1
|
Brunel J, Paganini J, Galloux M, Charvet B, Perron H. HERV-W ENV transcription in B cells predicting symptomatic COVID-19 and risk for long COVID can express a full-length protein despite stop codon in mRNA from chromosome X via a ribosome readthrough. Microbes Infect 2024:105431. [PMID: 39419470 DOI: 10.1016/j.micinf.2024.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024]
Abstract
The human genome comprises 8 % of endogenous retroviruses (HERVs). Though HERVS contribute to physiological functions, copies retained pathogenic potential. The HERV-W ENV protein was shown expressed in patients with worse COVID-19 symptoms and post-COVID syndrome. A significant detection of the mRNA encoding HERV-W ENV from patients with COVID-19 in B cells from RNAseq reads obtained from peripheral blond mononuclear cells. This data stratified with increased COVID-19 symptoms or with post-acute sequelae of COVID-19 (long COVID) after 3 months. The HERV-W ENV-U3R RNA was confirmed to display the best alignment with chromosome X ERVWE2 locus. However, a stop codon precluding its translation was re-addressed after recent understandings of ribosome readthrough mechanisms. Experimental results evidenced that this HERV gene can effectively express a full-length protein in the presence of molecules allowing translation via a readthrough mechanism at the ribosome level. Results not only confirm HERV-W ENV RNA origin in these patients but show for the first time how a defective HERV copy can be translated into a complete protein when specific factors make it possible at the ribosome level. The present proof of concept now requires further studies to identify the factors involved in this newly understood mechanism, following SARS-CoV-2 exposure.
Collapse
Affiliation(s)
- Joanna Brunel
- GeNeuro Innovation, 60A Avenue Rockefeller, 69008, Lyon, France
| | | | | | | | - Hervé Perron
- GeNeuro Innovation, 60A Avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
2
|
Pizzioli E, Minutolo A, Balestrieri E, Matteucci C, Magiorkinis G, Horvat B. Crosstalk between human endogenous retroviruses and exogenous viruses. Microbes Infect 2024:105427. [PMID: 39349096 DOI: 10.1016/j.micinf.2024.105427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral infections of human germ-line cells, which are mostly silenced during evolution, but could be de-repressed and play a pathological role. Infection with some exogenous viruses, including herpesviruses, HIV-1 and SARS-CoV-2, was demonstrated to induce the expression of HERV RNAs and proteins.
Collapse
Affiliation(s)
- Edoardo Pizzioli
- CIRI, Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, France
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Gkikas Magiorkinis
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Branka Horvat
- CIRI, Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, France.
| |
Collapse
|
3
|
Perron H. A tale of a hidden family of genetic immigrants. Microbes Infect 2024:105387. [PMID: 38944111 DOI: 10.1016/j.micinf.2024.105387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/01/2024]
Abstract
Though not usual for the editors of a scientific journal to ask that a story be told to its readers, this special issue is offering an opportunity to pay tribute to all those who have made it possible for a long scientific journey to open up many research avenues, to access the discoveries of what was not known and to the understanding of what was unveiled in the field of human endogenous retroviruses. In particular, and beyond a simple fortuitous association, to show their pathogenic involvement in certain diseases whose causality has been the subject of numerous and variable hypotheses.
Collapse
Affiliation(s)
- Hervé Perron
- GeNeuro, 3 chemin du Pré-Fleuri, 1228 Plan-les-ouates, Geneva, Switzerland; Geneuro-Innovation, 60A, Avenue Rockefeller, 69008 Lyon, France.
| |
Collapse
|
4
|
Gruchot J, Reiche L, Werner L, Herrero F, Schira-Heinen J, Meyer U, Küry P. Molecular dissection of HERV-W dependent microglial- and astroglial cell polarization. Microbes Infect 2024:105382. [PMID: 38944109 DOI: 10.1016/j.micinf.2024.105382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
The endogenous retrovirus type W (HERV-W) is a human-specific entity, which was initially discovered in multiple sclerosis (MS) patient derived cells. We initially found that the HERV-W envelope (ENV) protein negatively affects oligodendrogenesis and controls microglial cell polarization towards a myelinated axon associated and damaging phenotype. Such first functional assessments were conducted ex vivo, given the human-specific origin of HERV-W. Recent experimental evidence gathered on a novel transgenic mouse model, mimicking activation and expression of the HERV-W ENV protein, revealed that all glial cell types are impacted and that cellular fates, differentiation, and functions were changed. In order to identify HERV-W-specific signatures in glial cells, the current study analyzed the transcriptome of ENV protein stimulated microglial- and astroglial cells and compared the transcriptomic signatures to lipopolysaccharide (LPS) stimulated cells, owing to the fact that both ligands can activate toll-like receptor-4 (TLR-4). Additionally, a comparison between published disease associated glial signatures and the transcriptome of HERV-W ENV stimulated glial cells was conducted. We, therefore, provide here for the first time a detailed molecular description of specific HERV-W ENV evoked effects on those glial cell populations that are involved in smoldering neuroinflammatory processes relevant for progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Joel Gruchot
- Heinrich-Heine-University Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Department of Neurology, D-40225 Düsseldorf, Germany
| | - Laura Reiche
- Heinrich-Heine-University Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Department of Neurology, D-40225 Düsseldorf, Germany
| | - Luisa Werner
- Heinrich-Heine-University Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Department of Neurology, D-40225 Düsseldorf, Germany
| | - Felisa Herrero
- Institute of Veterinary Pharmacology and Toxicology, University of Zürich, Vetsuisse, Zürich, Switzerland
| | - Jessica Schira-Heinen
- Heinrich-Heine-University Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Department of Neurology, D-40225 Düsseldorf, Germany
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zürich, Vetsuisse, Zürich, Switzerland; Neuroscience Center Zürich, University of Zürich and ETH Zürich, Zürich, Switzerland
| | - Patrick Küry
- Heinrich-Heine-University Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Department of Neurology, D-40225 Düsseldorf, Germany; Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Adler GL, Le K, Fu Y, Kim WS. Human Endogenous Retroviruses in Neurodegenerative Diseases. Genes (Basel) 2024; 15:745. [PMID: 38927681 PMCID: PMC11202925 DOI: 10.3390/genes15060745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Human endogenous retroviruses (HERVs) are DNA transposable elements that have integrated into the human genome via an ancestral germline infection. The potential importance of HERVs is underscored by the fact that they comprise approximately 8% of the human genome. HERVs have been implicated in the pathogenesis of neurodegenerative diseases, a group of CNS diseases characterized by a progressive loss of structure and function of neurons, resulting in cell death and multiple physiological dysfunctions. Much evidence indicates that HERVs are initiators or drivers of neurodegenerative processes in multiple sclerosis and amyotrophic lateral sclerosis, and clinical trials have been designed to target HERVs. In recent years, the role of HERVs has been explored in other major neurodegenerative diseases, including frontotemporal dementia, Alzheimer's disease and Parkinson's disease, with some interesting discoveries. This review summarizes and evaluates the past and current research on HERVs in neurodegenerative diseases. It discusses the potential role of HERVs in disease manifestation and neurodegeneration. It critically reviews antiretroviral strategies used in the therapeutic intervention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gabrielle L. Adler
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kelvin Le
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - YuHong Fu
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - Woojin Scott Kim
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2050, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
6
|
Gruchot J, Lewen I, Dietrich M, Reiche L, Sindi M, Hecker C, Herrero F, Charvet B, Weber-Stadlbauer U, Hartung HP, Albrecht P, Perron H, Meyer U, Küry P. Transgenic expression of the HERV-W envelope protein leads to polarized glial cell populations and a neurodegenerative environment. Proc Natl Acad Sci U S A 2023; 120:e2308187120. [PMID: 37695891 PMCID: PMC10515160 DOI: 10.1073/pnas.2308187120] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/07/2023] [Indexed: 09/13/2023] Open
Abstract
The human endogenous retrovirus type W (HERV-W) has been identified and repeatedly confirmed as human-specific pathogenic entity affecting many cell types in multiple sclerosis (MS). Our recent contributions revealed the encoded envelope (ENV) protein to disturb myelin repair by interfering with oligodendroglial precursor differentiation and by polarizing microglial cells toward an axon-damage phenotype. Indirect proof of ENV's antiregenerative and degenerative activities has been gathered recently in clinical trials using a neutralizing anti-ENV therapeutic antibody. Yet direct proof of its mode of action can only be presented here based on transgenic ENV expression in mice. Upon demyelination, we observed myelin repair deficits, neurotoxic microglia and astroglia, and increased axon degeneration. Experimental autoimmune encephalomyelitis activity progressed faster in mutant mice equally accompanied by activated glial cells. This study therefore provides direct evidence on HERV-W ENV's contribution to the overall negative impact of this activated viral entity in MS.
