by enabling IFN-β production, thereby restricting viral propagation. Inhibition of miR-301a in mouse brain rescued the production of IRF1 and SOCS5, increased the generation of IFN-β, and reduced the extent of JEV replication, thus improving mouse survival. Thus, our study suggests that the JEV-induced expression of miR-301a assists viral pathogenesis by suppressing IFN production, which might be targeted by antiviral therapies.
Collapse
Affiliation(s)
- Bibhabasu Hazra
- National Brain Research Centre, Manesar, Haryana 122051, India
| | | | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana 122051, India.
| |
Collapse
|
174
|
Wang W, Wu J, Zhang X, Hao C, Zhao X, Jiao G, Shan X, Tai W, Yu G. Inhibition of Influenza A Virus Infection by Fucoidan Targeting Viral Neuraminidase and Cellular EGFR Pathway. Sci Rep 2017; 7:40760. [PMID: 28094330 PMCID: PMC5240104 DOI: 10.1038/srep40760] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 12/09/2016] [Indexed: 11/21/2022] Open
Abstract
Development of novel anti-influenza A virus (IAV) drugs with high efficiency and low toxicity is critical for preparedness against influenza outbreaks. Herein, we investigated the anti-IAV activities and mechanisms of fucoidan in vitro and in vivo. The results showed that a fucoidan KW derived from brown algae Kjellmaniella crassifolia effectively blocked IAV infection in vitro with low toxicity. KW possessed broad anti-IAV spectrum and low tendency of induction of viral resistance, superior to the anti-IAV drug amantadine. KW was capable of inactivating virus particles before infection and blocked some stages after adsorption. KW could bind to viral neuraminidase (NA) and inhibit the activity of NA to block the release of IAV. KW also interfered with the activation of EGFR, PKCα, NF-κB, and Akt, and inhibited both IAV endocytosis and EGFR internalization in IAV-infected cells, suggesting that KW may also inhibit cellular EGFR pathway. Moreover, intranasal administration of KW markedly improved survival and decreased viral titers in IAV-infected mice. Therefore, fucoidan KW has the potential to be developed into a novel nasal drop or spray for prevention and treatment of influenza in the future.
Collapse
Affiliation(s)
- Wei Wang
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Jiandong Wu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Xiaoshuang Zhang
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Cui Hao
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University Medical College, Qingdao, 266003, P. R. China
| | - Xiaoliang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Guangling Jiao
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Xindi Shan
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
| | - Wenjing Tai
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education, Ocean University of China, Qingdao, 266003, P.R. China
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, Ocean University of China, Qingdao, 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| |
Collapse
|
175
|
Haghani A, Mehrbod P, Safi N, Kadir FAA, Omar AR, Ideris A. Edible bird's nest modulate intracellular molecular pathways of influenza A virus infected cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:22. [PMID: 28056926 PMCID: PMC5216576 DOI: 10.1186/s12906-016-1498-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Edible Bird's Nest (EBN) as a popular traditional Chinese medicine is believed to have health enhancing and antiviral activities against influenza A virus (IAV); however, the molecular mechanism behind therapeutic effects of EBN is not well characterized. METHODS In this study, EBNs that underwent different enzymatic preparation were tested against IAV infected cells. 50% cytotoxic concentration (CC50) and 50% inhibitory concentration (IC50) of the EBNs against IAV strain A/Puerto Rico/8/1934(H1N1) were determined by HA and MTT assays. Subsequently, the sialic acid content of the used EBNs were analyzed by fluorometric HPLC. Western Blotting and immunofluorescent staining were used to investigate the effects of EBNs on early endosomal trafficking and autophagy process of influenza virus. RESULTS This study showed that post inoculations of EBNs after enzymatic preparations have the highest efficacy to inhibit IAV. While CC50 of the tested EBNs ranged from 27.5-32 mg/ml, the IC50 of these compounds ranged between 2.5-4.9 mg/ml. EBNs could inhibit IAV as efficient as commercial antiviral agents, such as amantadine and oseltamivir with different mechanisms of action against IAV. The antiviral activity of these EBNs correlated with the content of N-acetyl neuraminic acid. EBNs could affect early endosomal trafficking of the virus by reducing Rab5 and RhoA GTPase proteins and also reoriented actin cytoskeleton of IAV infected cells. In addition, for the first time this study showed that EBNs can inhibit intracellular autophagy process of IAV life cycle as evidenced by reduction of LC3-II and increasing of lysosomal degradation. CONCLUSIONS The results procured in this study support the potential of EBNs as supplementary medication or alternative to antiviral agents to inhibit influenza infections. Evidently, EBNs can be a promising antiviral agent; however, these natural compounds should be screened for their metabolites prior to usage as therapeutic approach.
Collapse
Affiliation(s)
- Amin Haghani
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Parvaneh Mehrbod
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nikoo Safi
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Abdul Rahman Omar
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Aini Ideris
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
176
|
Abstract
At every step of their replication cycle influenza viruses depend heavily on their host cells. The multifaceted interactions that occur between the virus and its host cell determine the outcome of the infection, including efficiency of progeny virus production, tropism, and pathogenicity. In order to understand viral disease and develop therapies for influenza it is therefore pertinent to study the intricate interplay between influenza viruses and their required host factors. Here, we review the current knowledge on host cell factors required by influenza virus at the different stages of the viral replication cycle. We also discuss the roles of host factors in zoonotic transmission of influenza viruses and their potential for developing novel antivirals.
Collapse
|
177
|
Davey RA, Shtanko O, Anantpadma M, Sakurai Y, Chandran K, Maury W. Mechanisms of Filovirus Entry. Curr Top Microbiol Immunol 2017; 411:323-352. [PMID: 28601947 DOI: 10.1007/82_2017_14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Filovirus entry into cells is complex, perhaps as complex as any viral entry mechanism identified to date. However, over the past 10 years, the important events required for filoviruses to enter into the endosomal compartment and fuse with vesicular membranes have been elucidated (Fig. 1). Here, we highlight the important steps that are required for productive entry of filoviruses into mammalian cells.
