1
|
Dipalo LL, Mikkelsen JG, Gijsbers R, Carlon MS. Trojan Horse-Like Vehicles for CRISPR-Cas Delivery: Engineering Extracellular Vesicles and Virus-Like Particles for Precision Gene Editing in Cystic Fibrosis. Hum Gene Ther 2025. [PMID: 40295092 DOI: 10.1089/hum.2024.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
The advent of genome editing has kindled the hope to cure previously uncurable, life-threatening genetic diseases. However, whether this promise can be ultimately fulfilled depends on how efficiently gene editing agents can be delivered to therapeutically relevant cells. Over time, viruses have evolved into sophisticated, versatile, and biocompatible nanomachines that can be engineered to shuttle payloads to specific cell types. Despite the advances in safety and selectivity, the long-term expression of gene editing agents sustained by viral vectors remains a cause for concern. Cell-derived vesicles (CDVs) are gaining traction as elegant alternatives. CDVs encompass extracellular vesicles (EVs), a diverse set of intrinsically biocompatible and low-immunogenic membranous nanoparticles, and virus-like particles (VLPs), bioparticles with virus-like scaffold and envelope structures, but devoid of genetic material. Both EVs and VLPs can efficiently deliver ribonucleoprotein cargo to the target cell cytoplasm, ensuring that the editing machinery is only transiently active in the cell and thereby increasing its safety. In this review, we explore the natural diversity of CDVs and their potential as delivery vectors for the clustered regularly interspaced short palindromic repeats (CRISPR) machinery. We illustrate different strategies for the optimization of CDV cargo loading and retargeting, highlighting the versatility and tunability of these vehicles. Nonetheless, the lack of robust and standardized protocols for CDV production, purification, and quality assessment still hinders their widespread adoption to further CRISPR-based therapies as advanced "living drugs." We believe that a collective, multifaceted effort is urgently needed to address these critical issues and unlock the full potential of genome-editing technologies to yield safe, easy-to-manufacture, and pharmacologically well-defined therapies. Finally, we discuss the current clinical landscape of lung-directed gene therapies for cystic fibrosis and explore how CDVs could drive significant breakthroughs in in vivo gene editing for this disease.
Collapse
Affiliation(s)
- Laudonia Lidia Dipalo
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | | | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, Advanced Disease Modelling, Targeted Drug Discovery, and Gene Therapy (ADVANTAGE) labs, KU Leuven, Leuven, Belgium
- Leuven Viral Vector Core, group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Marianne S Carlon
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Xie Q, Wang L, Liao X, Huang B, Luo C, Liao G, Yuan L, Liu X, Luo H, Shu Y. Research Progress into the Biological Functions of IFITM3. Viruses 2024; 16:1543. [PMID: 39459876 PMCID: PMC11512382 DOI: 10.3390/v16101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are upregulated by interferons. They are not only highly conserved in evolution but also structurally consistent and have almost identical structural domains and functional domains. They are all transmembrane proteins and have multiple heritable variations in genes. The IFITM protein family is closely related to a variety of biological functions, including antiviral immunity, tumor formation, bone metabolism, cell adhesion, differentiation, and intracellular signal transduction. The progress of the research on its structure and related functions, as represented by IFITM3, is reviewed.
Collapse
Affiliation(s)
- Qian Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Liangliang Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), WHO Collaborating Center for Standardization and Evaluation of Biologicals NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Xinzhong Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Bi Huang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Guancheng Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Lifang Yuan
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Xuejie Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China; (Q.X.); (X.L.); (B.H.); (C.L.); (G.L.); (L.Y.); (X.L.)
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 102629, China
| |
Collapse
|
3
|
Wang J, Luo Y, Katiyar H, Liang C, Liu Q. The Antiviral Activity of Interferon-Induced Transmembrane Proteins and Virus Evasion Strategies. Viruses 2024; 16:734. [PMID: 38793616 PMCID: PMC11125860 DOI: 10.3390/v16050734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Interferons (IFNs) are antiviral cytokines that defend against viral infections by inducing the expression of interferon-stimulated genes (ISGs). Interferon-inducible transmembrane proteins (IFITMs) 1, 2, and 3 are crucial ISG products and members of the CD225 protein family. Compelling evidence shows that IFITMs restrict the infection of many unrelated viruses by inhibiting the virus-cell membrane fusion at the virus entry step via the modulation of lipid composition and membrane properties. Meanwhile, viruses can evade IFITMs' restrictions by either directly interacting with IFITMs via viral glycoproteins or by altering the native entry pathway. At the same time, cumulative evidence suggests context-dependent and multifaceted roles of IFITMs in modulating virus infections and cell signaling. Here, we review the diverse antiviral mechanisms of IFITMs, the viral antagonizing strategies, and the regulation of IFITM activity in host cells. The mechanisms behind the antiviral activity of IFITMs could aid the development of broad-spectrum antivirals and enhance preparedness for future pandemics.
Collapse
Affiliation(s)
- Jingjing Wang
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada; (J.W.); (Y.L.)
| | - Yuhang Luo
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada; (J.W.); (Y.L.)
| | - Harshita Katiyar
- McGill Center for Viral Diseases, Lady Davis Institute, Montreal, QC H3T 1E2, Canada; (H.K.); (C.L.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Chen Liang
- McGill Center for Viral Diseases, Lady Davis Institute, Montreal, QC H3T 1E2, Canada; (H.K.); (C.L.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Qian Liu
- Institute of Parasitology, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada; (J.W.); (Y.L.)
- McGill Center for Viral Diseases, Lady Davis Institute, Montreal, QC H3T 1E2, Canada; (H.K.); (C.L.)
| |
Collapse
|
4
|
Twentyman J, Emerman M, Ohainle M. Capsid-dependent lentiviral restrictions. J Virol 2024; 98:e0030824. [PMID: 38497663 PMCID: PMC11019884 DOI: 10.1128/jvi.00308-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Host antiviral proteins inhibit primate lentiviruses and other retroviruses by targeting many features of the viral life cycle. The lentiviral capsid protein and the assembled viral core are known to be inhibited through multiple, directly acting antiviral proteins. Several phenotypes, including those known as Lv1 through Lv5, have been described as cell type-specific blocks to infection against some but not all primate lentiviruses. Here we review important features of known capsid-targeting blocks to infection together with several blocks to infection for which the genes responsible for the inhibition still remain to be identified. We outline the features of these blocks as well as how current methodologies are now well suited to find these antiviral genes and solve these long-standing mysteries in the HIV and retrovirology fields.
Collapse
Affiliation(s)
- Joy Twentyman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michael Emerman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Molly Ohainle
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
5
|
Yadav K, Rana VS, Anjali, Saurav GK, Rawat N, Kumar A, Sunil S, Singh OP, Rajagopal R. Mucin Protein of Aedes aegypti Interacts with Dengue Virus 2 and Influences Viral Infection. Microbiol Spectr 2023; 11:e0250322. [PMID: 36847498 PMCID: PMC10101019 DOI: 10.1128/spectrum.02503-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
Dengue, caused by dengue virus (DENV), is the most prevalent vector-borne viral disease, posing a serious health concern to 2.5 billion people worldwide. DENV is primarily transmitted among humans by its mosquito vector Aedes aegypti; hence, the identification of a novel dengue virus receptor in mosquitoes is critical for the development of new anti-mosquito measures. In the current study, we have identified peptides which potentially interact with the surface of the virion particles and facilitate virus infection and movement during their life cycle in the mosquito vector. To identify these candidate proteins, we performed phage-display library screening against domain III of the envelope protein (EDIII), which plays an essential role during host cell receptor binding for viral entry. The mucin protein, which shared sequence similarity with the peptide identified in the screening, was cloned, expressed, and purified for in vitro interaction studies. Using in vitro pulldown and virus overlay protein-binding assay (VOPBA), we confirmed the positive interaction of mucin with purified EDIII and whole virion particles. Finally, blocking of mucin protein with anti-mucin antibodies partially reduced DENV titers in infected mosquitos. Moreover, mucin protein was found to be localized in the midgut of Ae. aegypti. IMPORTANCE Identification of interacting protein partners of DENV in the insect vector Aedes aegypti is crucial for designing vector control-based strategies and for understanding the molecular mechanism DENV uses to modulate the host, gain entry, and survive successfully. Similar proteins can be used in generating transmission-blocking vaccines.