Collapse
Affiliation(s)
- Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Isabel Lewen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Mustafa Sindi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | - Felisa Herrero
- Institute of Veterinary Pharmacology and Toxicology, University of Zürich-Vetsuisse, CH-8057Zürich, Switzerland
| | | | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zürich-Vetsuisse, CH-8057Zürich, Switzerland
- Neuroscience Center Zurich, University of Zürich and ETH Zürich, CH-8057Zürich, Switzerland
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
- Brain and Mind Center, University of Sydney, NSW 2050Sydney, Australia
- Department of Neurology, Palacky University Olomouc, 77146Olomouc, Czech Republic
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
| | | | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zürich-Vetsuisse, CH-8057Zürich, Switzerland
- Neuroscience Center Zurich, University of Zürich and ETH Zürich, CH-8057Zürich, Switzerland
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225Düsseldorf, Germany
- Department of Neurology, University of Bern, CH-3010Bern, Switzerland
| |
Collapse
|
7
|
Dawson T, Rentia U, Sanford J, Cruchaga C, Kauwe JSK, Crandall KA. Locus specific endogenous retroviral expression associated with Alzheimer's disease. Front Aging Neurosci 2023; 15:1186470. [PMID: 37484691 PMCID: PMC10359044 DOI: 10.3389/fnagi.2023.1186470] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Human endogenous retroviruses (HERVs) are transcriptionally-active remnants of ancient retroviral infections that may play a role in Alzheimer's disease. Methods We combined two, publicly available RNA-Seq datasets with a third, novel dataset for a total cohort of 103 patients with Alzheimer's disease and 45 healthy controls. We use telescope to perform HERV quantification for these samples and simultaneously perform gene expression analysis. Results We identify differentially expressed genes and differentially expressed HERVs in Alzheimer's disease patients. Differentially expressed HERVs are scattered throughout the genome; many of them are members of the HERV-K superfamily. A number of HERVs are correlated with the expression of dysregulated genes in Alzheimer's and are physically proximal to genes which drive disease pathways. Discussion Dysregulated expression of ancient retroviral insertions in the human genome are present in Alzheimer's disease and show localization patterns that may explain how these elements drive pathogenic gene expression.
Collapse
Affiliation(s)
- Tyson Dawson
- Computational Biology Institute, The George Washington University, Washington, DC, United States
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Uzma Rentia
- Computational Biology Institute, The George Washington University, Washington, DC, United States
| | - Jessie Sanford
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, UT, United States
| | - Keith A. Crandall
- Computational Biology Institute, The George Washington University, Washington, DC, United States
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| |
Collapse
|
8
|
Charvet B, Brunel J, Pierquin J, Iampietro M, Decimo D, Queruel N, Lucas A, Encabo-Berzosa MDM, Arenaz I, Marmolejo TP, Gonzalez AI, Maldonado AC, Mathieu C, Küry P, Flores-Rivera J, Torres-Ruiz F, Avila-Rios S, Salgado Montes de Oca G, Schoorlemmer J, Perron H, Horvat B. SARS-CoV-2 awakens ancient retroviral genes and the expression of proinflammatory HERV-W envelope protein in COVID-19 patients. iScience 2023; 26:106604. [PMID: 37091988 PMCID: PMC10079620 DOI: 10.1016/j.isci.2023.106604] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/29/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Patients with COVID-19 may develop abnormal inflammatory response, followed in some cases by severe disease and long-lasting syndromes. We show here that in vitro exposure to SARS-CoV-2 activates the expression of the human endogenous retrovirus (HERV) HERV-W proinflammatory envelope protein (ENV) in peripheral blood mononuclear cells from a subset of healthy donors, in ACE2 receptor and infection-independent manner. Plasma and/or sera of 221 COVID-19 patients from different cohorts, infected with successive SARS-CoV-2 variants including the Omicron, had detectable HERV-W ENV, which correlated with ENV expression in T lymphocytes and peaked with the disease severity. HERV-W ENV was also found in postmortem tissues of lungs, heart, gastrointestinal tract, brain olfactory bulb, and nasal mucosa from COVID-19 patients. Altogether, these results demonstrate that SARS-CoV-2 could induce HERV-W envelope protein expression and suggest its involvement in the immunopathogenesis of certain COVID-19-associated syndromes and thereby its relevance in the development of personalized treatment of patients.
Collapse
Affiliation(s)
| | | | | | - Mathieu Iampietro
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Didier Decimo
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Alexandre Lucas
- We-Met platform, I2MC/Inserm/Université Paul Sabatier UMR1297, Toulouse, France
| | | | - Izaskun Arenaz
- Biobanco del Sistema de Salud de Aragón, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Tania Perez Marmolejo
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | - Arturo Ivan Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | | | - Cyrille Mathieu
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Dusseldorf, Germany
| | - Jose Flores-Rivera
- Department of Neurology, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Santiago Avila-Rios
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Gonzalo Salgado Montes de Oca
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Jon Schoorlemmer
- ARAID Fundación; Instituto Aragonés de Ciencias de la Salud (IACS); Grupo B46_20R de la DGA and GIIS-028 del IISA; all Zaragoza, Spain
| | - Hervé Perron
- GeNeuro Innovation, Lyon, France
- GeNeuro, Plan les Ouates, Geneva, Switzerland
| | - Branka Horvat
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
9
|
Yushkova E, Moskalev A. Transposable elements and their role in aging. Ageing Res Rev 2023; 86:101881. [PMID: 36773759 DOI: 10.1016/j.arr.2023.101881] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/16/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Transposable elements (TEs) are an important part of eukaryotic genomes. The role of somatic transposition in aging, carcinogenesis, and other age-related diseases has been determined. This review discusses the fundamental properties of TEs and their complex interactions with cellular processes, which are crucial for understanding the diverse effects of their activity on the genetics and epigenetics of the organism. The interactions of TEs with recombination, replication, repair, and chromosomal regulation; the ability of TEs to maintain a balance between their own activity and repression, the involvement of TEs in the creation of new or alternative genes, the expression of coding/non-coding RNA, and the role in DNA damage and modification of regulatory networks are reviewed. The contribution of the derepressed TEs to age-dependent effects in individual cells/tissues in different organisms was assessed. Conflicting information about TE activity under stress as well as theories of aging mechanisms related to TEs is discussed. On the one hand, transposition activity in response to stressors can lead to organisms acquiring adaptive innovations of great importance for evolution at the population level. On the other hand, the TE expression can cause decreased longevity and stress tolerance at the individual level. The specific features of TE effects on aging processes in germline and soma and the ways of their regulation in cells are highlighted. Recent results considering somatic mutations in normal human and animal tissues are indicated, with the emphasis on their possible functional consequences. In the context of aging, the correlation between somatic TE activation and age-related changes in the number of proteins required for heterochromatin maintenance and longevity regulation was analyzed. One of the original features of this review is a discussion of not only effects based on the TEs insertions and the associated consequences for the germline cell dynamics and somatic genome, but also the differences between transposon- and retrotransposon-mediated structural genome changes and possible phenotypic characteristics associated with aging and various age-related pathologies. Based on the analysis of published data, a hypothesis about the influence of the species-specific features of number, composition, and distribution of TEs on aging dynamics of different animal genomes was formulated.