Collapse
Affiliation(s)
- R A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - O Shtanko
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - M Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Y Sakurai
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - K Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W Maury
- Department of Microbiology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
178
|
Ni R, Zhou J, Hossain N, Chau Y. Virus-inspired nucleic acid delivery system: Linking virus and viral mimicry. Adv Drug Deliv Rev 2016; 106:3-26. [PMID: 27473931 DOI: 10.1016/j.addr.2016.07.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/02/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
Targeted delivery of nucleic acids into disease sites of human body has been attempted for decades, but both viral and non-viral vectors are yet to meet our expectations. Safety concerns and low delivery efficiency are the main limitations of viral and non-viral vectors, respectively. The structure of viruses is both ordered and dynamic, and is believed to be the key for effective transfection. Detailed understanding of the physical properties of viruses, their interaction with cellular components, and responses towards cellular environments leading to transfection would inspire the development of safe and effective non-viral vectors. To this goal, this review systematically summarizes distinctive features of viruses that are implied for efficient nucleic acid delivery but not yet fully explored in current non-viral vectors. The assembly and disassembly of viral structures, presentation of viral ligands, and the subcellular targeting of viruses are emphasized. Moreover, we describe the current development of cationic material-based viral mimicry (CVM) and structural viral mimicry (SVM) in these aspects. In light of the discrepancy, we identify future opportunities for rational design of viral mimics for the efficient delivery of DNA and RNA.
Collapse
Affiliation(s)
- Rong Ni
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Junli Zhou
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Naushad Hossain
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
179
|
Alymova IV, York IA, Air GM, Cipollo JF, Gulati S, Baranovich T, Kumar A, Zeng H, Gansebom S, McCullers JA. Glycosylation changes in the globular head of H3N2 influenza hemagglutinin modulate receptor binding without affecting virus virulence. Sci Rep 2016; 6:36216. [PMID: 27796371 PMCID: PMC5086918 DOI: 10.1038/srep36216] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022] Open
Abstract
Since the emergence of human H3N2 influenza A viruses in the pandemic of 1968, these viruses have become established as strains of moderate severity. A decline in virulence has been accompanied by glycan accumulation on the hemagglutinin globular head, and hemagglutinin receptor binding has changed from recognition of a broad spectrum of glycan receptors to a narrower spectrum. The relationship between increased glycosylation, binding changes, and reduction in H3N2 virulence is not clear. We evaluated the effect of hemagglutinin glycosylation on receptor binding and virulence of engineered H3N2 viruses. We demonstrate that low-binding virus is as virulent as higher binding counterparts, suggesting that H3N2 infection does not require either recognition of a wide variety of, or high avidity binding to, receptors. Among the few glycans recognized with low-binding virus, there were two structures that were bound by the vast majority of H3N2 viruses isolated between 1968 and 2012. We suggest that these two structures support physiologically relevant binding of H3N2 hemagglutinin and that this physiologically relevant binding has not changed since the 1968 pandemic. Therefore binding changes did not contribute to reduced severity of seasonal H3N2 viruses. This work will help direct the search for factors enhancing influenza virulence.
Collapse
Affiliation(s)
- Irina V Alymova
- Influenza Division, National Center for Immunization & Respiratory Diseases, Centers for Disease Control &Prevention, Atlanta, GA, USA
| | - Ian A York
- Influenza Division, National Center for Immunization & Respiratory Diseases, Centers for Disease Control &Prevention, Atlanta, GA, USA
| | - Gillian M Air
- Department of Biochemistry &Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John F Cipollo
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Shelly Gulati
- Department of Biochemistry &Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tatiana Baranovich
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amrita Kumar
- Influenza Division, National Center for Immunization & Respiratory Diseases, Centers for Disease Control &Prevention, Atlanta, GA, USA.,Battelle Memorial Institute, Atlanta, GA, USA
| | - Hui Zeng
- Influenza Division, National Center for Immunization & Respiratory Diseases, Centers for Disease Control &Prevention, Atlanta, GA, USA
| | - Shane Gansebom
- Influenza Division, National Center for Immunization & Respiratory Diseases, Centers for Disease Control &Prevention, Atlanta, GA, USA
| | - Jonathan A McCullers
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, TN, USA
| |
Collapse
|
180
|
Ubiquitin in Influenza Virus Entry and Innate Immunity. Viruses 2016; 8:v8100293. [PMID: 27783058 PMCID: PMC5086625 DOI: 10.3390/v8100293] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/14/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022] Open
Abstract
Viruses are obligatory cellular parasites. Their mission is to enter a host cell, to transfer the viral genome, and to replicate progeny whilst diverting cellular immunity. The role of ubiquitin is to regulate fundamental cellular processes such as endocytosis, protein degradation, and immune signaling. Many viruses including influenza A virus (IAV) usurp ubiquitination and ubiquitin-like modifications to establish infection. In this focused review, we discuss how ubiquitin and unanchored ubiquitin regulate IAV host cell entry, and how histone deacetylase 6 (HDAC6), a cytoplasmic deacetylase with ubiquitin-binding activity, mediates IAV capsid uncoating. We also discuss the roles of ubiquitin in innate immunity and its implications in the IAV life cycle.
Collapse
|
181
|
Acciani TH, Suzuki T, Trapnell BC, Le Cras TD. Epidermal growth factor receptor signalling regulates granulocyte-macrophage colony-stimulating factor production by airway epithelial cells and established allergic airway disease. Clin Exp Allergy 2016; 46:317-28. [PMID: 26263242 DOI: 10.1111/cea.12612] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 07/06/2015] [Accepted: 07/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Airway epithelial cells (AEC) are increasingly recognized as a major signalling centre in the pathogenesis of allergic asthma. A previous study demonstrated that epithelial growth factor receptor (EGFR) signalling in AEC regulated key features of allergic airway disease. However, it is unclear what mediators are regulated by EGFR signalling in AEC, although the production of the pro-inflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) is EGFR dependent in keratinocytes. OBJECTIVES To determine whether EGFR signalling regulates GM-CSF production by human AEC downstream of the clinically relevant mediators house dust mite (HDM) and interleukin (IL)-17A and in a mouse model of established allergic asthma. METHODS EGFR inhibitors were used to determine whether EGFR signalling regulates GM-CSF production by cultured human AEC in response to HDM and IL-17A. The roles of EGFR ligands, p38 mitogen-activated protein kinase (MAPK) and tumour necrosis factor-alpha (TNF-α) converting enzyme (TACE) were also assessed. To determine whether EGFR regulates GM-CSF as well as key asthma characteristics in vivo, mice were chronically exposed to HDM to establish allergic airway disease and then treated with the EGFR inhibitor Erlotinib. RESULTS EGFR inhibition reduced HDM and IL-17A induced GM-CSF production in a dose-dependent manner in cultured human AEC. GM-CSF production also required amphiregulin, p38 MAPK signalling and protease/TACE activity. In mice with established allergic airway disease, EGFR inhibition reduced levels of GM-CSF and TNF-α, as well as airway hyperreactivity, cellular inflammation, smooth muscle thickening and goblet cell metaplasia without changes in IgE and Th1, Th2 and Th17 cytokines. CONCLUSIONS AND CLINICAL RELEVANCE Results link HDM, IL-17A, amphiregulin, EGFR and GM-CSF in a mechanistic pathway in AEC and demonstrate that EGFR regulates GM-CSF production and the severity of established disease in a clinically relevant asthma model. These results identify the EGFR→GM-CSF axis as a target for therapeutic development.