Collapse
Affiliation(s)
- Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Anjali
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Zoology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Nitish Rawat
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Ankit Kumar
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Om P. Singh
- National Institute of Malaria Research, New Delhi, India
| | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| |
Collapse
|
6
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
7
|
Regino-Zamarripa NE, Ramírez-Martínez G, Jiménez-Álvarez LA, Cruz-Lagunas A, Gómez-García IA, Ignacio-Cortés S, Márquez-García JE, Pacheco-Hernández LM, Ramírez-Noyola JA, Barquera R, Mendoza-Milla C, Luna-Rivero C, Domínguez-Cherit JG, Ramírez-Rangel R, Rodríguez-Reyna TS, Hernández-Cárdenas CM, Choreño-Parra JA, León-Ávila G, Zúñiga J. Differential Leukocyte Expression of IFITM1 and IFITM3 in Patients with Severe Pandemic Influenza A(H1N1) and COVID-19. J Interferon Cytokine Res 2022; 42:430-443. [PMID: 35708622 PMCID: PMC9422779 DOI: 10.1089/jir.2022.0036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Interferon-induced transmembrane (IFITM) proteins mediate protection against enveloped viruses by blocking membrane fusion at endosomes. IFITM1 and IFITM3 are crucial for protection against influenza, and various single nucleotide polymorphisms altering their function have been linked to disease susceptibility. However, bulk IFITM1 and IFITM3 mRNA expression dynamics and their correlation with clinical outcomes have not been extensively addressed in patients with respiratory infections. In this study, we evaluated the expression of IFITM1 and IFITM3 in peripheral leukocytes from healthy controls and individuals with severe pandemic influenza A(H1N1) or coronavirus disease 2019 (COVID-19). Comparisons between participants grouped according to their clinical characteristics, underlying disease, and outcomes showed that the downregulation of IFITM1 was a distinctive characteristic of severe pandemic influenza A(H1N1) that correlated with outcomes, including mortality. Conversely, increased IFITM3 expression was a common feature of severe pandemic influenza A(H1N1) and COVID-19. Using a high-dose murine model of infection, we confirmed not only the downregulation of IFITM1 but also of IFITM3 in the lungs of mice with severe influenza, as opposed to humans. Analyses in the comparative cohort also indicate the possible participation of IFITM3 in COVID-19. Our results add to the evidence supporting a protective function of IFITM proteins against viral respiratory infections in humans.
Collapse
Affiliation(s)
- Nora E Regino-Zamarripa
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Programa de Doctorado en Ciencias Quimicobiológicas, Sección de Estudios de Posgrado e Investigación, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio and Plan de Ayala s/n, Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Gustavo Ramírez-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - Luis Armando Jiménez-Álvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Sergio Ignacio-Cortés
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - José Eduardo Márquez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - Lynette Miroslava Pacheco-Hernández
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Jazmín Ariadna Ramírez-Noyola
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Programa de Maestría en Ciencias de la Salud, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Salvador Díaz Mirón and Plan de San Luis, Mexico City, Mexico
| | - Rodrigo Barquera
- Department of Archaeogenetics, Max Planck Institute for Science of Human History, Jena, Germany
| | - Criselda Mendoza-Milla
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Cesar Luna-Rivero
- Deparment of Pathology, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - José Guillermo Domínguez-Cherit
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico.,Critical Care Unit, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán, Mexico City, Mexico
| | - Remedios Ramírez-Rangel
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Investigación Científica, Mexico City, Mexico
| | - Tatiana Sofía Rodríguez-Reyna
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán, Mexico City, Mexico
| | - Carmen M Hernández-Cárdenas
- Respiratory Critical Care Unit, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Gloria León-Ávila
- Zoology Deparment, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio and Plan de Ayala s/n, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas," Mexico City, Mexico.,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| |
Collapse
|
8
|
Inchingolo AD, Malcangi G, Inchingolo AM, Piras F, Settanni V, Garofoli G, Palmieri G, Ceci S, Patano A, De Leonardis N, Di Pede C, Montenegro V, Azzollini D, Garibaldi MG, Kruti Z, Tarullo A, Coloccia G, Mancini A, Rapone B, Semjonova A, Hazballa D, D’Oria MT, Jones M, Macchia L, Bordea IR, Scarano A, Lorusso F, Tartaglia GM, Maspero C, Del Fabbro M, Nucci L, Ferati K, Ferati AB, Brienza N, Corriero A, Inchingolo F, Dipalma G. Benefits and Implications of Resveratrol Supplementation on Microbiota Modulations: A Systematic Review of the Literature. Int J Mol Sci 2022; 23:4027. [PMID: 35409389 PMCID: PMC8999966 DOI: 10.3390/ijms23074027] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/31/2022] [Indexed: 01/27/2023] Open
Abstract
Resveratrol is a polyphenol that has been shown to possess many applications in different fields of medicine. This systematic review has drawn attention to the axis between resveratrol and human microbiota, which plays a key role in maintaining an adequate immune response that can lead to different diseases when compromised. Resveratrol can also be an asset in new technologies, such as gene therapy. PubMed, Cochrane Library, Scopus, Web of Science, and Google Scholar were searched to find papers that matched our topic dating from 1 January 2017 up to 18 January 2022, with English-language restriction using the following Boolean keywords: ("resveratrol" AND "microbio*"). Eighteen studies were included as relevant papers matching the purpose of our investigation. Immune response, prevention of thrombotic complications, microbiota, gene therapy, and bone regeneration were retrieved as the main topics. The analyzed studies mostly involved resveratrol supplementation and its effects on human microbiota by trials in vitro, in vivo, and ex vivo. The beneficial activity of resveratrol is evident by analyzing the changes in the host's genetic expression and the gastrointestinal microbial community with its administration. The possibility of identifying individual microbial families may allow to tailor therapeutic plans with targeted polyphenolic diets when associated with microbial dysbiosis, such as inflammatory diseases of the gastrointestinal tract, degenerative diseases, tumors, obesity, diabetes, bone tissue regeneration, and metabolic syndrome.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Fabio Piras
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Vito Settanni
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Grazia Garofoli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Giulia Palmieri
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Sabino Ceci
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Nicole De Leonardis
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Chiara Di Pede
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Valentina Montenegro
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Daniela Azzollini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Maria Grazia Garibaldi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Zamira Kruti
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Antonella Tarullo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Giovanni Coloccia
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Biagio Rapone
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Alexandra Semjonova
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Denisa Hazballa
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
- Kongresi Elbasanit, Aqif Pasha, Rruga, 3001 Elbasan, Albania
| | - Maria Teresa D’Oria
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
- Department of Medical and Biological Sciences, University of Udine, Via delle Scienze, 206, 33100 Udine, Italy
| | - Megan Jones
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Luigi Macchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), University of Bari “Aldo Moro”, 70121 Bari, Italy;
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Gianluca Martino Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20122 Milan, Italy; (G.M.T.); (C.M.); (M.D.F.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Cinzia Maspero
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20122 Milan, Italy; (G.M.T.); (C.M.); (M.D.F.)
- UOC Maxillo-Facial Surgery and Dentistry, Fondazione IRCCS Ca Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Massimo Del Fabbro
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20122 Milan, Italy; (G.M.T.); (C.M.); (M.D.F.)