Collapse
Affiliation(s)
- Elena Yushkova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russian Federation
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st., 167982 Syktyvkar, Russian Federation; Laboratory of Genetics and Epigenetics of Aging, Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Moscow 129226, Russian Federation; Longaevus Technologies, London, UK.
| |
Collapse
|
10
|
Tovo PA, Marozio L, Abbona G, Calvi C, Frezet F, Gambarino S, Dini M, Benedetto C, Galliano I, Bergallo M. Pregnancy Is Associated with Impaired Transcription of Human Endogenous Retroviruses and of TRIM28 and SETDB1, Particularly in Mothers Affected by Multiple Sclerosis. Viruses 2023; 15:v15030710. [PMID: 36992419 PMCID: PMC10051116 DOI: 10.3390/v15030710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Accumulating evidence highlights the pathogenetic role of human endogenous retroviruses (HERVs) in eliciting and maintaining multiple sclerosis (MS). Epigenetic mechanisms, such as those regulated by TRIM 28 and SETDB1, are implicated in HERV activation and in neuroinflammatory disorders, including MS. Pregnancy markedly improves the course of MS, but no study explored the expressions of HERVs and of TRIM28 and SETDB1 during gestation. Using a polymerase chain reaction real-time Taqman amplification assay, we assessed and compared the transcriptional levels of pol genes of HERV-H, HERV-K, HERV-W; of env genes of Syncytin (SYN)1, SYN2, and multiple sclerosis associated retrovirus (MSRV); and of TRIM28 and SETDB1 in peripheral blood and placenta from 20 mothers affected by MS; from 27 healthy mothers, in cord blood from their neonates; and in blood from healthy women of child-bearing age. The HERV mRNA levels were significantly lower in pregnant than in nonpregnant women. Expressions of all HERVs were downregulated in the chorion and in the decidua basalis of MS mothers compared to healthy mothers. The former also showed lower mRNA levels of HERV-K-pol and of SYN1, SYN2, and MSRV in peripheral blood. Significantly lower expressions of TRIM28 and SETDB1 also emerged in pregnant vs. nonpregnant women and in blood, chorion, and decidua of mothers with MS vs. healthy mothers. In contrast, HERV and TRIM28/SETDB1 expressions were comparable between their neonates. These results show that gestation is characterized by impaired expressions of HERVs and TRIM28/SETDB1, particularly in mothers with MS. Given the beneficial effects of pregnancy on MS and the wealth of data suggesting the putative contribution of HERVs and epigenetic processes in the pathogenesis of the disease, our findings may further support innovative therapeutic interventions to block HERV activation and to control aberrant epigenetic pathways in MS-affected patients.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Correspondence: (P.-A.T.); (M.B.)
| | - Luca Marozio
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Giancarlo Abbona
- Pathology Unit, Department Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Cristina Calvi
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Federica Frezet
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Stefano Gambarino
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Maddalena Dini
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Chiara Benedetto
- Department of Surgical Sciences, Obstetrics and Gynecology 1, University of Turin, 10126 Turin, Italy
| | - Ilaria Galliano
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Massimiliano Bergallo
- Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
- Correspondence: (P.-A.T.); (M.B.)
| |
Collapse
|
11
|
Interplay between activation of endogenous retroviruses and inflammation as common pathogenic mechanism in neurological and psychiatric disorders. Brain Behav Immun 2023; 107:242-252. [PMID: 36270439 DOI: 10.1016/j.bbi.2022.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/21/2022] [Accepted: 10/13/2022] [Indexed: 12/05/2022] Open
Abstract
Human endogenous retroviruses (ERVs) are ancestorial retroviral elements that were integrated into our genome through germline infections and insertions during evolution. They have repeatedly been implicated in the aetiology and pathophysiology of numerous human disorders, particularly in those that affect the central nervous system. In addition to the known association of ERVs with multiple sclerosis and amyotrophic lateral sclerosis, a growing number of studies links the induction and expression of these retroviral elements with the onset and severity of neurodevelopmental and psychiatric disorders. Although these disorders differ in terms of overall disease pathology and causalities, a certain degree of (subclinical) chronic inflammation can be identified in all of them. Based on these commonalities, we discuss the bidirectional relationship between ERV expression and inflammation and highlight that numerous entry points to this reciprocal sequence of events exist, including initial infections with ERV-activating pathogens, exposure to non-infectious inflammatory stimuli, and conditions in which epigenetic silencing of ERV elements is disrupted.
Collapse
|
12
|
Rangel SC, da Silva MD, da Silva AL, dos Santos JDMB, Neves LM, Pedrosa A, Rodrigues FM, Trettel CDS, Furtado GE, de Barros MP, Bachi ALL, Romano CM, Nali LHDS. Human endogenous retroviruses and the inflammatory response: A vicious circle associated with health and illness. Front Immunol 2022; 13:1057791. [PMID: 36518758 PMCID: PMC9744114 DOI: 10.3389/fimmu.2022.1057791] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Human Endogenous Retroviruses (HERVs) are derived from ancient exogenous retroviral infections that have infected our ancestors' germline cells, underwent endogenization process, and were passed throughout the generations by retrotransposition and hereditary transmission. HERVs comprise 8% of the human genome and are critical for several physiological activities. Yet, HERVs reactivation is involved in pathological process as cancer and autoimmune diseases. In this review, we summarize the multiple aspects of HERVs' role within the human genome, as well as virological and molecular aspects, and their fusogenic property. We also discuss possibilities of how the HERVs are possibly transactivated and participate in modulating the inflammatory response in health conditions. An update on their role in several autoimmune, inflammatory, and aging-related diseases is also presented.
Collapse
Affiliation(s)
- Sara Coelho Rangel
- UNISA Research Center, Universidade Santo Amaro, Post-Graduation in Health Sciences, São Paulo, Brazil
| | | | - Amanda Lopes da Silva
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | | | - Lucas Melo Neves
- UNISA Research Center, Universidade Santo Amaro, Post-Graduation in Health Sciences, São Paulo, Brazil
| | - Ana Pedrosa
- CNC-Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, (3004-504), Coimbra, Portugal
| | | | - Caio dos Santos Trettel
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo, Brazil
| | - Guilherme Eustáquio Furtado
- Polytechnic Institute of Coimbra, Applied Research Institute, Rua da Misericórdia, Lagar dos Cortiços – S. Martinho do Bispo, Coimbra, Portugal
| | - Marcelo Paes de Barros
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo, Brazil
| | - André Luis Lacerda Bachi
- UNISA Research Center, Universidade Santo Amaro, Post-Graduation in Health Sciences, São Paulo, Brazil
| | - Camila Malta Romano
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, Universidade de São Paulo, São Paulo, Brazil
- Hospital das Clínicas HCFMUSP (LIM52), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
13
|
Beyond fusion: A novel role for ERVW-1 in trophoblast proliferation and type I interferon receptor expression. Placenta 2022; 126:150-159. [PMID: 35816776 DOI: 10.1016/j.placenta.2022.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/23/2022] [Accepted: 06/26/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Throughout human pregnancy there is a delicate balance between the maintenance of a proliferative, trophoblast stem cell pool (TSC) and the differentiation from TSC to placental cell sub-lineages like the syncytiotrophoblast (STB). The STB is comprised of multinucleated cells that come into direct contact with maternal blood and provides the first line of defense to protect the fetus from maternal infections. The differentiation of TSC towards STB is primarily driven by human endogenous retroviruses (HERV), specifically Syncytin-1 (ERVW-1) and Syncytin-2 (ERVFRD-1). Beyond cell fusion, there is also evidence to suggest they can regulate cell proliferation and an antiviral response in other cell types. Therefore, we hypothesized that HERV can regulate cell proliferation as well as an antiviral response in TSCs. METHOD shRNA was used to knockdown ERVW-1 in TSCs and revealed reduction in cell proliferation, differentiation, and cell fusion. RT-qPCR and flow cytometry was used to measure other HERV and the presence of Type I and Type II interferon receptors. RESULTS ERVW-1 knockdown (KD) TSCs had a significantly longer cell doubling time and reduced expression of the proliferation marker Ki67. ERVW-1 KD cells also demonstrated a marked deficiency in the ability to differentiate. Interestingly, ERVFRD-1 was upregulated in both ERVW-1 KD TSC and STB cells compared to controls. Finally, we found that the Type I interferon receptors, IFNAR1 and IFNAR2 were significantly increased in ERVW-1 KD STB cells. DISCUSSION These findings uncover critical HERV functions in the trophoblasts and a novel role for ERVW-1 during early human placental development.
Collapse
|
14
|
DeRosa H, Richter T, Wilkinson C, Hunter RG. Bridging the Gap Between Environmental Adversity and Neuropsychiatric Disorders: The Role of Transposable Elements. Front Genet 2022; 13:813510. [PMID: 35711940 PMCID: PMC9196244 DOI: 10.3389/fgene.2022.813510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/13/2022] [Indexed: 12/21/2022] Open
Abstract
Long regarded as “junk DNA,” transposable elements (TEs) have recently garnered much attention for their role in promoting genetic diversity and plasticity. While many processes involved in mammalian development require TE activity, deleterious TE insertions are a hallmark of several psychiatric disorders. Moreover, stressful events including exposure to gestational infection and trauma, are major risk factors for developing psychiatric illnesses. Here, we will provide evidence demonstrating the intersection of stressful events, atypical TE expression, and their epigenetic regulation, which may explain how neuropsychiatric phenotypes manifest. In this way, TEs may be the “bridge” between environmental perturbations and psychopathology.