Collapse
Affiliation(s)
- T H Acciani
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - T Suzuki
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Translational Pulmonary Science Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - B C Trapnell
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Translational Pulmonary Science Center, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Pulmonary Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - T D Le Cras
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
182
|
Wang R, Wang X, Wu JQ, Ni B, Wen LB, Huang L, Liao Y, Tong GZ, Ding C, Mao X. Efficient porcine reproductive and respiratory syndrome virus entry in MARC-145 cells requires EGFR-PI3K-AKT-LIMK1-COFILIN signaling pathway. Virus Res 2016; 225:23-32. [DOI: 10.1016/j.virusres.2016.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 07/16/2016] [Accepted: 09/08/2016] [Indexed: 01/24/2023]
|
183
|
Entry of Classical Swine Fever Virus into PK-15 Cells via a pH-, Dynamin-, and Cholesterol-Dependent, Clathrin-Mediated Endocytic Pathway That Requires Rab5 and Rab7. J Virol 2016; 90:9194-208. [PMID: 27489278 DOI: 10.1128/jvi.00688-16] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 07/26/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Classical swine fever virus (CSFV), a member of the genus Pestivirus within the family Flaviviridae, is a small, enveloped, positive-strand RNA virus. Due to its economic importance to the pig industry, the biology and pathogenesis of CSFV have been investigated extensively. However, the mechanisms of CSFV entry into cells are not well characterized. In this study, we used systematic approaches to dissect CSFV cell entry. We first observed that CSFV infection was inhibited by chloroquine and NH4Cl, suggesting that viral entry required a low-pH environment. By using the specific inhibitor dynasore, or by expressing the dominant negative (DN) K44A mutant, we verified that dynamin is required for CSFV entry. CSFV particles were observed to colocalize with clathrin at 5 min postinternalization, and CSFV infection was significantly reduced by chlorpromazine treatment, overexpression of a dominant negative form of the EPS15 protein, or knockdown of the clathrin heavy chain by RNA interference. These results suggested that CSFV entry depends on clathrin. Additionally, we found that endocytosis of CSFV was dependent on membrane cholesterol, while neither the overexpression of a dominant negative caveolin mutant nor the knockdown of caveolin had an effect. These results further suggested that CSFV entry required cholesterol and not caveolae. Importantly, the effect of DN mutants of three Rab proteins that regulate endosomal traffic on CSFV infection was examined. Expression of DN Rab5 and Rab7 mutants, but not the DN Rab11 mutant, significantly inhibited CSFV replication. These results were confirmed by silencing of Rab5 and Rab7. Confocal microscopy showed that virus particles colocalized with Rab5 or Rab7 during the early phase of infection within 45 min after virus entry. These results indicated that after internalization, CSFV moved to early and late endosomes before releasing its RNA. Taken together, our findings demonstrate for the first time that CSFV enters cells through the endocytic pathway, providing new insights into the life cycle of pestiviruses. IMPORTANCE Bovine viral diarrhea virus (BVDV), a single-stranded, positive-sense pestivirus within the family Flaviviridae, is internalized by clathrin-dependent receptor-mediated endocytosis. However, the detailed mechanism of cell entry is unknown for other pestiviruses, such as classical swine fever (CSF) virus (CSFV). CSFV is the etiological agent of CSF, a highly contagious disease of swine that causes numerous deaths in pigs and enormous economic losses in China. Understanding the entry pathway of CSFV will not only advance our knowledge of CSFV infection and pathogenesis but also provide novel drug targets for antiviral intervention. Based on this objective, we used systematic approaches to dissect the pathway of entry of CSFV into PK-15 cells. This is the first report to show that the entry of CSFV into PK-15 cells requires a low-pH environment and involves dynamin- and cholesterol-dependent, clathrin-mediated endocytosis that requires Rab5 and Rab7.
Collapse
|
184
|
Wiedemann A, Mijouin L, Ayoub MA, Barilleau E, Canepa S, Teixeira-Gomes AP, Le Vern Y, Rosselin M, Reiter E, Velge P. Identification of the epidermal growth factor receptor as the receptor for Salmonella Rck-dependent invasion. FASEB J 2016; 30:4180-4191. [PMID: 27609774 DOI: 10.1096/fj.201600701r] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/22/2016] [Indexed: 11/11/2022]
Abstract
The Salmonella Rck outer membrane protein binds to the cell surface, which leads to bacterial internalization via a Zipper mechanism. This invasion process requires induction of cellular signals, including phosphorylation of tyrosine proteins, and activation of c-Src and PI3K, which arises as a result of an interaction with a host cell surface receptor. In this study, epidermal growth factor receptor (EGFR) was identified as the cell signaling receptor required for Rck-mediated adhesion and internalization. First, Rck-mediated adhesion and internalization were shown to be altered when EGFR expression and activity were modulated. Then, immunoprecipitations were performed to demonstrate the Rck-EGFR interaction. Furthermore, surface plasmon resonance biosensor and homogeneous time-resolved fluorescence technologies were used to demonstrate the direct interaction of Rck with the extracellular domain of human EGFR. Finally, our study strongly suggests a noncompetitive binding of Rck and EGF to EGFR. Overall, these results demonstrate that Rck is able to bind to EGFR and thereby establish a tight adherence to provide a signaling cascade, which leads to internalization of Rck-expressing bacteria.-Wiedemann, A., Mijouin, L., Ayoub, M. A., Barilleau, E., Canepa, S., Teixeira-Gomes, A. P., Le Vern, Y., Rosselin, M., Reiter, E., Velge, P. Identification of the epidermal growth factor receptor as the receptor for Salmonella Rck-dependent invasion.