- IRCCS Orthopedic Institute Galeazzi, 20161 Milan, Italy
| | - Ludovica Nucci
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, Via Luigi de Crecchio, 6, 80138 Naples, Italy;
| | - Kenan Ferati
- Faculty of Medical Sciences, University of Tetovo, 1220 Tetovo, North Macedonia; (K.F.); (A.B.F.)
| | - Arberesha Bexheti Ferati
- Faculty of Medical Sciences, University of Tetovo, 1220 Tetovo, North Macedonia; (K.F.); (A.B.F.)
| | - Nicola Brienza
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (N.B.); (A.C.)
| | - Alberto Corriero
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (N.B.); (A.C.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.D.I.); (G.M.); (A.M.I.); (F.P.); (V.S.); (G.G.); (G.P.); (S.C.); (A.P.); (N.D.L.); (C.D.P.); (V.M.); (D.A.); (M.G.G.); (Z.K.); (A.T.); (G.C.); (A.M.); (B.R.); (A.S.); (D.H.); (M.T.D.); (M.J.); (F.I.); (G.D.)
| |
Collapse
|
9
|
Liu P, Zhang Y, Zhang S, Peng C, Yang W, Li X, Zhang C, Li M, Han J, Lu Y. Integrative overview of IFITMs family based on Bioinformatics analysis. Intractable Rare Dis Res 2021; 10:165-172. [PMID: 34466338 PMCID: PMC8397817 DOI: 10.5582/irdr.2021.01041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/26/2021] [Accepted: 05/13/2021] [Indexed: 01/11/2023] Open
Abstract
Human interferon-induced transmembrane proteins (IFITMs) family is a multi-functional biomacromolecule family playing a critical role in various physiological processes, such as, antiviral immunity, tumor suppression, and bone formation. Although there are many studies proving that a subset of tumors strongly links to the changes of IFITMs, the link between different IFITMs mutant types and diverse tumors has not been studied thoroughly. To investigate the law of expression among IFITMs internal members and the linking of IFITMs mutant types and cancers, online databases were used to pool together relevant data for bioinformatics analysis. Here, we summarize mutations, expression, and functions of human IFITMs, analyze diverse expression levels of IFITMs in physiological and pathological tissues, predict protein-protein interaction (PPI) networks, and target miRNAs and relevant signaling pathways of IFITMs. The results show that IFITM1, IFITM2, and IFITM3 have similar motif pattern constructions and physiological functions, while IFITM5 and IFITM10 show far diversity from them. Particularly, IFITM1-3, in conjunction with interacting proteins, is strongly related to development and overall survival rates of a portion of cancers, including renal cancer and uveal melanoma (UVM). This trait may make IFITM1-3 become a prognostic marker of cancers. Meanwhile, hsa_circ_0116375 has been found as the common circRNA for IFITM2, IFITM3, IFITM5, and IFITM10.
Collapse
Affiliation(s)
- Pengchao Liu
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Yongtao Zhang
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Shanshan Zhang
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Chuanming Peng
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Wei Yang
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Xianxian Li
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Chao Zhang
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Mian Li
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Jinxiang Han
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Yanqin Lu
- Department of Endocrinology, The First Affiliated Hospital of Shandong First Medical University, Ji’nan, Shandong, China
- Key Laboratory for Biotech-Drugs of National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
- Address correspondence to:Yanqin Lu, Shandong First Medical University & Shandong Academy of Medical Sciences. # 6699 Qingdao Road, Ji'nan 250117, China. E-mail:
| |
Collapse
|
10
|
Mourad M, Jacob T, Sadovsky E, Bejerano S, Simone GSD, Bagalkot TR, Zucker J, Yin MT, Chang JY, Liu L, Debelenko L, Shawber CJ, Firestein M, Ouyang Y, Gyamfi-Bannerman C, Penn A, Sorkin A, Wapner R, Sadovsky Y. Placental response to maternal SARS-CoV-2 infection. Sci Rep 2021; 11:14390. [PMID: 34257394 PMCID: PMC8277865 DOI: 10.1038/s41598-021-93931-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic affected people at all ages. Whereas pregnant women seemed to have a worse course of disease than age-matched non-pregnant women, the risk of feto-placental infection is low. Using a cohort of 66 COVID-19-positive women in late pregnancy, we correlated clinical parameters with disease severity, placental histopathology, and the expression of viral entry and Interferon-induced transmembrane (IFITM) antiviral transcripts. All newborns were negative for SARS-CoV-2. None of the demographic parameters or placental histopathological characteristics were associated with disease severity. The fetal-maternal transfer ratio for IgG against the N or S viral proteins was commonly less than one, as recently reported. We found that the expression level of placental ACE2, but not TMPRSS2 or Furin, was higher in women with severe COVID-19. Placental expression of IFITM1 and IFITM3, which have been implicated in antiviral response, was higher in participants with severe disease. We also showed that IFITM3 protein expression, which localized to early and late endosomes, was enhanced in severe COVID-19. Our data suggest an association between disease severity and placental SARS-CoV-2 processing and antiviral pathways, implying a role for these proteins in placental response to SARS-CoV-2.
Collapse
Affiliation(s)
- Mirella Mourad
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Taylor Jacob
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Shai Bejerano
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Glicella Salazar-De Simone
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Jason Zucker
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael T Yin
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Jennifer Y Chang
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Lihong Liu
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Larisa Debelenko
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Carrie J Shawber
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Morgan Firestein
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Cynthia Gyamfi-Bannerman
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Penn
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ronald Wapner
- Department of Obstetrics, Gynecology and Reproductive Science, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Marziali F, Cimarelli A. Membrane Interference Against HIV-1 by Intrinsic Antiviral Factors: The Case of IFITMs. Cells 2021; 10:cells10051171. [PMID: 34065027 PMCID: PMC8151167 DOI: 10.3390/cells10051171] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
HIV-1 is a complex retrovirus that is adapted to replicate in cells of the immune system. To do so, HIV-1, like other viruses, developed strategies to use several cellular processes to its advantage, but had also to come to terms with an arsenal of cellular innate defense proteins, or antiviral factors, that target more or less efficiently, virtually every step of the virus replicative cycle. Among antiviral restriction factors, the family of interferon-induced transmembrane proteins (IFITMs) has emerged as a crucial component of cellular innate defenses for their ability to interfere with both early and late phases of viral replication by inhibiting cellular and viral membranes fusion. Here, we review the enormous advances made since the discovery of IFITMs as interferon-regulated genes more than thirty years ago, with a particular focus on HIV-1 and on the elements that modulate its susceptibility or resistance towards members of this family. Given the recent advances of the field in the elucidation of the mechanism of IFITM inhibition and on the mechanism(s) of viral resistance, we expect that future years will bring novel insights into the definition of the multiple facets of IFITMs and on their possible use for novel therapeutical approaches.
Collapse
Affiliation(s)
- Federico Marziali
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm U1111, CNRS, UMR5308, ENS de Lyon, Université Claude Bernard Lyon 1, 46 Allée d'Italie, 69007 Lyon, France
| | - Andrea Cimarelli
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, Inserm U1111, CNRS, UMR5308, ENS de Lyon, Université Claude Bernard Lyon 1, 46 Allée d'Italie, 69007 Lyon, France
| |
Collapse
|
12
|
Ren L, Du S, Xu W, Li T, Wu S, Jin N, Li C. Current Progress on Host Antiviral Factor IFITMs. Front Immunol 2020; 11:543444. [PMID: 33329509 PMCID: PMC7734444 DOI: 10.3389/fimmu.2020.543444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022] Open
Abstract
Host antiviral factor interferon-induced transmembrane proteins (IFITMs) are a kind of small-molecule transmembrane proteins induced by interferon. Their broad-spectrum antiviral activity and unique ability to inhibit viral invasion have made them a hot molecule in antiviral research in recent years. Since the first demonstration of their natural ability to resist viral infection in 1996, IFITMs have been reported to limit a variety of viral infections, including some major pathogens that seriously endanger human health and social stability, such as influenza A, Ebol, severe acute respiratory syndrome, AIDS, and Zika viruses, etc. Studies show that IFITMs mainly exert antiviral activity during virus entry, specifically interfering with the fusion of the envelope and the endosome membrane or forming fusion micropores to block the virus from entering the cytoplasm. However, their specific mechanism is still unclear. This article mainly reviews the research progress in the structure, evolution, function, and mechanism of IFITMs, which may provide a theoretical basis for clarifying the molecular mechanism of interaction between the molecules and viruses and the research and development of new antiviral drugs based on IFITMs.