Collapse
Affiliation(s)
- Holly DeRosa
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Troy Richter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Cooper Wilkinson
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Richard G Hunter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
15
|
Hamdy A, Leonardi A. Superantigens and SARS-CoV-2. Pathogens 2022; 11:390. [PMID: 35456065 PMCID: PMC9026686 DOI: 10.3390/pathogens11040390] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 12/31/2022] Open
Abstract
It has been posited SARS-CoV-2 contains at least one unique superantigen-like motif not found in any other SARS or endemic coronaviruses. Superantigens are potent antigens that can send the immune system into overdrive. SARS-CoV-2 causes many of the biological and clinical consequences of a superantigen, and, in the context of reinfection and waning immunity, it is important to better understand the impact of a widely circulating, airborne pathogen that may be a superantigen, superantigen-like or trigger a superantigenic host response. Urgent research is needed to better understand the long-term risks being taken by governments whose policies enable widespread transmission of a potential superantigenic pathogen, and to more clearly define the vaccination and public health policies needed to protect against the consequences of repeat exposure to the pathogen.
Collapse
Affiliation(s)
- Adam Hamdy
- Panres Pandemic Research, Newport TF10 8PG, UK
| | - Anthony Leonardi
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| |
Collapse
|
16
|
Giovannoni G, Popescu V, Wuerfel J, Hellwig K, Iacobaeus E, Jensen MB, García-Domínguez JM, Sousa L, De Rossi N, Hupperts R, Fenu G, Bodini B, Kuusisto HM, Stankoff B, Lycke J, Airas L, Granziera C, Scalfari A. Smouldering multiple sclerosis: the 'real MS'. Ther Adv Neurol Disord 2022; 15:17562864211066751. [PMID: 35096143 PMCID: PMC8793117 DOI: 10.1177/17562864211066751] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/28/2021] [Indexed: 12/25/2022] Open
Abstract
Using a philosophical approach or deductive reasoning, we challenge the dominant clinico-radiological worldview that defines multiple sclerosis (MS) as a focal inflammatory disease of the central nervous system (CNS). We provide a range of evidence to argue that the 'real MS' is in fact driven primarily by a smouldering pathological disease process. In natural history studies and clinical trials, relapses and focal activity revealed by magnetic resonance imaging (MRI) in MS patients on placebo or on disease-modifying therapies (DMTs) were found to be poor predictors of long-term disease evolution and were dissociated from disability outcomes. In addition, the progressive accumulation of disability in MS can occur independently of relapse activity from early in the disease course. This scenario is underpinned by a more diffuse smouldering pathological process that may affect the entire CNS. Many putative pathological drivers of smouldering MS can be potentially modified by specific therapeutic strategies, an approach that may have major implications for the management of MS patients. We hypothesise that therapeutically targeting a state of 'no evident inflammatory disease activity' (NEIDA) cannot sufficiently prevent disability accumulation in MS, meaning that treatment should also focus on other brain and spinal cord pathological processes contributing to the slow loss of neurological function. This should also be complemented with a holistic approach to the management of other systemic disease processes that have been shown to worsen MS outcomes.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark St., Whitechapel, London E1 2AT, UK
| | - Veronica Popescu
- Universitair MS Centrum, Hasselt, Belgium; Noorderhart Hospital, Pelt, Belgium; Hasselt University, Hasselt, Belgium
| | - Jens Wuerfel
- MIAC AG, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Charité - University Medicine Berlin, Berlin, Germany
| | - Kerstin Hellwig
- Katholisches Klinikum Bochum, Klinikum der Ruhr-Universität, Bochum, Germany
| | | | - Michael B Jensen
- Department of Neurology, Nordsjællands Hospital, Hillerød, Denmark
| | | | - Livia Sousa
- Centro Hospitalar e Universitário de Coimbra, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | | | - Raymond Hupperts
- Zuyderland Medisch Centrum, Sittard-Geleen, The Netherlands; Maastricht University Medical Center, Maastricht, The Netherlands
| | - Giuseppe Fenu
- Department of Neurology, Brotzu Hospital, Cagliari, Italy
| | - Benedetta Bodini
- Paris Brain Institute, Sorbonne University, Paris, France; Department of Neurology, APHP, Saint-Antoine Hospital, Paris, France
| | - Hanna-Maija Kuusisto
- Department of Neurology, Tampere University Hospital, Tampere, Finland; Department of Customer and Patient Safety, University of Eastern Finland, Kuopio, Finland
| | - Bruno Stankoff
- Paris Brain Institute, Sorbonne University, ICM, CNRS, Inserm, Paris, France; APHP, Saint-Antoine Hospital, Paris, France
| | - Jan Lycke
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | | | - Cristina Granziera
- Neurologic Clinic and Policlinic, Departments of Medicine, Clinical Research and Biomedical Engineering, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Antonio Scalfari
- Centre for Neuroscience, Department of Medicine, Charing Cross Hospital, Imperial College London, London, UK
| |
Collapse
|
17
|
Latifi T, Zebardast A, Marashi SM. The role of human endogenous retroviruses (HERVs) in Multiple Sclerosis and the plausible interplay between HERVs, Epstein-Barr virus infection, and vitamin D. Mult Scler Relat Disord 2022; 57:103318. [PMID: 35158423 DOI: 10.1016/j.msard.2021.103318] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 12/30/2022]
Abstract
Multiple Sclerosis (MS) is one of the chronic inflammatory diseases with neurological disability in the central nervous system (CNS). Although the exact cause of MS is still largely unknown, both genetic and environmental factors are thought to play a role in disease risk. Human Endogenous Retroviruses (HERVs) are endogenous viral elements of the human genome whose expression is associated with MS. HERVs are normally silenced or expressed at low levels, although their expression is higher in MS than in the healthy population. Several studies highlighted the plausible interaction between HERVs and other MS risk factors, including viral infection like Epstein-Barr viruses and vitamin D deficiency which may lead to high expression of HERVs in these patients. Understanding how HERVs act in this scenario can improve our understanding towards MS etiology and may lead to the development of antiretroviral therapies in these patients. Here in this review, we try to examine the different HERVs expression implicated in MS and their association with EBV infection and vitamin D status.
Collapse
Affiliation(s)
- Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arghavan Zebardast
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Nali LH, Olival GS, Montenegro H, da Silva IT, Dias-Neto E, Naya H, Spangenberg L, Penalva-de-Oliveira AC, Romano CM. Human endogenous retrovirus and multiple sclerosis: A review and transcriptome findings. Mult Scler Relat Disord 2021; 57:103383. [PMID: 34922254 DOI: 10.1016/j.msard.2021.103383] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/15/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022]
Abstract
Multiple Sclerosis is an autoimmune disease with an unknown etiology. Both genetic and environmental factors are believed to trigger MS autoimmunity. Among the environmental factors, infectious agents have been extensively investigated, and the Human Endogenous Retroviruses (HERVs), especially HERV-W, are believed to be associated with MS pathogenesis. HERVs are derived from ancestral infections and comprise around 8% of the human genome. Although most HERVs are silenced, retroviral genes may be expressed with virion formation. There is extensive evidence of the relationship between HERV-W and MS, including higher levels of HERV-W expression in MS patients, HERV-W protein detection in MS plaques, and the HERV-W env protein inducing an inflammatory response in in vitro and in vivo models. Here we discuss possible links of HERVs and the pathogenesis of MS and present new data regarding the diversity of HERVs expression in samples derived from MS patients.