Collapse
Affiliation(s)
- Agnès Wiedemann
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France; .,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Lily Mijouin
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Mohammed Akli Ayoub
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France.,Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France.,Le Studium Loire Valley Institute for Advanced Studies, Orléans, France; and
| | - Emilie Barilleau
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Sylvie Canepa
- Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Ana Paula Teixeira-Gomes
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Yves Le Vern
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Manon Rosselin
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Eric Reiter
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France.,Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France
| | - Philippe Velge
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| |
Collapse
|
185
|
Liu Q, Zhou YH, Ye F, Yang ZQ. Antivirals for Respiratory Viral Infections: Problems and Prospects. Semin Respir Crit Care Med 2016; 37:640-6. [PMID: 27486742 PMCID: PMC7171711 DOI: 10.1055/s-0036-1584803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the past two decades, several newly emerging and reemerging viral respiratory pathogens including several influenza viruses (avian influenza and pandemic influenza), severe acute respiratory syndrome coronavirus (SARS-CoV), and Middle East respiratory syndrome coronavirus (MERS-CoV), have continued to challenge medical and public health systems. Thereafter, the development of cost-effective, broad-spectrum antiviral agents is the urgent mission of both virologists and pharmacologists. Current antiviral developments have focused targets on viral entry, replication, release, and intercellular pathways essential for viral life cycle. Here, we review the current literature on challenges and prospects in the development of these antivirals.
Collapse
Affiliation(s)
- Qiang Liu
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Yuan-Hong Zhou
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Feng Ye
- The First College of Clinical Medical Science, China Three Gorges University/Yichang Central People's Hospital, Yichang, China
| | - Zhan-Qiu Yang
- State Key Laboratory of Virology, Institute of Medical Virology, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
186
|
Kumar R, Agrawal T, Khan NA, Nakayama Y, Medigeshi GR. Identification and characterization of the role of c-terminal Src kinase in dengue virus replication. Sci Rep 2016; 6:30490. [PMID: 27457684 PMCID: PMC4960526 DOI: 10.1038/srep30490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/06/2016] [Indexed: 01/01/2023] Open
Abstract
We screened a siRNA library targeting human tyrosine kinases in Huh-7 cells and identified c-terminal Src kinase (Csk) as one of the kinases involved in dengue virus replication. Knock-down of Csk expression by siRNAs or inhibition of Csk by an inhibitor reduced dengue virus RNA levels but did not affect viral entry. Csk partially colocalized with viral replication compartments. Dengue infection was drastically reduced in cells lacking the three ubiquitous src family kinases, Src, Fyn and Yes. Csk knock-down in these cells failed to block dengue virus replication suggesting that the effect of Csk is via regulation of Src family kinases. Csk was found to be hyper-phosphorylated during dengue infection and inhibition of protein kinase A led to a block in Csk phosphorylation and dengue virus replication. Overexpression studies suggest an important role for the kinase and SH3 domains in this process. Our results identified a novel role for Csk as a host tyrosine kinase involved in dengue virus replication and provide further insights into the role of host factors in dengue replication.
Collapse
Affiliation(s)
- Rinki Kumar
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Haryana, India.,Department of Biotechnology, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Tanvi Agrawal
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Haryana, India
| | - Naseem Ahmed Khan
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Haryana, India
| | - Yuji Nakayama
- Department of Biochemistry &Molecular Biology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Guruprasad R Medigeshi
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Haryana, India
| |
Collapse
|
187
|
Becker B, Shaebani MR, Rammo D, Bubel T, Santen L, Schmitt MJ. Cargo binding promotes KDEL receptor clustering at the mammalian cell surface. Sci Rep 2016; 6:28940. [PMID: 27353000 PMCID: PMC4926219 DOI: 10.1038/srep28940] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/24/2016] [Indexed: 01/06/2023] Open
Abstract
Transmembrane receptor clustering is a ubiquitous phenomenon in pro- and eukaryotic cells to physically sense receptor/ligand interactions and subsequently translate an exogenous signal into a cellular response. Despite that receptor cluster formation has been described for a wide variety of receptors, ranging from chemotactic receptors in bacteria to growth factor and neurotransmitter receptors in mammalian cells, a mechanistic understanding of the underlying molecular processes is still puzzling. In an attempt to fill this gap we followed a combined experimental and theoretical approach by dissecting and modulating cargo binding, internalization and cellular response mediated by KDEL receptors (KDELRs) at the mammalian cell surface after interaction with a model cargo/ligand. Using a fluorescent variant of ricin toxin A chain as KDELR-ligand (eGFP-RTAH/KDEL), we demonstrate that cargo binding induces dose-dependent receptor cluster formation at and subsequent internalization from the membrane which is associated and counteracted by anterograde and microtubule-assisted receptor transport to preferred docking sites at the plasma membrane. By means of analytical arguments and extensive numerical simulations we show that cargo-synchronized receptor transport from and to the membrane is causative for KDELR/cargo cluster formation at the mammalian cell surface.
Collapse
Affiliation(s)
- Björn Becker
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66041 Saarbrücken, Germany
| | - M Reza Shaebani
- Department of Theoretical Physics, Saarland University, D-66041 Saarbrücken, Germany
| | - Domenik Rammo
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66041 Saarbrücken, Germany
| | - Tobias Bubel
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66041 Saarbrücken, Germany
| | - Ludger Santen
- Department of Theoretical Physics, Saarland University, D-66041 Saarbrücken, Germany
| | - Manfred J Schmitt
- Molecular and Cell Biology, Department of Biosciences and Center of Human and Molecular Biology (ZHMB), Saarland University, D-66041 Saarbrücken, Germany
| |
Collapse
|
188
|
Wang R, Wang X, Ni B, Huan CC, Wu JQ, Wen LB, Liao Y, Tong GZ, Ding C, Fan HJ, Mao X. Syndecan-4, a PRRSV attachment factor, mediates PRRSV entry through its interaction with EGFR. Biochem Biophys Res Commun 2016; 475:230-7. [PMID: 27208778 DOI: 10.1016/j.bbrc.2016.05.084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
Abstract
The causative agent of porcine reproductive and respiratory syndrome is the PRRS virus (PRRSV), an enveloped, single-stranded and positive-sense RNA virus. The host factors and mechanisms that are involved in PRRSV entry are still largely unknown. In our present studies, we found that syndecan-4, one of the heparan sulfate proteoglycans, plays a critical role in PRRSV entry, especially in PRRSV attachment. Moreover, EGFR interacts with syndecan-4 in MACR-145 cells and disruption of their interaction impaired PRRSV entry. Furthermore, EGFR inhibitor AG1478 or syndecan-4 derived peptide SSTN87-131 inhibited syndecan-4 endocytosis induced by PRRSV entry. Altogether, syndecan-4, a PRRSV attachment factor, mediated PRRSV entry by interacting with EGFR.