Collapse
Affiliation(s)
- Linzhu Ren
- Key Lab for Zoonoses Research, Ministry of Education, Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China
| | - Shouwen Du
- Department of Infectious Diseases, Shenzhen People's Hospital, Second Clinical Hospital of Jinan University, Shenzhen, China
| | - Wang Xu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Academy of Military Medical Sciences, Changchun, China
| | - Tiyuan Li
- Department of Infectious Diseases, Shenzhen People's Hospital, Second Clinical Hospital of Jinan University, Shenzhen, China
| | - Shipin Wu
- Department of Infectious Diseases, Shenzhen People's Hospital, Second Clinical Hospital of Jinan University, Shenzhen, China
| | - Ningyi Jin
- Key Lab for Zoonoses Research, Ministry of Education, Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China.,Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Academy of Military Medical Sciences, Changchun, China
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Academy of Military Medical Sciences, Changchun, China
| |
Collapse
|
13
|
Xing H, Ye L, Fan J, Fu T, Li C, Zhang S, Ren L, Bai J. IFITMs of African Green Monkey Can Inhibit Replication of SFTSV but Not MNV In Vitro. Viral Immunol 2020; 33:634-641. [PMID: 33185509 DOI: 10.1089/vim.2020.0132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interferon-induced transmembrane proteins (IFITMs) are transmembrane proteins induced by interferon that can provide broad-spectrum antiviral activities. However, there are few reports on the antiviral activity of monkey-derived IFITMs. In this study, the IFITM1 and IFITM3 genes of African green monkey (AGM) were cloned and overexpressed in Vero cells, followed by infection with mouse norovirus (MNV) and severe fever with thrombocytopenia syndrome virus (SFTSV). The results showed that monkey IFITM1 and IFITM3 can be stably overexpressed in Vero cells. Both IFITM1 and IFITM3 from AGM could effectively restrict infection by SFTSV, and the viral inhibition rate of IFITM3 was more obvious compared with IFITM1. However, both monkey IFITM1 and IFITM3 had no significant effect on the replication of MNV. These results indicate that different IFITMs have different functions, which may be related to the structure of the host IFITMs and the types of pathogens.
Collapse
Affiliation(s)
- Huanwei Xing
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, China.,PKU-Nanjing Joint Institute of Translational Medicine, Nanjing Raygen Health Molecular Medicine Technology Co., Ltd., Nanjing, China.,Non-Human Primate Research Center, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Li Ye
- Beijing Research Center for Information Technology in Agriculture, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Junwen Fan
- China Beijing Animal Disease Control and Prevention Center, Beijing, China
| | - Tingting Fu
- PKU-Nanjing Joint Institute of Translational Medicine, Nanjing Raygen Health Molecular Medicine Technology Co., Ltd., Nanjing, China.,Non-Human Primate Research Center, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Chang Li
- Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, China
| | - Suhua Zhang
- Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, China
| | - Linzhu Ren
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, Changchun, China
| | - Jieying Bai
- PKU-Nanjing Joint Institute of Translational Medicine, Nanjing Raygen Health Molecular Medicine Technology Co., Ltd., Nanjing, China.,Non-Human Primate Research Center, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
14
|
Resveratrol trimer enhances gene delivery to hematopoietic stem cells by reducing antiviral restriction at endosomes. Blood 2020; 134:1298-1311. [PMID: 31416800 DOI: 10.1182/blood.2019000040] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022] Open
Abstract
Therapeutic gene delivery to hematopoietic stem cells (HSCs) holds great potential as a life-saving treatment of monogenic, oncologic, and infectious diseases. However, clinical gene therapy is severely limited by intrinsic HSC resistance to modification with lentiviral vectors (LVs), thus requiring high doses or repeat LV administration to achieve therapeutic gene correction. Here we show that temporary coapplication of the cyclic resveratrol trimer caraphenol A enhances LV gene delivery efficiency to human and nonhuman primate hematopoietic stem and progenitor cells with integrating and nonintegrating LVs. Although significant ex vivo, this effect was most dramatically observed in human lineages derived from HSCs transplanted into immunodeficient mice. We further show that caraphenol A relieves restriction of LV transduction by altering the levels of interferon-induced transmembrane (IFITM) proteins IFITM2 and IFITM3 and their association with late endosomes, thus augmenting LV core endosomal escape. Caraphenol A-mediated IFITM downregulation did not alter the LV integration pattern or bias lineage differentiation. Taken together, these findings compellingly demonstrate that the pharmacologic modification of intrinsic immune restriction factors is a promising and nontoxic approach for improving LV-mediated gene therapy.
Collapse
|
15
|
Olejnik J, Hume AJ, Leung DW, Amarasinghe GK, Basler CF, Mühlberger E. Filovirus Strategies to Escape Antiviral Responses. Curr Top Microbiol Immunol 2019; 411:293-322. [PMID: 28685291 PMCID: PMC5973841 DOI: 10.1007/82_2017_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Adam J Hume
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Christopher F Basler
- Microbial Pathogenesis, Georgia State University, Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
16
|
Liao Y, Goraya MU, Yuan X, Zhang B, Chiu SH, Chen JL. Functional Involvement of Interferon-Inducible Transmembrane Proteins in Antiviral Immunity. Front Microbiol 2019; 10:1097. [PMID: 31156602 PMCID: PMC6532022 DOI: 10.3389/fmicb.2019.01097] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/30/2019] [Indexed: 01/03/2023] Open
Abstract
Interferons (IFNs) play crucial roles in host defense against viral infections by inducing the expression of numerous IFN-stimulated genes (ISGs) that can activate host antiviral immunity. Interferon-inducible transmembrane proteins (IFITMs), a family of small transmembrane proteins, are critical ISG products. Compelling evidence has implicated that IFITMs can establish an innate immune state to eliminate pathogens efficiently. IFITM proteins can impede broad-spectrum viral infection through various mechanisms. It is generally believed that IFITMs can block the viral entry by suppressing viral membrane fusion. However, some findings indicated that IFITMs might also inhibit viral gene expression and viral protein synthesis and thereby impair viral replication. IFITMs may incorporate into virions during viral assembly and thus reduce the infectivity of nascent virions. The precise inhibitory mechanism of IFITMs on viral infection and replication still requires further exploration. In this review, we highlight the recent findings regarding critical roles of IFITMs in host-virus interaction. We also discuss the molecular mechanisms underlying their functions in antiviral responses.
Collapse
Affiliation(s)
- Yuan Liao
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mohsan Ullah Goraya
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xu Yuan
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baoge Zhang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shih-Hsin Chiu
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Abstract
Rapamycin and its derivatives are specific inhibitors of mammalian target of rapamycin (mTOR) kinase and, as a result, are well-established immunosuppressants and antitumorigenic agents. Additionally, this class of drug promotes gene delivery by facilitating lentiviral vector entry into cells, revealing its potential to improve gene therapy efforts. However, the precise mechanism was unknown. Here, we report that mTOR inhibitor treatment results in down-regulation of the IFN-induced transmembrane (IFITM) proteins. IFITM proteins, especially IFITM3, are potent inhibitors of virus-cell fusion and are broadly active against a range of pathogenic viruses. We found that the effect of rapamycin treatment on lentiviral transduction is diminished upon IFITM silencing or knockout in primary and transformed cells, and the extent of transduction enhancement depends on basal expression of IFITM proteins, with a major contribution from IFITM3. The effect of rapamycin treatment on IFITM3 manifests at the level of protein, but not mRNA, and is selective, as many other endosome-associated transmembrane proteins are unaffected. Rapamycin-mediated degradation of IFITM3 requires endosomal trafficking, ubiquitination, endosomal sorting complex required for transport (ESCRT) machinery, and lysosomal acidification. Since IFITM proteins exhibit broad antiviral activity, we show that mTOR inhibition also promotes infection by another IFITM-sensitive virus, Influenza A virus, but not infection by Sendai virus, which is IFITM-resistant. Our results identify the molecular basis by which mTOR inhibitors enhance virus entry into cells and reveal a previously unrecognized immunosuppressive feature of these clinically important drugs. In addition, this study uncovers a functional convergence between the mTOR pathway and IFITM proteins at endolysosomal membranes.