Collapse
Affiliation(s)
- Luiz H Nali
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo 05403-000, Brazil; Post-graduation Program in Health Sciences, Santo Amaro University, Rua Prof. Enéas de Siqueira Neto, 340, São Paulo 04829-300, Brazil
| | - Guilherme S Olival
- Departamento de Neurologia Santa Casa de Misericórdia de São Paulo, R. Dr. Cesário Mota Júnior, 112, São Paulo 01221-020 Brazil
| | | | - Israel T da Silva
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01525-001, Brazil
| | - Emmanuel Dias-Neto
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01525-001, Brazil; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, São Paulo Medical School, University of São Paulo, São Paulo, Brazil
| | - Hugo Naya
- Unidad de Bioinformática Institut Pasteur de Montevideo, Mataojo 2020, CP11400 Montevideo, Uruguay; Departamento de Producción Animal y Pasturas, Facultad de Agronomía, Universidad de la República, Av. Gral. Eugenio Garzón 780, CP12900 Montevideo, Uruguay
| | - Lucia Spangenberg
- Unidad de Bioinformática Institut Pasteur de Montevideo, Mataojo 2020, CP11400 Montevideo, Uruguay
| | - Augusto C Penalva-de-Oliveira
- Departamento de Neurologia Santa Casa de Misericórdia de São Paulo, R. Dr. Cesário Mota Júnior, 112, São Paulo 01221-020 Brazil; Departamento de Neurologia, Instituto de Infectologia Emilio Ribas, Avenida Doutor Arnaldo, 165, São Paulo 01246-900, Brazil
| | - Camila M Romano
- Laboratório de Virologia, Instituto de Medicina Tropical de São Paulo, LIM-52 (LIMHC) Universidade de São Paulo, Rua Dr. Enéas de Carvalho Aguiar, 470, São Paulo 05403-000, Brazil; Hospital das Clinicas HCFMUSP (LIM52), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
19
|
Wang C, Milgate AW, Solomon PS, McDonald MC. The identification of a transposon affecting the asexual reproduction of the wheat pathogen Zymoseptoria tritici. MOLECULAR PLANT PATHOLOGY 2021; 22:800-816. [PMID: 33949756 PMCID: PMC8232023 DOI: 10.1111/mpp.13064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 05/13/2023]
Abstract
Zymoseptoria tritici, the causal agent of Septoria tritici blotch, is a fungal wheat pathogen that causes significant global yield losses. Within Z. tritici populations, quantitative differences in virulence among different isolates are commonly observed; however, the genetic components that underpin these differences remain elusive. In this study, intraspecific comparative transcriptomic analysis was used to identify candidate genes that contribute to differences in virulence on the wheat cultivar WW2449. This led to the identification of a multicopy gene that was not expressed in the high-virulence isolate when compared to the medium- and low-virulence isolates. Further investigation suggested this gene resides in a 7.9-kb transposon. Subsequent long-read sequencing of the isolates used in the transcriptomic analysis confirmed that this gene did reside in an active Class II transposon, which is composed of four genes named REP9-1 to -4. Silencing and overexpression of REP9-1 in two distinct genetic backgrounds demonstrated that its expression alone reduces the number of pycnidia produced by Z. tritici during infection. The REP9-1 gene identified within a Class II transposon is the first discovery of a gene in a transposable element that influences the virulence of Z. tritici. This discovery adds further complexity to genetic loci that contribute to quantitative virulence in this important pathogen.
Collapse
Affiliation(s)
- Chen Wang
- Division of Plant SciencesResearch School of BiologyThe Australian National UniversityCanberraACTAustralia
| | - Andrew W. Milgate
- NSW Department of Primary IndustriesWagga Wagga Agricultural InstituteWagga WaggaNSWAustralia
| | - Peter S. Solomon
- Division of Plant SciencesResearch School of BiologyThe Australian National UniversityCanberraACTAustralia
| | - Megan C. McDonald
- Division of Plant SciencesResearch School of BiologyThe Australian National UniversityCanberraACTAustralia
- School of BiosciencesUniversity of BirminghamEdgbastonBirminghamUK
| |
Collapse
|
20
|
Bello-Morales R, Andreu S, Ripa I, López-Guerrero JA. HSV-1 and Endogenous Retroviruses as Risk Factors in Demyelination. Int J Mol Sci 2021; 22:ijms22115738. [PMID: 34072259 PMCID: PMC8199333 DOI: 10.3390/ijms22115738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/19/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a neurotropic alphaherpesvirus that can infect the peripheral and central nervous systems, and it has been implicated in demyelinating and neurodegenerative processes. Transposable elements (TEs) are DNA sequences that can move from one genomic location to another. TEs have been linked to several diseases affecting the central nervous system (CNS), including multiple sclerosis (MS), a demyelinating disease of unknown etiology influenced by genetic and environmental factors. Exogenous viral transactivators may activate certain retrotransposons or class I TEs. In this context, several herpesviruses have been linked to MS, and one of them, HSV-1, might act as a risk factor by mediating processes such as molecular mimicry, remyelination, and activity of endogenous retroviruses (ERVs). Several herpesviruses have been involved in the regulation of human ERVs (HERVs), and HSV-1 in particular can modulate HERVs in cells involved in MS pathogenesis. This review exposes current knowledge about the relationship between HSV-1 and human ERVs, focusing on their contribution as a risk factor for MS.
Collapse
Affiliation(s)
- Raquel Bello-Morales
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (S.A.); (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
- Correspondence:
| | - Sabina Andreu
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (S.A.); (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| | - Inés Ripa
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (S.A.); (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| | - José Antonio López-Guerrero
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain; (S.A.); (I.R.); (J.A.L.-G.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
21
|
Giménez-Orenga K, Oltra E. Human Endogenous Retrovirus as Therapeutic Targets in Neurologic Disease. Pharmaceuticals (Basel) 2021; 14:495. [PMID: 34073730 PMCID: PMC8225122 DOI: 10.3390/ph14060495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 01/16/2023] Open
Abstract
Human endogenous retroviruses (HERVs) are ancient retroviral DNA sequences established into germline. They contain regulatory elements and encoded proteins few of which may provide benefits to hosts when co-opted as cellular genes. Their tight regulation is mainly achieved by epigenetic mechanisms, which can be altered by environmental factors, e.g., viral infections, leading to HERV activation. The aberrant expression of HERVs associates with neurological diseases, such as multiple sclerosis (MS) or amyotrophic lateral sclerosis (ALS), inflammatory processes and neurodegeneration. This review summarizes the recent advances on the epigenetic mechanisms controlling HERV expression and the pathogenic effects triggered by HERV de-repression. This article ends by describing new, promising therapies, targeting HERV elements, one of which, temelimab, has completed phase II trials with encouraging results in treating MS. The information gathered here may turn helpful in the design of new strategies to unveil epigenetic failures behind HERV-triggered diseases, opening new possibilities for druggable targets and/or for extending the use of temelimab to treat other associated diseases.
Collapse
Affiliation(s)
- Karen Giménez-Orenga
- Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Elisa Oltra
- School of Medicine and Health Sciences, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain
| |
Collapse
|
22
|
Wieland L, Engel K, Volkmer I, Krüger A, Posern G, Kornhuber ME, Staege MS, Emmer A. Overexpression of Endogenous Retroviruses and Malignancy Markers in Neuroblastoma Cell Lines by Medium-Induced Microenvironmental Changes. Front Oncol 2021; 11:637522. [PMID: 34026614 PMCID: PMC8138558 DOI: 10.3389/fonc.2021.637522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma (NB) is the commonest solid tumor outside the central nervous system in infancy and childhood with a unique biological heterogeneity. In patients with advanced, metastasizing neuroblastoma, treatment failure and poor prognosis is often marked by resistance to chemo- or immunotherapy. Thus, identification of robust biomarkers seems essential for understanding tumor progression and developing effective therapy. Here, we have studied the expression of human endogenous retroviruses (HERV) as potential targets in NB cell lines during stem-cell medium-induced microenvironmental change. Quantitative PCR revealed that relative expression of the HERV-K family and HERV-W1 ENV were increased in all three NB cell lines after incubation in stem-cell medium. Virus transcriptome analyses revealed the transcriptional activation of three endogenous retrovirus elements: HERV-R ENV (ERV3-1), HERV-E1 and HERV-Fc2 ENV (ERVFC1-1). Known malignancy markers in NB, e.g. proto-oncogenic MYC or MYCN were expressed highly heterogeneously in the three investigated NB cell lines with up-regulation of MYC and MYCN upon medium-induced microenvironmental change. In addition, SiMa cells exclusively showed a phenotype switching from loosely-adherent monolayers to low proliferating grape-like cellular aggregates, which was accompanied by an enhanced CD133 expression. Interestingly, the overexpression of HERV was associated with a significant elevation of immune checkpoint molecule CD200 in both quantitative PCR and RNA-seq analysis suggesting tumor escape mechanism in NB cell lines after incubation in serum-free stem cell medium.
Collapse
Affiliation(s)
- Lisa Wieland
- Department of Neurology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany.,Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Kristina Engel
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ines Volkmer
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Anna Krüger
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Malte E Kornhuber
- Department of Neurology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Martin S Staege
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Alexander Emmer
- Department of Neurology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
23
|
Kristensen MK, Christensen T. Regulation of the expression of human endogenous retroviruses: elements in fetal development and a possible role in the development of cancer and neurological diseases. APMIS 2021; 129:241-253. [PMID: 33683784 DOI: 10.1111/apm.13130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Human endogenous retroviruses (HERVs) are remnants of ancient retroviral germline infections. Most HERV sequences are silenced in somatic cells, but interest is emerging on the involvement of HERV derived transcripts and proteins in human physiology and disease. A HERV-W encoded protein, syncytin-1, has been co-opted into fetal physiology, where it plays a role in trophoblast formation. Altered HERV transcription and expression of HERV derived proteins are associated with various cancer types and neurological diseases such as multiple sclerosis (MS). The implication of HERVs as potential mediators of both health and disease suggests important roles of regulatory mechanisms and alterations of these in physiological and pathological processes. The regulation of HERV sequences is mediated by a wide variety of mechanisms, and the focus of this review is on selected aspects of these, including epigenetic mechanisms such as CpG methylation and histone modifications of the HP1-H3K9me axis, viral transactivation events, and regulatory perspectives of transient stimuli in the microenvironment. Increasing knowledge of the regulation of HERV sequences will not only contribute to the understanding of complex pathogeneses, but also may pinpoint potential targets for better diagnosis and treatment in complex diseases as MS.