Collapse
Affiliation(s)
- Rui Wang
- College of Veterinary Medicine, Nanjing Agricultural University, China.
| | - Xin Wang
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Bo Ni
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Chang-Chao Huan
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Jia-Qiang Wu
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, China
| | - Li-Bin Wen
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Science, China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Guang-Zhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China
| | - Hong-Jie Fan
- College of Veterinary Medicine, Nanjing Agricultural University, China
| | - Xiang Mao
- College of Veterinary Medicine, Nanjing Agricultural University, China; Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, China.
| |
Collapse
|
189
|
Lépinoux-Chambaud C, Barreau K, Eyer J. The Neurofilament-Derived Peptide NFL-TBS.40-63 Targets Neural Stem Cells and Affects Their Properties. Stem Cells Transl Med 2016; 5:901-13. [PMID: 27177578 DOI: 10.5966/sctm.2015-0221] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/23/2016] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Targeting neural stem cells (NSCs) in the adult brain represents a promising approach for developing new regenerative strategies, because these cells can proliferate, self-renew, and differentiate into new neurons, astrocytes, and oligodendrocytes. Previous work showed that the NFL-TBS.40-63 peptide, corresponding to the sequence of a tubulin-binding site on neurofilaments, can target glioblastoma cells, where it disrupts their microtubules and inhibits their proliferation. We show that this peptide targets NSCs in vitro and in vivo when injected into the cerebrospinal fluid. Although neurosphere formation was not altered by the peptide, the NSC self-renewal capacity and proliferation were reduced and were associated with increased adhesion and differentiation. These results indicate that the NFL-TBS.40-63 peptide represents a new molecular tool to target NSCs to develop new strategies for regenerative medicine and the treatment of brain tumors. SIGNIFICANCE In the present study, the NFL-TBS.40-63 peptide targeted neural stem cells in vitro when isolated from the subventricular zone and in vivo when injected into the cerebrospinal fluid present in the lateral ventricle. The in vitro formation of neurospheres was not altered by the peptide; however, at a high concentration of the peptide, the neural stem cell (NSC) self-renewal capacity and proliferation were reduced and associated with increased adhesion and differentiation. These results indicate that the NFL-TBS.40-63 peptide represents a new molecular tool to target NSCs to develop new strategies for regenerative medicine and the treatment of brain tumors.
Collapse
Affiliation(s)
- Claire Lépinoux-Chambaud
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| | - Kristell Barreau
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| | - Joël Eyer
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| |
Collapse
|
190
|
Yamauchi Y, Greber UF. Principles of Virus Uncoating: Cues and the Snooker Ball. Traffic 2016; 17:569-92. [PMID: 26875443 PMCID: PMC7169695 DOI: 10.1111/tra.12387] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/17/2022]
Abstract
Viruses are spherical or complex shaped carriers of proteins, nucleic acids and sometimes lipids and sugars. They are metastable and poised for structural changes. These features allow viruses to communicate with host cells during entry, and to release the viral genome, a process known as uncoating. Studies have shown that hundreds of host factors directly or indirectly support this process. The cell provides molecules that promote stepwise virus uncoating, and direct the virus to the site of replication. It acts akin to a snooker player who delivers accurate and timely shots (cues) to the ball (virus) to score. The viruses, on the other hand, trick (snooker) the host, hijack its homeostasis systems, and dampen innate immune responses directed against danger signals. In this review, we discuss how cellular cues, facilitators, and built‐in viral mechanisms promote uncoating. Cues come from receptors, enzymes and chemicals that act directly on the virus particle to alter its structure, trafficking and infectivity. Facilitators are defined as host factors that are involved in processes which indirectly enhance entry or uncoating. Unraveling the mechanisms of virus uncoating will continue to enhance understanding of cell functions, and help counteracting infections with chemicals and vaccines.