Collapse
|
18
|
Dornfeld D, Dudek AH, Vausselin T, Günther SC, Hultquist JF, Giese S, Khokhlova-Cubberley D, Chew YC, Pache L, Krogan NJ, Garcia-Sastre A, Schwemmle M, Shaw ML. SMARCA2-regulated host cell factors are required for MxA restriction of influenza A viruses. Sci Rep 2018; 8:2092. [PMID: 29391557 PMCID: PMC5794779 DOI: 10.1038/s41598-018-20458-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/18/2018] [Indexed: 11/29/2022] Open
Abstract
The human interferon (IFN)-induced MxA protein is a key antiviral host restriction factor exhibiting broad antiviral activity against many RNA viruses, including highly pathogenic avian influenza A viruses (IAV) of the H5N1 and H7N7 subtype. To date the mechanism for how MxA exerts its antiviral activity is unclear, however, additional cellular factors are believed to be essential for this activity. To identify MxA cofactors we performed a genome-wide siRNA-based screen in human airway epithelial cells (A549) constitutively expressing MxA using an H5N1 reporter virus. These data were complemented with a proteomic screen to identify MxA-interacting proteins. The combined data identified SMARCA2, the ATPase subunit of the BAF chromatin remodeling complex, as a crucial factor required for the antiviral activity of MxA against IAV. Intriguingly, our data demonstrate that although SMARCA2 is essential for expression of some IFN-stimulated genes (ISGs), and the establishment of an antiviral state, it is not required for expression of MxA, suggesting an indirect effect on MxA activity. Transcriptome analysis of SMARCA2-depleted A549-MxA cells identified a small set of SMARCA2-regulated factors required for activity of MxA, in particular IFITM2 and IGFBP3. These findings reveal that several virus-inducible factors work in concert to enable MxA restriction of IAV.
Collapse
Affiliation(s)
- Dominik Dornfeld
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Alexandra H Dudek
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Thibaut Vausselin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sira C Günther
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Judd F Hultquist
- Quantitative Biosciences Institute, QBI, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA
- J. David Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Sebastian Giese
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | | | - Yap C Chew
- Zymo Research Corp, Irvine, CA, 92614, USA
| | - Lars Pache
- Sanford Burnham Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Nevan J Krogan
- Quantitative Biosciences Institute, QBI, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA
- J. David Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Adolfo Garcia-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Martin Schwemmle
- Institute of Virology, Medical Center, University of Freiburg, 79104, Freiburg, Germany.
- Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
| | - Megan L Shaw
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
19
|
The Interferon-Stimulated Gene Ifitm3 Restricts West Nile Virus Infection and Pathogenesis. J Virol 2016; 90:8212-25. [PMID: 27384652 DOI: 10.1128/jvi.00581-16] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/28/2016] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The interferon-induced transmembrane protein (IFITM) family of proteins inhibit infection of several different enveloped viruses in cell culture by virtue of their ability to restrict entry and fusion from late endosomes. As few studies have evaluated the importance of Ifitm3 in vivo in restricting viral pathogenesis, we investigated its significance as an antiviral gene against West Nile virus (WNV), an encephalitic flavivirus, in cells and mice. Ifitm3(-/-) mice were more vulnerable to lethal WNV infection, and this was associated with greater virus accumulation in peripheral organs and central nervous system tissues. As no difference in viral burden in the brain or spinal cord was observed after direct intracranial inoculation, Ifitm3 likely functions as an antiviral protein in nonneuronal cells. Consistent with this, Ifitm3(-/-) fibroblasts but not dendritic cells resulted in higher yields of WNV in multistep growth analyses. Moreover, transcomplementation experiments showed that Ifitm3 inhibited WNV infection independently of Ifitm1, Ifitm2, Ifitm5, and Ifitm6. Beyond a direct effect on viral infection in cells, analysis of the immune response in WNV-infected Ifitm3(-/-) mice showed decreases in the total number of B cells, CD4(+) T cells, and antigen-specific CD8(+) T cells. Finally, bone marrow chimera experiments demonstrated that Ifitm3 functioned in both radioresistant and radiosensitive cells, as higher levels of WNV were observed in the brain only when Ifitm3 was absent from both compartments. Our analyses suggest that Ifitm3 restricts WNV pathogenesis likely through multiple mechanisms, including the direct control of infection in subsets of cells. IMPORTANCE As part of the mammalian host response to viral infections, hundreds of interferon-stimulated genes (ISGs) are induced. The inhibitory activity of individual ISGs varies depending on the specific cell type and viral pathogen. Among ISGs, the genes encoding interferon-induced transmembrane protein (IFITM) have been reported to inhibit multiple families of viruses in cell culture. However, few reports have evaluated the impact of IFITM genes on viral pathogenesis in vivo In this study, we characterized the antiviral activity of Ifitm3 against West Nile virus (WNV), an encephalitic flavivirus, using mice with a targeted gene deletion of Ifitm3 Based on extensive virological and immunological analyses, we determined that Ifitm3 protects mice from WNV-induced mortality by restricting virus accumulation in peripheral organs and, subsequently, in central nervous system tissues. Our data suggest that Ifitm3 restricts WNV pathogenesis by multiple mechanisms and functions in part by controlling infection in different cell types.
Collapse
|
20
|
Blondel D, Maarifi G, Nisole S, Chelbi-Alix MK. Resistance to Rhabdoviridae Infection and Subversion of Antiviral Responses. Viruses 2015; 7:3675-702. [PMID: 26198243 PMCID: PMC4517123 DOI: 10.3390/v7072794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022] Open
Abstract
Interferon (IFN) treatment induces the expression of hundreds of IFN-stimulated genes (ISGs). However, only a selection of their products have been demonstrated to be responsible for the inhibition of rhabdovirus replication in cultured cells; and only a few have been shown to play a role in mediating the antiviral response in vivo using gene knockout mouse models. IFNs inhibit rhabdovirus replication at different stages via the induction of a variety of ISGs. This review will discuss how individual ISG products confer resistance to rhabdoviruses by blocking viral entry, degrading single stranded viral RNA, inhibiting viral translation or preventing release of virions from the cell. Furthermore, this review will highlight how these viruses counteract the host IFN system.
Collapse
Affiliation(s)
- Danielle Blondel
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS UMR 9198, Université Paris-Sud, Gif-sur-Yvette 91190, France.
| | - Ghizlane Maarifi
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| | - Sébastien Nisole
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| | - Mounira K Chelbi-Alix
- INSERM UMR-S 1124, Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris (FR 3567, CNRS), 75270 Paris Cedex 6, France.
| |
Collapse
|
21
|
Suo S, Ren X. Importance of interferon inducible trans-membrane proteins and retinoic acid inducible gene I for influenza virus replication: A review. Crit Rev Microbiol 2015; 42:75-82. [PMID: 25811941 DOI: 10.3109/1040841x.2014.886553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Understanding the interplay between Influenza viruses and host cells is key to elucidating the pathogenesis of these viruses. Several host factors have been identified that exert antiviral functions; however, influenza viruses continue to replicate utilizing host cell machinery. Herein, we review the mechanisms of action of two host-derived proteins on conferring cellular resistance to the influenza virus; (1) the interferon inducible trans-membrane proteins, 1, 2 and 3, a recently identified family of early restriction factors; and (2) retinoic acid inducible gene I, a key mediator of antiviral immunity. These data may contribute to the design of novel and efficient anti-influenza treatments.
Collapse
Affiliation(s)
- Siqingaowa Suo
- a College of Veterinary Medicine, Northeast Agricultural University , Xiangfang , Harbin , China
| | - Xiaofeng Ren
- a College of Veterinary Medicine, Northeast Agricultural University , Xiangfang , Harbin , China
| |
Collapse
|
22
|
Jia R, Ding S, Pan Q, Liu SL, Qiao W, Liang C. The C-terminal sequence of IFITM1 regulates its anti-HIV-1 activity. PLoS One 2015; 10:e0118794. [PMID: 25738301 PMCID: PMC4349745 DOI: 10.1371/journal.pone.0118794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/11/2015] [Indexed: 11/28/2022] Open
Abstract
The interferon-inducible transmembrane (IFITM) proteins inhibit a wide range of viruses. We previously reported the inhibition of human immunodeficiency virus type 1 (HIV-1) strain BH10 by human IFITM1, 2 and 3. It is unknown whether other HIV-1 strains are similarly inhibited by IFITMs and whether there exists viral countermeasure to overcome IFITM inhibition. We report here that the HIV-1 NL4-3 strain (HIV-1NL4-3) is not restricted by IFITM1 and its viral envelope glycoprotein is partly responsible for this insensitivity. However, HIV-1NL4-3 is profoundly inhibited by an IFITM1 mutant, known as Δ(117–125), which is deleted of 9 amino acids at the C-terminus. In contrast to the wild type IFITM1, which does not affect HIV-1 entry, the Δ(117–125) mutant diminishes HIV-1NL4-3 entry by 3-fold. This inhibition correlates with the predominant localization of Δ(117–125) to the plasma membrane where HIV-1 entry occurs. In spite of strong conservation of IFITM1 among most species, mouse IFITM1 is 19 amino acids shorter at its C-terminus as compared to human IFITM1 and, like the human IFITM1 mutant Δ(117–125), mouse IFITM1 also inhibits HIV-1 entry. This is the first report illustrating the role of viral envelope protein in overcoming IFITM1 restriction. The results also demonstrate the importance of the C-terminal region of IFITM1 in modulating the antiviral function through controlling protein subcellular localization.