Collapse
|
24
|
Glinsky GV. Genomics-Guided Drawing of Molecular and Pathophysiological Components of Malignant Regulatory Signatures Reveals a Pivotal Role in Human Diseases of Stem Cell-Associated Retroviral Sequences and Functionally-Active hESC Enhancers. Front Oncol 2021; 11:638363. [PMID: 33869024 PMCID: PMC8044830 DOI: 10.3389/fonc.2021.638363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/10/2021] [Indexed: 12/31/2022] Open
Abstract
Repetitive DNA sequences (repeats) colonized two-third of human genome and a majority of repeats comprised of transposable genetic elements (TE). Evolutionary distinct categories of TE represent nucleic acid sequences that are repeatedly copied from and pasted into chromosomes at multiple genomic locations and acquired a multitude of regulatory functions. Here, genomics-guided maps of stemness regulatory signatures were drawn to dissect the contribution of TE to clinical manifestations of malignant phenotypes of human cancers. From patients’ and physicians’ perspectives, the clinical definition of a tumor’s malignant phenotype could be restricted to the early diagnosis of sub-types of malignancies with the increased risk of existing therapy failure and high likelihood of death from cancer. It is the viewpoint from which the understanding of stemness and malignant regulatory signatures is considered in this contribution. Genomics-guided analyses of experimental and clinical observations revealed the pivotal role of human stem cell-associated retroviral sequences (SCARS) in the origin and pathophysiology of clinically-lethal malignancies. SCARS were defined as the evolutionary- and biologically-related family of genomic regulatory sequences, the principal physiological function of which is to create and maintain the stemness phenotype during human preimplantation embryogenesis. For cell differentiation to occur, SCARS expression must be silenced and SCARS activity remains repressed in most terminally-differentiated human cells which are destined to perform specialized functions in the human body. Epigenetic reprogramming, de-repression, and sustained activity of SCARS results in various differentiation-defective phenotypes. One of the most prominent tissue- and organ-specific clinical manifestations of sustained SCARS activities is diagnosed as a pathological condition defined by a consensus of morphological, molecular, and genetic examinations as the malignant growth. Here, contemporary evidence are acquired, analyzed, and reported defining both novel diagnostic tools and druggable molecular targets readily amenable for diagnosis and efficient therapeutic management of clinically-lethal malignancies. These diagnostic and therapeutic approaches are based on monitoring of high-fidelity molecular signals of continuing SCARS activities in conjunction with genomic regulatory networks of thousands’ functionally-active embryonic enhancers affecting down-stream phenotype-altering genetic loci. Collectively, reported herein observations support a model of SCARS-activation triggered singular source code facilitating the intracellular propagation and intercellular (systemic) dissemination of disease states in the human body.
Collapse
Affiliation(s)
- Gennadi V Glinsky
- Institute of Engineering in Medicine, University of California, San Diego, CA, United States.,Department of Functional & Translational Genomics, OncoSCAR, Inc., Portland, OR, United States
| |
Collapse
|
25
|
Charvet B, Pierquin J, Brunel J, Gorter R, Quétard C, Horvat B, Amor S, Portoukalian J, Perron H. Human Endogenous Retrovirus Type W Envelope from Multiple Sclerosis Demyelinating Lesions Shows Unique Solubility and Antigenic Characteristics. Virol Sin 2021; 36:1006-1026. [PMID: 33770381 PMCID: PMC8558138 DOI: 10.1007/s12250-021-00372-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
In multiple sclerosis (MS), human endogenous retrovirus W family (HERV-W) envelope protein, pHERV-W ENV, limits remyelination and induces microglia-mediated neurodegeneration. To better understand its role, we examined the soluble pHERV-W antigen from MS brain lesions detected by specific antibodies. Physico-chemical and antigenic characteristics confirmed differences between pHERV-W ENV and syncytin-1. pHERV-W ENV monomers and trimers remained associated with membranes, while hexamers self-assembled from monomers into a soluble macrostructure involving sulfatides in MS brain. Extracellular hexamers are stabilized by internal hydrophobic bonds and external hydrophilic moieties. HERV-W studies in MS also suggest that this diffusible antigen may correspond to a previously described high-molecular-weight neurotoxic factor secreted by MS B-cells and thus represents a major agonist in MS pathogenesis. Adapted methods are now needed to identify encoding HERV provirus(es) in affected cells DNA. The properties and origin of MS brain pHERV-W ENV soluble antigen will allow a better understanding of the role of HERVs in MS pathogenesis. The present results anyhow pave the way to an accurate detection of the different forms of pHERV-W ENV antigen with appropriate conditions that remained unseen until now.
Collapse
Affiliation(s)
- Benjamin Charvet
- GeNeuro Innovation, Lyon, 69008, France. .,CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France. .,Université Claude Bernard Lyon 1, Lyon, 69000, France.
| | | | - Joanna Brunel
- GeNeuro Innovation, Lyon, 69008, France.,CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France.,Université Claude Bernard Lyon 1, Lyon, 69000, France
| | - Rianne Gorter
- Department of Pathology, Amsterdam UMC, Location VUMC, 1007 MB, Amsterdam, The Netherlands
| | | | - Branka Horvat
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, University of Lyon, ENS Lyon, France.,Université Claude Bernard Lyon 1, Lyon, 69000, France
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Location VUMC, 1007 MB, Amsterdam, The Netherlands.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | | | - Hervé Perron
- GeNeuro Innovation, Lyon, 69008, France. .,Université Claude Bernard Lyon 1, Lyon, 69000, France.
| |
Collapse
|
26
|
Römer C. Viruses and Endogenous Retroviruses as Roots for Neuroinflammation and Neurodegenerative Diseases. Front Neurosci 2021; 15:648629. [PMID: 33776642 PMCID: PMC7994506 DOI: 10.3389/fnins.2021.648629] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Many neurodegenerative diseases are associated with chronic inflammation in the brain and periphery giving rise to a continuous imbalance of immune processes. Next to inflammation markers, activation of transposable elements, including long intrespersed nuclear elements (LINE) elements and endogenous retroviruses (ERVs), has been identified during neurodegenerative disease progression and even correlated with the clinical severity of the disease. ERVs are remnants of viral infections in the human genome acquired during evolution. Upon activation, they produce transcripts and the phylogenetically youngest ones are still able to produce viral-like particles. In addition, ERVs can bind transcription factors and modulate immune response. Being between own and foreign, ERVs are reviewed in the context of viral infections of the central nervous system, in aging and neurodegenerative diseases. Moreover, this review tests the hypothesis that viral infection may be a trigger at the onset of neuroinflammation and that ERVs sustain the inflammatory imbalance by summarizing existing data of neurodegenerative diseases associated with viruses and/or ERVs.
Collapse
Affiliation(s)
- Christine Römer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, The Berlin Institute for Medical Systems Biology, Berlin, Germany
| |
Collapse
|
27
|
Gröger V, Emmer A, Staege MS, Cynis H. Endogenous Retroviruses in Nervous System Disorders. Pharmaceuticals (Basel) 2021; 14:ph14010070. [PMID: 33467098 PMCID: PMC7829834 DOI: 10.3390/ph14010070] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Human endogenous retroviruses (HERV) have been implicated in the pathogenesis of several nervous system disorders including multiple sclerosis and amyotrophic lateral sclerosis. The toxicity of HERV-derived RNAs and proteins for neuronal cells has been demonstrated. The involvement of HERV in the pathogenesis of currently incurable diseases might offer new treatment strategies based on the inhibition of HERV activities by small molecules or therapeutic antibodies.
Collapse
Affiliation(s)
- Victoria Gröger
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany;
| | - Alexander Emmer
- Department of Neurology, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Martin S. Staege
- Department of Surgical and Conservative Pediatrics and Adolescent Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Correspondence: (M.S.S.); (H.C.); Tel.: +49-345-557-7280 (M.S.S.); +49-345-13142835 (H.C.)