Collapse
Affiliation(s)
- Yohei Yamauchi
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Urs F Greber
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
191
|
Gschweitl M, Ulbricht A, Barnes CA, Enchev RI, Stoffel-Studer I, Meyer-Schaller N, Huotari J, Yamauchi Y, Greber UF, Helenius A, Peter M. A SPOPL/Cullin-3 ubiquitin ligase complex regulates endocytic trafficking by targeting EPS15 at endosomes. eLife 2016; 5:e13841. [PMID: 27008177 PMCID: PMC4846373 DOI: 10.7554/elife.13841] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/23/2016] [Indexed: 01/01/2023] Open
Abstract
Cullin-3 (CUL3)-based ubiquitin ligases regulate endosome maturation and trafficking of endocytic cargo to lysosomes in mammalian cells. Here, we report that these functions depend on SPOPL, a substrate-specific CUL3 adaptor. We find that SPOPL associates with endosomes and is required for both the formation of multivesicular bodies (MVBs) and the endocytic host cell entry of influenza A virus. In SPOPL-depleted cells, endosomes are enlarged and fail to acquire intraluminal vesicles (ILVs). We identify a critical substrate ubiquitinated by CUL3-SPOPL as EPS15, an endocytic adaptor that also associates with the ESCRT-0 complex members HRS and STAM on endosomes. Indeed, EPS15 is ubiquitinated in a SPOPL-dependent manner, and accumulates with HRS in cells lacking SPOPL. Together, our data indicates that a CUL3-SPOPL E3 ubiquitin ligase complex regulates endocytic trafficking and MVB formation by ubiquitinating and degrading EPS15 at endosomes, thereby influencing influenza A virus infection as well as degradation of EGFR and other EPS15 targets. DOI:http://dx.doi.org/10.7554/eLife.13841.001 Individual cells can move material, collectively referred to as cargo, from the outside environment into the cell interior via a process known as endocytosis. The cell then has different routes to transport the packages of cargo, called endocytic vesicles, to specific locations within the cell. Protein-based molecular machines move the cargo and control how it is selected and targeted to different destinations. For example, a molecular machine that contains a protein called CUL3 labels other components of the system with a chemical tag to regulate the route cargo takes in mammalian cells. However, it was not clear how CUL3 can selectively attach the chemical labels. Gschweitl, Ulbricht et al. have now found that another protein called SPOPL provides selectivity for the CUL3-based machine during endocytosis in human cells. The experiments show that SPOPL attaches to endocytic vesicles, and that CUL3 and SPOPL work together to label a specific component of these vesicles called EPS15. The label changes how EPS15 interacts with other proteins. When SPOPL is not present in a cell, EPS15 is unnaturally stable and occupies many of the routes used by endocytic cargos. The cargo directly interacting with EPS15 is then routed on the fast lane to its destination, while other cargo accumulate in a kind of molecular traffic jam. Other proteins like SPOPL are specific for the endocytic system. Exchange of SPOPL with these similar proteins in the CUL3 machine is likely to chemically label a different set of endocytic proteins. Gschweitl, Ulbricht et al.’s next challenge is to identify the selectivity, targeting and coordination of these exchangeable components in the endocytic system. DOI:http://dx.doi.org/10.7554/eLife.13841.002
Collapse
Affiliation(s)
- Michaela Gschweitl
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Anna Ulbricht
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Christopher A Barnes
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Radoslav I Enchev
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Ingrid Stoffel-Studer
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Nathalie Meyer-Schaller
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Jatta Huotari
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Yohei Yamauchi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Urs F Greber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ari Helenius
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, Eidgenössische Technische Hochschule Zürich, Zurich, Switzerland
| |
Collapse
|
192
|
Stindt S, Cebula P, Albrecht U, Keitel V, Schulte am Esch J, Knoefel WT, Bartenschlager R, Häussinger D, Bode JG. Hepatitis C Virus Activates a Neuregulin-Driven Circuit to Modify Surface Expression of Growth Factor Receptors of the ErbB Family. PLoS One 2016; 11:e0148711. [PMID: 26886748 PMCID: PMC4757098 DOI: 10.1371/journal.pone.0148711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/20/2016] [Indexed: 01/19/2023] Open
Abstract
Recently, the epidermal growth factor (EGF) receptor (EGFR), a member of the ErbB receptor family, and its down-stream signalling have been identified as co-factors for HCV entry and replication. Since EGFR also functions as a heterodimer with other ErbB receptor family members, the subject of the present study was to investigate a possible viral interference with these cellular components. By using genotype 1b replicon cells as well as an infection-based system we found that while transcript and protein levels of EGFR and ErbB2 were up-regulated or unaffected, respectively, HCV induced a substantial reduction of ErbB3 and ErbB4 expression. Down-regulation of ErbB3 expression by HCV involves specificity protein (Sp)1-mediated induction of Neuregulin (NRG)1 expression as well as activation of Akt. Consistently, at transcript level disruption of ErbB3 expression by HCV can be prevented by knockdown of NRG1 or Sp1 expression, whereas reconstitution of ErbB3 protein levels requires inhibition of HCV-induced NRG1 expression and of Akt activity. Interestingly, the NRG1-mediated suppression of ErbB3 expression by HCV results in an enhanced expression of EGFR and ErbB2 on the cell surface, which can be mimicked by siRNA-mediated knockdown of ErbB3 expression. These data delineate a novel mechanism enabling HCV to sway the composition of the ErbB family members on the surface of its host cell by an NRG1-driven circuit and unravels a yet unknown cross-regulation between ErbB3 and the two other family members ErbB2 and EGFR. The shift of the receptor surface expression of the ErbB family towards enhanced expression of ErbB2 and EGFR triggered by HCV was found to promote viral RNA replication and infectivity. This suggests that HCV rearranges expression of ErbB family members to adapt the cellular environment to its requirements.
Collapse
Affiliation(s)
- Sabine Stindt
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Patricia Cebula
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ute Albrecht
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jan Schulte am Esch
- Department of General, Visceral, and Pediatric Surgery, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Wolfram T. Knoefel
- Department of General, Visceral, and Pediatric Surgery, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Ralf Bartenschlager
- Department for Infectious Diseases, Molecular Virology, Heidelberg University, Heidelberg, Germany
- Division for Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Johannes G. Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
193
|
Endocytic function is critical for influenza A virus infection via DC-SIGN and L-SIGN. Sci Rep 2016; 6:19428. [PMID: 26763587 PMCID: PMC4725901 DOI: 10.1038/srep19428] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/11/2015] [Indexed: 01/09/2023] Open
Abstract
The ubiquitous presence of cell-surface sialic acid (SIA) has complicated efforts to identify specific transmembrane glycoproteins that function as bone fide entry receptors for influenza A virus (IAV) infection. The C-type lectin receptors (CLRs) DC-SIGN (CD209) and L-SIGN (CD209L) enhance IAV infection however it is not known if they act as attachment factors, passing virions to other unknown receptors for virus entry, or as authentic entry receptors for CLR-mediated virus uptake and infection. Sialic acid-deficient Lec2 Chinese Hamster Ovary (CHO) cell lines were resistant to IAV infection whereas expression of DC-SIGN/L-SIGN restored susceptibility of Lec2 cells to pH- and dynamin-dependent infection. Moreover, Lec2 cells expressing endocytosis-defective DC-SIGN/L-SIGN retained capacity to bind IAV but showed reduced susceptibility to infection. These studies confirm that DC-SIGN and L-SIGN are authentic endocytic receptors for IAV entry and infection.