Collapse
Affiliation(s)
- Rui Jia
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin, 300071, China
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
| | - Shilei Ding
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
| | - Shan-Lu Liu
- Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, 65211–7310, United States of America
| | - Wentao Qiao
- Key Laboratory of Molecular Microbiology and Biotechnology (Ministry of Education) and Key Laboratory of Microbial Functional Genomics (Tianjin), College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, H3T 1E2, Canada
- Department of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
- * E-mail:
| |
Collapse
|
23
|
Bailey CC, Zhong G, Huang IC, Farzan M. IFITM-Family Proteins: The Cell's First Line of Antiviral Defense. Annu Rev Virol 2014; 1:261-283. [PMID: 25599080 DOI: 10.1146/annurev-virology-031413-085537] [Citation(s) in RCA: 353] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal cells use a wide variety of mechanisms to slow or prevent replication of viruses. These mechanisms are usually mediated by antiviral proteins whose expression and activities can be constitutive but are frequently amplified by interferon induction. Among these interferon-stimulated proteins, members of the IFITM (interferon-induced transmembrane) family are unique because they prevent infection before a virus can traverse the lipid bilayer of the cell. At least three human IFITM proteins-IFITM1, IFITM2, and IFITM3-have antiviral activities. These activities limit infection in cultured cells by many viruses, including dengue virus, Ebola virus, influenza A virus, severe acute respiratory syndrome coronavirus, and West Nile virus. Murine Ifitm3 controls influenza A virus infection in vivo, and polymorphisms in human IFITM3 correlate with the severity of both seasonal and highly pathogenic avian influenza virus. Here we review the discovery and characterization of the IFITM proteins, describe the spectrum of their antiviral activities, and discuss potential mechanisms underlying these effects.
Collapse
Affiliation(s)
- Charles C Bailey
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, Florida 33458
| | - Guocai Zhong
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, Florida 33458
| | - I-Chueh Huang
- Department of Cell Biology and Neuroscience, College of Natural and Agricultural Sciences, University of California, Riverside, California 92521
| | - Michael Farzan
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, Florida 33458
| |
Collapse
|
24
|
Ding S, Pan Q, Liu SL, Liang C. HIV-1 mutates to evade IFITM1 restriction. Virology 2014; 454-455:11-24. [PMID: 24725927 PMCID: PMC4274668 DOI: 10.1016/j.virol.2014.01.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 11/25/2013] [Accepted: 01/25/2014] [Indexed: 12/04/2022]
Abstract
Interferon-induced transmembrane (IFITM) proteins inhibit the infection of a wide range of viruses including human immunodeficiency virus type 1 (HIV-1). At present, little is known about how viruses overcome IFITM restriction. In this study, we have utilized HIV-1 as a model and selected IFITM1-resistant viruses after multiple passages of HIV-1 in IFITM1-expressing SupT1 cells. Sequencing the entire viral genome revealed several mutations in the vpu and envelope genes, among which mutations Vpu34 and EnvG367E together enable efficient HIV-1 replication in IFITM1-expressing cells. Vpu34 introduces a stop codon at amino acid position 35 of Vpu, whereas EnvG367E changes the G367 residue at the CD4-binding site of gp120. These two mutations do not appear to overcome the downregulation of viral p24 expression caused by IFITM1, but rather enhance HIV-1 replication by promoting cell-to-cell virus transmission. Altogether, our data demonstrate that HIV-1 can mutate to evade IFITM1 restriction by increasing cell-to-cell transmission. IFITM1 inhibits HIV-1 replication in SupT1 cells. HIV-1 evolves to escape IFITM1 inhibition by mutating Vpu and Env. The Vpu and Env mutations overcome IFITM1 through promoting HIV-1 cell to cell transmission.
Collapse
Affiliation(s)
- Shilei Ding
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada H3T 1E2; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada H3A 2B4
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada H3T 1E2
| | - Shan-Lu Liu
- Department of Molecular Microbiology & Immunology, School of Medicine, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211-7310, USA.
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada H3T 1E2; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada H3A 2B4; Department of Medicine, McGill University, Montreal, QC, Canada H3A 2B4.
| |
Collapse
|
25
|
Abstract
Interferon-induced transmembrane (IFITM) proteins are a family of viral restriction factors that inhibit the entry processes of several pathogenic viruses, including influenza A virus (IAV), in vitro. Here we report that IAV-infected knockout mice lacking the Ifitm locus on chromosome 7 exhibited accelerated disease progression, greater mortality, and higher pulmonary and systemic viral burdens as compared to wild type controls. We further observed that the phenotype of Ifitm3-specific knockout mice was indistinguishable from that of mice lacking the entire Ifitm locus. Ifitm3 was expressed by IAV target cells including alveolar type II pneumocytes and tracheal/bronchial respiratory epithelial cells. Robust Ifitm3 expression was also observed in several tissues in the absence of infection. Among murine Ifitm promoters, only that of Ifitm3 could be induced by type I and II interferons. Ifitm3 could also be upregulated by the gp130 cytokines IL-6 and oncostatin M on cells expressing appropriate receptors, suggesting that multiple cytokine signals could contribute to Ifitm3 expression in a cell or tissue-specific manner. Collectively, these findings establish a central role for Ifitm3 in limiting acute influenza in vivo, and provide further insight into Ifitm3 expression and regulation. The human genome contains many genes devoted to combating viral infections. Some of these genes encode a family of proteins called interferon-induced transmembrane (IFITM) proteins which were recently discovered to inhibit infection by influenza A viruses in cell culture experiments. Here we show that genetically engineered mice lacking the murine equivalents of the human IFITM genes are more susceptible to influenza than mice with a full complement of these genes. In addition, deletion of one of these genes alone, Ifitm3, made mice equally susceptible to infection, showing that the Ifitm3 protein plays a central role in the control of influenza A virus in living animals. We also show that murine Ifitm proteins are expressed on cells targeted by influenza A viruses and that the control of their expression in animals is more complex than suggested by previous cell culture studies.
Collapse
|
26
|
Strobl B, Leitner NR, Müller M. Multifaceted Antiviral Actions of Interferon-stimulated Gene Products. JAK-STAT SIGNALING : FROM BASICS TO DISEASE 2012. [PMCID: PMC7121797 DOI: 10.1007/978-3-7091-0891-8_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Interferons (IFNs) are extremely powerful cytokines for the host defence against viral infections. Binding of IFNs to their receptors activates the JAK/STAT signalling pathway with the Janus kinases JAK1, 2 and TYK2 and the signal transducer and activators of transcription (STAT) 1 and STAT2. Depending on the cellular setting, additional STATs (STAT3-6) and additional signalling pathways are activated. The actions of IFNs on infected cells and the surrounding tissue are mediated by the induction of several hundred IFN-stimulated genes (ISGs). Since the cloning of the first ISGs, considerable progress has been made in describing antiviral effector proteins and their many modes of action. Effector proteins individually target distinct steps in the viral life cycle, including blocking virus entry, inhibition of viral transcription and translation, modification of viral nucleic acids and proteins and, interference with virus assembly and budding. Novel pathways of viral inhibition are constantly being elucidated and, additionally, unanticipated functions of known antiviral effector proteins are discovered. Herein, we outline IFN-induced antiviral pathways and review recent developments in this fascinating area of research.