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120 Halle (Saale), Germany;
- Correspondence: (M.S.S.); (H.C.); Tel.: +49-345-557-7280 (M.S.S.); +49-345-13142835 (H.C.)
| |
Collapse
|
28
|
Dechaumes A, Bertin A, Sane F, Levet S, Varghese J, Charvet B, Gmyr V, Kerr-Conte J, Pierquin J, Arunkumar G, Pattou F, Perron H, Hober D. Coxsackievirus-B4 Infection Can Induce the Expression of Human Endogenous Retrovirus W in Primary Cells. Microorganisms 2020; 8:E1335. [PMID: 32883004 PMCID: PMC7563422 DOI: 10.3390/microorganisms8091335] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Human Endogenous Retrovirus W Envelope (HERV-W ENV) mRNA or protein can be found in peripheral blood mononuclear cells (PBMCs) and exocrine pancreas of patients with type 1 diabetes (T1D). Further, previous observations have shown an association between enteroviral infection and development of T1D; specifically, coxsackievirus-B (CV-B) has been detected in the blood and pancreas of patients with T1D. Notably, viruses can activate HERV-W expression. Hence, we evaluated the effect of CV-B4 infection on HERV-W ENV mRNA expression. Primary human pancreatic ductal cells were obtained from five brain-dead donors. In the pancreatic cells of three donors, the HERV-W ENV mRNA level measured using RT-qPCR was upregulated upon CV-B4 infection. The HERV-W ENV protein was detected in the infected cells using the immunoblot assay. In human PBMCs inoculated with CV-B4 or when CV-B4 was incubated with an enhancing serum, the HERV-W ENV mRNA level was higher than the background RNA level. In monocyte-derived macrophages obtained from 5 of 13 donors, the HERV-W ENV mRNA level was higher in cultures inoculated with CV-B4 than in the control. Therefore, CV-B4 can upregulate or induce the transcription of a certain HERV-W ENV copy (or copies) in primary cell cultures, such as monocytes, macrophages, and pancreatic cells.
Collapse
Affiliation(s)
- Arthur Dechaumes
- Laboratoire de Virologie ULR3610 Univ Lille, CHU Lille, 59000 Lille, France; (A.D.); (A.B.); (F.S.); (J.V.)
| | - Antoine Bertin
- Laboratoire de Virologie ULR3610 Univ Lille, CHU Lille, 59000 Lille, France; (A.D.); (A.B.); (F.S.); (J.V.)
| | - Famara Sane
- Laboratoire de Virologie ULR3610 Univ Lille, CHU Lille, 59000 Lille, France; (A.D.); (A.B.); (F.S.); (J.V.)
| | - Sandrine Levet
- Geneuro Innovation, 69008 Lyon, France; (S.L.); (B.C.); (J.P.); (H.P.)
| | - Jennifer Varghese
- Laboratoire de Virologie ULR3610 Univ Lille, CHU Lille, 59000 Lille, France; (A.D.); (A.B.); (F.S.); (J.V.)
- Manipal Institute of Virology, Manipal Academy of Higher Education, Karnataka 576104, India;
| | - Benjamin Charvet
- Geneuro Innovation, 69008 Lyon, France; (S.L.); (B.C.); (J.P.); (H.P.)
| | - Valéry Gmyr
- U1190 Univ Lille, Inserm, CHU Lille, European Genomic Institute for Diabetes, 59000 Lille, France; (V.G.); (J.K.-C.); (F.P.)
| | - Julie Kerr-Conte
- U1190 Univ Lille, Inserm, CHU Lille, European Genomic Institute for Diabetes, 59000 Lille, France; (V.G.); (J.K.-C.); (F.P.)
| | - Justine Pierquin
- Geneuro Innovation, 69008 Lyon, France; (S.L.); (B.C.); (J.P.); (H.P.)
| | | | - François Pattou
- U1190 Univ Lille, Inserm, CHU Lille, European Genomic Institute for Diabetes, 59000 Lille, France; (V.G.); (J.K.-C.); (F.P.)
| | - Hervé Perron
- Geneuro Innovation, 69008 Lyon, France; (S.L.); (B.C.); (J.P.); (H.P.)
- Geneuro SA, 1228 Geneva, Switzerland
- Faculté de Médecine Laënnec, Université de Lyon, 69008 Lyon, France
| | - Didier Hober
- Laboratoire de Virologie ULR3610 Univ Lille, CHU Lille, 59000 Lille, France; (A.D.); (A.B.); (F.S.); (J.V.)
| |
Collapse
|
29
|
Abstract
Multiple sclerosis (MS), a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system, is today a leading cause of unpredictable lifelong disability in young adults. The treatment of patients in progressive stages remains highly challenging, alluding to our limited understanding of the underlying pathological processes. In this review, we provide insights into the mechanisms underpinning MS progression from a perspective of epigenetics, that refers to stable and mitotically heritable, yet reversible, changes in the genome activity and gene expression. We first recapitulate findings from epigenetic studies examining the brain tissue of progressive MS patients, which support a contribution of DNA and histone modifications in impaired oligodendrocyte differentiation, defective myelination/remyelination and sustained neuro-axonal vulnerability. We next explore possibilities for identifying factors affecting progression using easily accessible tissues such as blood by comparing epigenetic signatures in peripheral immune cells and brain tissue. Despite minor overlap at individual methylation sites, nearly 30% of altered genes reported in peripheral immune cells of progressive MS patients were found in brain tissue, jointly converging on alterations of neuronal functions. We further speculate about the mechanisms underlying shared epigenetic patterns between blood and brain, which likely imply the influence of internal (genetic control) and/or external (e.g. smoking and ageing) factors imprinting a common signature in both compartments. Overall, we propose that epigenetics might shed light on clinically relevant mechanisms involved in disease progression and open new avenues for the treatment of progressive MS patients in the future.
Collapse
Affiliation(s)
- L Kular
- From the, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - M Jagodic
- From the, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Talotta R, Atzeni F, Laska MJ. Retroviruses in the pathogenesis of systemic lupus erythematosus: Are they potential therapeutic targets? Autoimmunity 2020; 53:177-191. [PMID: 32321325 DOI: 10.1080/08916934.2020.1755962] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is characterised by the hyper-activation of immunologic pathways related to the antiviral response. Exogenous and endogenous retroviruses, by integrating their DNA templates in the host cell genome, may epigenetically control the transcription of genes involved in the immune response. Furthermore, their nucleic acids or neo-synthesized proteins could stimulate the sensor molecules placed upstream the inflammatory cascade. Exogenous retroviruses, like human immunodeficiency virus, have been associated to SLE-like manifestations or to a fair SLE diagnosis. In addition, there is some evidence confirming a pathogenic role of human endogenous retroviruses in SLE. In line with these data, the use of antiretroviral agents could represent an attractive opportunity in the future therapeutic algorithms of this disease, but studies are still missing.
Collapse
Affiliation(s)
- Rossella Talotta
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital "Gaetano Martino", Messina, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital "Gaetano Martino", Messina, Italy
| | | |
Collapse
|
31
|
Abstract
PURPOSE OF THE REVIEW The aim of this review is to discuss recent data pointing at an involvement of human endogenous retroviruses (HERVs) in type 1 diabetes (T1D) onset and progression. RECENT FINDINGS The envelope protein of HERV-W family, named HERV-W-Env, was detected in pancreata from T1D patients and was shown to display pro-inflammatory properties and direct toxicity toward pancreatic beta cells. The etiopathogenesis of T1D remains elusive, even if conventional environmental viral infections have been recurrently involved. Nonetheless, a new category of pathogens may provide the missing link between genetic susceptibility and environmental factors long thought to contribute to T1D onset. A number of studies have now shown that HERV sequences, which are normally inactivated or repressed in the human genome, could be activated by environmental viruses. Thus, if similarly activated by viruses associated with T1D, disregarded HERV genes may underlie T1D genetic susceptibility. Moreover, once expressed, HERV elements may display broad pathogenic properties, which identify them as potential new therapeutic targets.