Collapse
|
194
|
Han SC, Guo HC, Sun SQ, Jin Y, Wei YQ, Feng X, Yao XP, Cao SZ, Xiang Liu D, Liu XT. Productive Entry of Foot-and-Mouth Disease Virus via Macropinocytosis Independent of Phosphatidylinositol 3-Kinase. Sci Rep 2016; 6:19294. [PMID: 26757826 PMCID: PMC4725844 DOI: 10.1038/srep19294] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/08/2015] [Indexed: 12/12/2022] Open
Abstract
Virus entry is an attractive target for therapeutic intervention. Here, using a combination of electron microscopy, immunofluorescence assay, siRNA interference, specific pharmacological inhibitors, and dominant negative mutation, we demonstrated that the entry of foot-and-mouth disease virus (FMDV) triggered a substantial amount of plasma membrane ruffling. We also found that the internalization of FMDV induced a robust increase in fluid-phase uptake, and virions internalized within macropinosomes colocalized with phase uptake marker dextran. During this stage, the Rac1-Pak1 signaling pathway was activated. After specific inhibition on actin, Na(+)/H(+) exchanger, receptor tyrosine kinase, Rac1, Pak1, myosin II, and protein kinase C, the entry and infection of FMDV significantly decreased. However, inhibition of phosphatidylinositol 3-kinase (PI3K) did not reduce FMDV internalization but increased the viral entry and infection to a certain extent, implying that FMDV entry did not require PI3K activity. Results showed that internalization of FMDV exhibited the main hallmarks of macropinocytosis. Moreover, intracellular trafficking of FMDV involves EEA1/Rab5-positive vesicles. The present study demonstrated macropinocytosis as another endocytic pathway apart from the clathrin-mediated pathway. The findings greatly expand our understanding of the molecular mechanisms of FMDV entry into cells, as well as provide potential insights into the entry mechanisms of other picornaviruses.
Collapse
Affiliation(s)
- Shi-Chong Han
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Hui-Chen Guo
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shi-Qi Sun
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Ye Jin
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yan-Quan Wei
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xia Feng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xue-Ping Yao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Sui-Zhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an, Sichuan, China
| | - Ding Xiang Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Xiang-Tao Liu
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
195
|
Smrt ST, Lorieau JL. Membrane Fusion and Infection of the Influenza Hemagglutinin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 966:37-54. [PMID: 27966108 DOI: 10.1007/5584_2016_174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The influenza virus is a major health concern associated with an estimated 5000 to 30,000 deaths every year (Reed et al. 2015) and a significant economic impact with the development of treatments, vaccinations and research (Molinari et al. 2007). The entirety of the influenza genome is comprised of only eleven coding genes. An enormous degree of variation in non-conserved regions leads to significant challenges in the development of inclusive inhibitors for treatment. The fusion peptide domain of the influenza A hemagglutinin (HA) is a promising candidate for treatment since it is one of the most highly conserved sequences in the influenza genome (Heiny et al. 2007), and it is vital to the viral life cycle. Hemagglutinin is a class I viral fusion protein that catalyzes the membrane fusion process during cellular entry and infection. Impediment of the hemagglutinin's function, either through incomplete post-translational processing (Klenk et al. 1975; Lazarowitz and Choppin 1975) or through mutations (Cross et al. 2001), leads to non-infective virus particles. This review will investigate current research on the role of hemagglutinin in the virus life cycle, its structural biology and mechanism as well as the central role of the hemagglutinin fusion peptide (HAfp) to influenza membrane fusion and infection.
Collapse
Affiliation(s)
- Sean T Smrt
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Justin L Lorieau
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
196
|
Ng WC, Londrigan SL, Nasr N, Cunningham AL, Turville S, Brooks AG, Reading PC. The C-type Lectin Langerin Functions as a Receptor for Attachment and Infectious Entry of Influenza A Virus. J Virol 2016; 90:206-21. [PMID: 26468543 PMCID: PMC4702526 DOI: 10.1128/jvi.01447-15] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/04/2015] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED It is well established that influenza A virus (IAV) attachment to and infection of epithelial cells is dependent on sialic acid (SIA) at the cell surface, although the specific receptors that mediate IAV entry have not been defined and multiple receptors may exist. Lec2 Chinese hamster ovary (CHO) cells are SIA deficient and resistant to IAV infection. Here we demonstrate that the expression of the C-type lectin receptor langerin in Lec2 cells (Lec2-Lg) rendered them permissive to IAV infection, as measured by replication of the viral genome, transcription of viral mRNA, and synthesis of viral proteins. Unlike SIA-dependent infection of parental CHO cells, IAV attachment and infection of Lec2-Lg cells was mediated via lectin-mediated recognition of mannose-rich glycans expressed by the viral hemagglutinin glycoprotein. Lec2 cells expressing endocytosis-defective langerin bound IAV efficiently but remained resistant to IAV infection, confirming that internalization via langerin was essential for infectious entry. Langerin-mediated infection of Lec2-Lg cells was pH and dynamin dependent, occurred via clathrin- and caveolin-mediated endocytic pathways, and utilized early (Rab5(+)) but not late (Rab7(+)) endosomes. This study is the first to demonstrate that langerin represents an authentic receptor that binds and internalizes IAV to facilitate infection. Moreover, it describes a unique experimental system to probe specific pathways and compartments involved in infectious entry following recognition of IAV by a single cell surface receptor. IMPORTANCE On the surface of host cells, sialic acid (SIA) functions as the major attachment factor for influenza A viruses (IAV). However, few studies have identified specific transmembrane receptors that bind and internalize IAV to facilitate infection. Here we identify human langerin as a transmembrane glycoprotein that can act as an attachment factor and a bone fide endocytic receptor for IAV infection. Expression of langerin by an SIA-deficient cell line resistant to IAV rendered cells permissive to infection. As langerin represented the sole receptor for IAV infection in this system, we have defined the pathways and compartments involved in infectious entry of IAV into cells following recognition by langerin.