Collapse
|
27
|
Siegrist F, Ebeling M, Certa U. The small interferon-induced transmembrane genes and proteins. J Interferon Cytokine Res 2010; 31:183-97. [PMID: 21166591 DOI: 10.1089/jir.2010.0112] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interferon-induced transmembrane (IFITM) genes are transcribed in most tissues and are with the exception of IFITM5 interferon inducible. They are involved in early development, cell adhesion, and control of cell growth. Most IFITM genes are activated in response to bacterial and viral infections, and the exact host immune defense mechanisms are still unknown. Elevated gene expression triggered by past or chronic inflammation could prevent spreading of pathogens by limiting host cell proliferation. Accordingly, induction in cells with low basal protein levels is sufficient to drive growth arrest and a senescence-like morphology. On the other hand, loss of IFITM levels in cancer is correlated with pronounced malignancy; thus, these genes are considered as tumor suppressors. However, several cancer cells have deregulated high levels of IFITM transcripts, indicating a tumor progression stage where at least one of the interferon-controlled antiproliferative pathways has been silenced. Phylogenetic analyses of the protein coding genomic sequences suggest a single interferon-inducible gene in the common ancestor of rodents and primates. Biological functions studied so far may have evolved in parallel, and functional characterization of IFITM proteins will provide insight into innate immune defense, cancer development, and other pathways.
Collapse
Affiliation(s)
- Fredy Siegrist
- Non-Clinical Safety, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | | |
Collapse
|
28
|
Palmitoylome profiling reveals S-palmitoylation-dependent antiviral activity of IFITM3. Nat Chem Biol 2010; 6:610-4. [PMID: 20601941 DOI: 10.1038/nchembio.405] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 06/03/2010] [Indexed: 02/06/2023]
Abstract
Identification of immune effectors and the post-translational modifications that control their activity is essential for dissecting mechanisms of immunity. Here we demonstrate that the antiviral activity of interferon-induced transmembrane protein 3 (IFITM3) is post-translationally regulated by S-palmitoylation. Large-scale profiling of palmitoylated proteins in a dendritic cell line using a chemical reporter strategy revealed over 150 lipid-modified proteins with diverse cellular functions, including innate immunity. We discovered that S-palmitoylation of IFITM3 on membrane-proximal cysteines controls its clustering in membrane compartments and its antiviral activity against influenza virus. The sites of S-palmitoylation are highly conserved among the IFITM family of proteins in vertebrates, which suggests that S-palmitoylation of these immune effectors may be an ancient post-translational modification that is crucial for host resistance to viral infections. The S-palmitoylation and clustering of IFITM3 will be important for elucidating its mechanism of action and for the design of antiviral therapeutics.
Collapse
|
29
|
The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 2010; 139:1243-54. [PMID: 20064371 PMCID: PMC2824905 DOI: 10.1016/j.cell.2009.12.017] [Citation(s) in RCA: 1023] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 12/01/2009] [Accepted: 12/09/2009] [Indexed: 12/14/2022]
Abstract
Influenza viruses exploit host cell machinery to replicate, resulting in epidemics of respiratory illness. In turn, the host expresses antiviral restriction factors to defend against infection. To find host cell modifiers of influenza A H1N1 viral infection, we used a functional genomic screen and identified over 120 influenza A virus-dependency factors with roles in endosomal acidification, vesicular trafficking, mitochondrial metabolism, and RNA splicing. We discovered that the interferon-inducible transmembrane proteins IFITM1, 2, and 3 restrict an early step in influenza A viral replication. The IFITM proteins confer basal resistance to influenza A virus but are also inducible by interferons type I and II and are critical for interferon's virustatic actions. Further characterization revealed that the IFITM proteins inhibit the early replication of flaviviruses, including dengue virus and West Nile virus. Collectively this work identifies a family of antiviral restriction factors that mediate cellular innate immunity to at least three major human pathogens.
Collapse
|
30
|
Salas S, Jézéquel P, Campion L, Deville JL, Chibon F, Bartoli C, Gentet JC, Charbonnel C, Gouraud W, Voutsinos-Porche B, Brouchet A, Duffaud F, Figarella-Branger D, Bouvier C. Molecular characterization of the response to chemotherapy in conventional osteosarcomas: predictive value of HSD17B10 and IFITM2. Int J Cancer 2009; 125:851-60. [PMID: 19449377 DOI: 10.1002/ijc.24457] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The therapy regimen of high-grade osteosarcoma includes chemotherapy followed by surgical resection and postoperative chemotherapy. The degree of necrosis following definitive surgery remains the only reliable prognostic factor and is used to guide the choice of postoperative chemotherapy. The aim of this study was to find molecular markers able to classify patients with an osteosarcoma as good or poor responders to chemotherapy before beginning treatment. Gene expression screening of 20 nonmetastatic high-grade osteosarcoma patients was performed using cDNA microarray. Expression of selected relevant genes was validated using QRT-PCR. Immunohistochemistry on tissue microarrays sections of 73 biopsies was performed to investigate protein expression. Fluorescent in situ hybridization was performed for RPL8 gene. We have found that HSD17B10 gene expression was up-regulated in poor responders and that immunohistochemistry expression of HSD17B10 on biopsy before treatment was correlated to response to chemotherapy. Other results include correlation of IFITM2, IFITM3, and RPL8 gene expression to chemotherapy response. A statistical correlation was found between polysomy 8 or gain of RPL8 and good response to chemotherapy. These data suggest that HSD17B10, RPL8, IFITM2, and IFITM3 genes are involved in the response to the chemotherapy and that HSD17B10 may be a therapeutic target. RPL8 and IFITM2 may be useful in the assessment at diagnosis and for stratifying patients taking part in randomized trials.
Collapse
Affiliation(s)
- Sébastien Salas
- Service Oncologie Médicale, Hôpital de la Timone, Assistance Publique-Hopitaux de Marseille, Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Normal germ line establishment in mice carrying a deletion of the Ifitm/Fragilis gene family cluster. Mol Cell Biol 2008; 28:4688-96. [PMID: 18505827 DOI: 10.1128/mcb.00272-08] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The family of interferon-inducible transmembrane proteins (Ifitm) consists of five highly sequence-related cell surface proteins, which are implicated in diverse cellular processes. Ifitm genes are conserved, widely expressed, and characteristically found in genomic clusters, such as the 67-kb Ifitm family locus on mouse chromosome 7. Recently, Ifitm1 and Ifitm3 have been suggested to mediate migration of early primordial germ cells (PGCs), a process that is little understood. To investigate Ifitm function during germ cell development, we used targeted chromosome engineering to generate mutants which either lack the entire Ifitm locus or carry a disrupted Ifitm3 gene only. Here we show that the mutations have no detectable effects on development of the germ line or on the generation of live young. Hence, contrary to previous reports, Ifitm genes are not essential for PGC migration. The Ifitm family is a striking example of a conserved gene cluster which appears to be functionally redundant during development.
Collapse
|
32
|
Liu L, Li P, Jiang L, Dong C, Wang L, Che Y, Zhao H, Dong S, Li Q. Using interferon-alpha to block expression of cellular ribosome subunit S24 variant 2 in human fibroblasts inhibits translation of the poliovirus genome. Viral Immunol 2007; 20:142-9. [PMID: 17425428 DOI: 10.1089/vim.2006.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Studies about the proteins induced by interferon (IFN-)-alpha stimulation have provided some data on their mechanism of antiviral effect. These proteins were confirmed to contribute to antiviral functions. In this study, IFN-alpha stimulation of human fibroblasts was shown to induce the inhibition of S24 variant 2 (a structural component of the ribosomal small subunit) at the mRNA and protein levels, implying a possible antiviral mechanism for IFN-alpha in human fibroblasts. The delay of poliovirus replication by IFN-alpha was partially compensated for by S24 variant 2 expressed in pcDNA vector-transfected cells, and the interference RNA of S24 variant 2 was able to induce mimetically, to some extent, this poliovirus replication delay. These observations revealed that S24 variant 2 could be involved in the antiviral effects of IFN-alpha in human fibroblasts.
Collapse
Affiliation(s)
- Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yang G, Xu Y, Chen X, Hu G. IFITM1 plays an essential role in the antiproliferative action of interferon-gamma. Oncogene 2006; 26:594-603. [PMID: 16847454 DOI: 10.1038/sj.onc.1209807] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interferon-gamma (IFN-gamma) is a pleiotropic cytokine involved in antiproliferative and anti-virus responses, immune surveillance and tumor suppression. These biological responses to IFN-gamma are mainly mediated by the regulation of gene expression. It has been reported that growth-inhibitory role of IFN-gamma is dependent on activation of signal transducers and activators of transcription 1 (STAT1); however, the molecular basis downstream of STAT1 remains unclear. Here, we report that an IFN-gamma-induced gene, interferon-induced transmembrane protein 1 (IFITM1), plays a key role in the antiproliferative action of IFN-gamma. Overexpression of IFITM1 negatively regulated cell growth, whereas suppression of IFITM1 blocked the antiproliferative effect of IFN-gamma, accelerated the cell growth rate and conferred tumorigenicity to a non-malignant hepatocyte in nude mice. Further, IFITM1 could inhibit the activity of extracellular signal-regulated kinase, enhance the transcriptional activity of p53 and stabilize the p53 protein by inhibiting p53 phosphorylation on Thr55. Suppression of p53 reduced the growth-inhibitory capacity of both IFITM1 and IFN-gamma. Therefore, these findings indicated that the antiproliferative action of IFN-gamma requires the induction of IFITM1, and provided a crosstalk between two well-known signaling mediators, STAT1 and p53, both of which play critical roles in tumor suppression.
Collapse
Affiliation(s)
- G Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | |
Collapse
|
34
|
Abstract
Interferons (IFNs) were first characterized as antiviral proteins. Since then, IFNs have proved to be involved in malignant, angiogenic, inflammatory, immune, and fibrous diseases and, thus, possess a broad spectrum of pathophysiologic properties. IFNs activate a cascade of intracellular signaling pathways leading to upregulation of more than 1000 IFN-stimulated genes (ISGs) within the cell. The function of some of the IFN-induced proteins is well described, whereas that of many others remain poorly characterized. This review focuses on three families of small intracellular and intrinsically nonsecreted proteins (10-20 kDa) separated into groups according to their amino acid sequence similarity: the ISG12 group (6-16, ISG12, and ISG12-S), the 1-8 group (9-27/Leu13, 1-8U, and 1-8D), and the ISG15 group (ISG15/UCRP). These IFN-induced genes are abundantly and widely expressed and mainly induced by type I IFN. ISG15 is very well described and is a member of the ubiquitin-like group of proteins. 9-27/Leu-13 associates with CD81/TAPA-1 and plays a role in B cell development. The functions of 1-8U, 1-8D, 6-16, ISG12, and ISG12-S proteins are unknown at present.
Collapse
|
35
|
Radaeva S, Jaruga B, Hong F, Kim WH, Fan S, Cai H, Strom S, Liu Y, El-Assal O, Gao B. Interferon-alpha activates multiple STAT signals and down-regulates c-Met in primary human hepatocytes. Gastroenterology 2002; 122:1020-1034. [PMID: 11910354 DOI: 10.1053/gast.2002.32388] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS Interferon (IFN)-alpha therapy is currently the primary choice for viral hepatitis and a promising treatment for hepatocellular carcinoma (HCC). Primary mouse and rat hepatocytes respond poorly to IFN-alpha stimulation. Thus, it is very important to examine the IFN-alpha signal pathway in primary human hepatocytes. METHODS The IFN-alpha-activated signals and genes in primary human hepatocytes and hepatoma cells were examined by Western blotting and microarray analyses. RESULTS Primary human hepatocytes respond very well to IFN-alpha stimulation as shown by activation of multiple signal transducer and activator of transcription factor (STAT) 1, 2, 3, 5, and multiple genes. The differential response to IFN-alpha stimulation in primary human and mouse hepatocytes may be caused by expression of predominant functional IFN-alpha receptor 2c (IFNAR2c) in primary human hepatocytes vs. expression of predominant inhibitory IFNAR2a in mouse hepatocytes. Microarray analyses of primary human hepatocytes show that IFN-alpha up-regulates about 44 genes by over 2-fold and down-regulates about 9 genes by 50%. The up-regulated genes include a variety of antiviral and tumor suppressors/proapoptotic genes. The down-regulated genes include c-myc and c-Met, the hepatocyte growth factor (HGF) receptor. Down-regulation of c-Met is caused by IFN-alpha suppression of the c-Met promoter through down-regulation of Sp1 binding and results in attenuation of HGF-induced signals and cell proliferation. CONCLUSIONS IFN-alpha directly targets human hepatocytes, followed by activation of multiple STATs and regulation of a wide variety of genes, which may contribute to the antiviral and antitumor activities of IFN-alpha in human liver.
Collapse
Affiliation(s)
- Svetlana Radaeva
- Section on Liver Biology, Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Type I interferons (IFNs) constitute a family of structurally related proteins that are all derived from the same ancestral gene and act on a common cell-surface receptor. Contrary to many other cytokines, the production of type I IFNs is not a specialized function, and all cells in the organism can produce them, usually as a result of induction by viruses, via the formation of double-stranded RNA. Type I IFNs are indeed responsible for the first line of defense during virus infection and act through the induction of a great number of proteins. Of these, at least thirty have been characterized, and there are probably many more. In addition to their direct antiviral effect, type I IFNs exert a wide variety of other activities, such as for example the induction of various cytokines and the stimulation of different effector cells of the immune system. Due to these pleiotropic effects, recombinant interferons are used in the clinic to treat a variety of diseases, among which cancer, viral hepatitis and multiple sclerosis.
Collapse
Affiliation(s)
- E De Maeyer
- Institut Curie, Université Paris-Sud, Orsay, France
| | | |
Collapse
|
37
|
Abstract
Interferons play key roles in mediating antiviral and antigrowth responses and in modulating immune response. The main signaling pathways are rapid and direct. They involve tyrosine phosphorylation and activation of signal transducers and activators of transcription factors by Janus tyrosine kinases at the cell membrane, followed by release of signal transducers and activators of transcription and their migration to the nucleus, where they induce the expression of the many gene products that determine the responses. Ancillary pathways are also activated by the interferons, but their effects on cell physiology are less clear. The Janus kinases and signal transducers and activators of transcription, and many of the interferon-induced proteins, play important alternative roles in cells, raising interesting questions as to how the responses to the interferons intersect with more general aspects of cellular physiology and how the specificity of cytokine responses is maintained.
Collapse
Affiliation(s)
- G R Stark
- Lerner Research Institute, Cleveland Clinic Foundation, Ohio 44195, USA.
| | | | | | | | | |
Collapse
|
38
|
Chelbi-Alix MK, Quignon F, Pelicano L, Koken MH, de Thé H. Resistance to virus infection conferred by the interferon-induced promyelocytic leukemia protein. J Virol 1998; 72:1043-51. [PMID: 9444998 PMCID: PMC124576 DOI: 10.1128/jvi.72.2.1043-1051.1998] [Citation(s) in RCA: 190] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/1997] [Accepted: 10/31/1997] [Indexed: 02/05/2023] Open
Abstract
The interferon (IFN)-induced promyelocytic leukemia (PML) protein is specifically associated with nuclear bodies (NBs) whose functions are yet unknown. Two of the NB-associated proteins, PML and Sp100, are induced by IFN. Here we show that overexpression of PML and not Sp100 induces resistance to infections by vesicular stomatitis virus (VSV) (a rhabdovirus) and influenza A virus (an orthomyxovirus) but not by encephalomyocarditis virus (a picornavirus). Inhibition of viral multiplication was dependent on both the level of PML expression and the multiplicity of infection and reached 100-fold. PML was shown to interfere with VSV mRNA and protein synthesis. Compared to the IFN mediator MxA protein, PML had less powerful antiviral activity. While nuclear body localization of PML did not seem to be required for the antiviral effect, deletion of the PML coiled-coil domain completely abolished it. Taken together, these results suggest that PML can contribute to the antiviral state induced in IFN-treated cells.
Collapse
Affiliation(s)
- M K Chelbi-Alix
- CNRS UPR 9051, Centre Hayem, Hôpital St. Louis, Paris, France.
| | | | | | | | | |
Collapse
|