Collapse
Affiliation(s)
- Wy Ching Ng
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Najla Nasr
- Westmead Millennium Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L Cunningham
- Westmead Millennium Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Stuart Turville
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
197
|
Nellimarla S, Baid K, Loo YM, Gale M, Bowdish DME, Mossman KL. Class A Scavenger Receptor-Mediated Double-Stranded RNA Internalization Is Independent of Innate Antiviral Signaling and Does Not Require Phosphatidylinositol 3-Kinase Activity. THE JOURNAL OF IMMUNOLOGY 2015; 195:3858-65. [PMID: 26363049 DOI: 10.4049/jimmunol.1501028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/17/2015] [Indexed: 11/19/2022]
Abstract
dsRNA is a potent trigger of innate immune signaling, eliciting effects within virally infected cells and after release from dying cells. Given its inherent stability, extracellular dsRNA induces both local and systemic effects. Although the class A scavenger receptors (SR-As) mediate dsRNA entry, it is unknown whether they contribute to signaling beyond ligand internalization. In this study, we investigated whether SR-As contribute to innate immune signaling independent of the classic TLR and retinoic acid-inducible gene-I-like receptor (RLR) pathways. We generated a stable A549 human epithelial cell line with inducible expression of the hepatitis C virus protease NS3/4A, which efficiently cleaves TRIF and IFN-β promoter stimulator 1, adaptors for TLR3 and the RLRs, respectively. Cells expressing NS3/4A and TLR3/MyD88/IFN-β promoter stimulator 1(-/-) mouse embryonic fibroblasts completely lacked antiviral activity to extracellular dsRNA relative to control cells, suggesting that SR-As do not possess signaling capacity independent of TLR3 or the RLRs. Previous studies implicated PI3K signaling in SR-A-mediated activities and in downstream production of type I IFN. We found that SR-A-mediated dsRNA internalization occurs independent of PI3K activation, whereas downstream signaling leading to IFN production was partially dependent on PI3K activity. Overall, these findings suggest that SR-A-mediated dsRNA internalization is independent of innate antiviral signaling.
Collapse
Affiliation(s)
- Srinivas Nellimarla
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| | - Kaushal Baid
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| | - Yueh-Ming Loo
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA 98109; and
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, School of Medicine, University of Washington, Seattle, WA 98109; and
| | - Dawn M E Bowdish
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| | - Karen L Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada L8S 4K1; Department of Biochemistry and Biomedical Sciences, McMaster Immunology Research Center, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| |
Collapse
|
198
|
Decoding protein networks during virus entry by quantitative proteomics. Virus Res 2015; 218:25-39. [PMID: 26365680 PMCID: PMC4914609 DOI: 10.1016/j.virusres.2015.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/31/2015] [Accepted: 09/08/2015] [Indexed: 01/05/2023]
Abstract
Virus entry into host cells relies on interactions between viral and host structures including lipids, carbohydrates and proteins. Particularly, protein–protein interactions between viral surface proteins and host proteins as well as secondary host protein–protein interactions play a pivotal role in coordinating virus binding and uptake. These interactions are dynamic and frequently involve multiprotein complexes. In the past decade mass spectrometry based proteomics methods have reached sensitivities and high throughput compatibilities of genomics methods and now allow the reliable quantitation of proteins in complex samples from limited material. As proteomics provides essential information on the biologically active entity namely the protein, including its posttranslational modifications and its interactions with other proteins, it is an indispensable method in the virologist's toolbox. Here we review protein interactions during virus entry and compare classical biochemical methods to study entry with novel technically advanced quantitative proteomics techniques. We highlight the value of quantitative proteomics in mapping functional virus entry networks, discuss the benefits and limitations and illustrate how the methodology will help resolve unsettled questions in virus entry research in the future.
Collapse
|
199
|
Hackett BA, Yasunaga A, Panda D, Tartell MA, Hopkins KC, Hensley SE, Cherry S. RNASEK is required for internalization of diverse acid-dependent viruses. Proc Natl Acad Sci U S A 2015; 112:7797-802. [PMID: 26056282 PMCID: PMC4485095 DOI: 10.1073/pnas.1424098112] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses must gain entry into cells to establish infection. In general, viruses enter either at the plasma membrane or from intracellular endosomal compartments. Viruses that use endosomal pathways are dependent on the cellular factors that control this process; however, these genes have proven to be essential for endogenous cargo uptake, and thus are of limited value for therapeutic intervention. The identification of genes that are selectively required for viral uptake would make appealing drug targets, as their inhibition would block an early step in the life cycle of diverse viruses. At this time, we lack pan-antiviral therapeutics, in part because of our lack of knowledge of such cellular factors. RNAi screening has begun to reveal previously unknown genes that play roles in viral infection. We identified dRNASEK in two genome-wide RNAi screens performed in Drosophila cells against West Nile and Rift Valley Fever viruses. Here we found that ribonuclease kappa (RNASEK) is essential for the infection of human cells by divergent and unrelated positive- and negative-strand-enveloped viruses from the Flaviviridae, Togaviridae, Bunyaviridae, and Orthomyxoviridae families that all enter cells from endosomal compartments. In contrast, RNASEK was dispensable for viruses, including parainfluenza virus 5 and Coxsackie B virus, that enter at the plasma membrane. RNASEK is dispensable for attachment but is required for uptake of these acid-dependent viruses. Furthermore, this requirement appears specific, as general endocytic uptake of transferrin is unaffected in RNASEK-depleted cells. Therefore, RNASEK is a potential host cell Achilles' heel for viral infection.
Collapse
Affiliation(s)
- Brent A Hackett
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Ari Yasunaga
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Debasis Panda
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Michael A Tartell
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Kaycie C Hopkins
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | | | - Sara Cherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104;
| |
Collapse
|
200
|
Boulant S, Stanifer M, Lozach PY. Dynamics of virus-receptor interactions in virus binding, signaling, and endocytosis. Viruses 2015; 7:2794-815. [PMID: 26043381 PMCID: PMC4488714 DOI: 10.3390/v7062747] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 02/06/2023] Open
Abstract
During viral infection the first challenge that viruses have to overcome is gaining access to the intracellular compartment. The infection process starts when the virus contacts the surface of the host cell. A complex series of events ensues, including diffusion at the host cell membrane surface, binding to receptors, signaling, internalization, and delivery of the genetic information. The focus of this review is on the very initial steps of virus entry, from receptor binding to particle uptake into the host cell. We will discuss how viruses find their receptor, move to sub-membranous regions permissive for entry, and how they hijack the receptor-mediated signaling pathway to promote their internalization.
Collapse
Affiliation(s)
- Steeve Boulant
- CellNetworks-Cluster of Excellence and Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Schaller research group at CellNetworks and DKFZ (German cancer research center), 69120 Heidelberg, Germany.
| | - Megan Stanifer
- CellNetworks-Cluster of Excellence and Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Schaller research group at CellNetworks and DKFZ (German cancer research center), 69120 Heidelberg, Germany.
| | - Pierre-Yves Lozach
- CellNetworks-Cluster of Excellence and Department of Infectious Diseases, Virology, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|