Collapse
Affiliation(s)
- Sandrine Levet
- GeNeuro Innovation, 60 avenue Rockefeller, 69008 Lyon, France
| | - B. Charvet
- GeNeuro Innovation, 60 avenue Rockefeller, 69008 Lyon, France
| | - A. Bertin
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Université Lille, F-59000 Lille, France
| | - A. Deschaumes
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Université Lille, F-59000 Lille, France
| | - H. Perron
- GeNeuro Innovation, 60 avenue Rockefeller, 69008 Lyon, France
- Laboratoire des déficits immunitaires, University of Lyon, Lyon, France
- Plan-les-Ouates, GeNeuro SA, Geneva, Switzerland
| | - D. Hober
- Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Université Lille, F-59000 Lille, France
| |
Collapse
|
32
|
Milo R, Korczyn AD, Manouchehri N, Stüve O. The temporal and causal relationship between inflammation and neurodegeneration in multiple sclerosis. Mult Scler 2019; 26:876-886. [PMID: 31682184 DOI: 10.1177/1352458519886943] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is currently incompletely understood whether inflammation and neurodegeneration are causally related in multiple sclerosis (MS). The sequence of a potential causal relationship is also unknown. Inflammation is present in rather all clinical stages of MS. Its role in the pathogenesis of MS is supported by histopathological analyses, genetic data, and numerous animal models of MS. All approved disease-modifying therapies that reduce clinical relapses and diminish the accumulation of lesions on neuroimaging are anti-inflammatory. Axonal loss and accelerated brain volume loss can also be detected from clinical disease onset throughout all stages. The expression of neurofilament light chain in cerebrospinal fluid and serum, a scaffolding protein in axons and dendrites, is a biomarker of neuronal injury associated with clinical relapses and reflects neuronal loss during episodes of acute inflammation. The recent association of human endogenous retrovirus (HERV) and its envelope proteins with MS illustrates a pathogenic pathway that causally links central nervous system (CNS)-intrinsic proinflammatory effects and inhibition of myelin repair and neuroregeneration. A review of current data on the causal relationship between inflammation and neurodegeneration in MS identified numerous plausible pathomechanisms that link the two events. Observations from most experimental models appear to favor a pathogenesis in which inflammation precedes neurodegeneration.
Collapse
Affiliation(s)
- Ron Milo
- Department of Neurology, Barzilai Medical Center, Ashkelon, Israel/Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Amos D Korczyn
- Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Navid Manouchehri
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA/Neurology Section, Medical Service, VA North Texas Health Care System, Dallas, TX, USA/Department of Neurology, Klinikum rechts der Isar, Technische Universität München, München, Germany
| |
Collapse
|
33
|
Gruchot J, Kremer D, Küry P. Neural Cell Responses Upon Exposure to Human Endogenous Retroviruses. Front Genet 2019; 10:655. [PMID: 31354794 PMCID: PMC6637040 DOI: 10.3389/fgene.2019.00655] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
Human endogenous retroviruses (HERVs) are ancient retroviral elements, which invaded the human germ line several million years ago. Subsequent retrotransposition events amplified these sequences, resulting in approximately 8% of the human genome being composed of HERV sequences today. These genetic elements, normally dormant within human genomes, can be (re)-activated by environmental factors such as infections with other viruses, leading to the expression of viral proteins and, in some instances, even to viral particle production. Several studies have shown that the expression of these retroviral elements correlates with the onset and progression of neurological diseases such as multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). Further studies provided evidence on additional roles for HERVs in schizophrenia (SCZ). Since these diseases are still not well understood, HERVs might constitute a new category of pathogenic components that could significantly change our understanding of these pathologies. Moreover, knowledge about their mode of action might also help to develop novel and more powerful approaches for the treatment of these complex diseases. Therefore, the main scope of this review is a description of the current knowledge on the involvement of HERV-W and HERV-K in neurological disease specifically focusing on the effects they exert on neural cells of the central nervous system.
Collapse
Affiliation(s)
- Joel Gruchot
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - David Kremer
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
34
|
Greenig M. HERVs, immunity, and autoimmunity: understanding the connection. PeerJ 2019; 7:e6711. [PMID: 30984482 PMCID: PMC6452852 DOI: 10.7717/peerj.6711] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/28/2019] [Indexed: 12/30/2022] Open
Abstract
Since their discovery in the 1960s, further investigation into endogenous retroviruses (ERVs) has challenged the conventional view of viral sequences as exclusively parasitic elements. Once presumed to be a group of passive genetic relics, it is becoming increasingly clear that this view of ERVs, while generally accurate, is incorrect in many specific cases. Research has identified ERV genes that appear to be co-opted by their mammalian hosts, but the biological function of ERV elements in humans remains a controversial subject. One area that has attracted some attention in this domain is the role of co-opted ERV elements in mammalian immune systems. The relationship between ERVs and human autoimmune diseases has also been investigated, but has historically been treated as a separate topic. This review will summarize the current evidence concerning the phenotypic significance of ERVs, both in the healthy immune system and in manifestations of autoimmunity. Furthermore, it will evaluate the relationship between these fields of study, and propose previously-unexplored molecular mechanisms through which human endogenous retroviruses might contribute to certain autoimmune pathologies. Investigation into these novel mechanisms could further our understanding of the molecular basis of autoimmune disease, and may one day provide new targets for treatment.
Collapse
Affiliation(s)
- Matthew Greenig
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
35
|
Diebold M, Derfuss T. The monoclonal antibody GNbAC1: targeting human endogenous retroviruses in multiple sclerosis. Ther Adv Neurol Disord 2019; 12:1756286419833574. [PMID: 30873219 PMCID: PMC6407165 DOI: 10.1177/1756286419833574] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Multiple sclerosis (MS) is an autoimmune demyelinating disorder of the central nervous system (CNS). Despite improvements of immunomodulatory therapies in relapsing–remitting MS, the pathomechanisms of progressive disease are poorly understood and therapeutically addressed to date. A pathophysiological role for proteins encoded by human endogenous retroviruses (HERVs) has been proposed. GNbAC1 is a monoclonal antibody directed against the envelope protein of a HERV with postulated involvement in MS. Methods: This review addresses the treatment concept of GNbAC1, the design, preclinical and clinical development of the antibody, as published by November 2018. All four in-human trials (of which two addressed MS) are discussed. Conclusion: The treatment concept of GNbAC1 is appealing but remains controversial due to conflicting results regarding the hypothesized underlying pathomechanism. Anticipated immunomodulatory effects were not observed in clinical or pharmacodynamic analyses of the currently available data. However, a magnetic resonance imaging sign compatible with the remyelinating potential of GNbAC1 encouraged further development of this antibody in progressive MS. No relevant issues with tolerability or safety have been described to date.
Collapse
Affiliation(s)
- Martin Diebold
- Neurological Policlinic and Clinic, University Hospital and University of Basel, Basel, Switzerland
| | - Tobias Derfuss
- Neurological Policlinic and Clinic, University Hospital and University of Basel, Petersgraben 4, Basel, 4031, Switzerland
| |
Collapse
|
36
|
Chen Y, Yan Q, Zhou P, Li S, Zhu F. HERV-W env regulates calcium influx via activating TRPC3 channel together with depressing DISC1 in human neuroblastoma cells. J Neurovirol 2019; 25:101-113. [PMID: 30397826 DOI: 10.1007/s13365-018-0692-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
The activation and involvement of human endogenous retroviruses W family envelope gene (HERV-W env, also called ERVWE1) have been reported in several neuropsychiatric disorders, including schizophrenia, as well as in multiple sclerosis (MS). Dysregulation of intracellular calcium content is also involved in the pathogenesis of these diseases. Our previous studies showed that HERV-W env overexpression results in activation of small conductance Ca2+-activated K+ channel protein 3 (SK3), a potential risk factor for schizophrenia. In the present study, we aimed to elucidate the relationship between HERV-W env and calcium signaling in schizophrenia. Our results showed that HERV-W env could induce Ca2+ influx in two human neuroblastoma cell lines and upregulate the expression and activation of TRPC3 in cells. The abnormal increase in intracellular Ca2+ concentration was inhibited by addition of the TRPC3 channel blocker pyr3, demonstrating that the Ca2+ influx induced by HERV-W env was TRPC3-dependent. Further experiments showed that HERV-W env overexpression downregulated DISC1, while knockdown of DISC1 promoted activation of TRPC3 without affecting TRPC3 expression. In conclusion, HERV-W env induced Ca2+ influx in human neuroblastoma cells by activating the TRPC3 channel through directly regulating its expression or downregulating DISC1, which could also increase TRPC3 activation without affecting TRPC3 expression. These findings provide new insights into how HERV-W env affects neuronal activity and contributes to the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Yatang Chen
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Qiujin Yan
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Ping Zhou
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China
| | - Shan Li
- Department of Integrated Medicine, Dongfeng Hospital, Hubei University of Medicine, Hubei, 442000, People's Republic of China
| | - Fan Zhu
- Department of Medical Microbiology, School of Medicine, Wuhan University, 185 Donghu Road, Wuhan, 430071, People's Republic of China.
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, 430071, People's Republic of China.
| |
Collapse
|
37
|
Barth M, Gröger V, Cynis H, Staege MS. Identification of human endogenous retrovirus transcripts in Hodgkin Lymphoma cells. Mol Biol Rep 2019; 46:1885-1893. [PMID: 30707417 DOI: 10.1007/s11033-019-04640-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/23/2019] [Indexed: 11/24/2022]
Abstract
During the last decades, the prognosis for patients with Hodgkin Lymphoma (HL) has been steadily improved. Nevertheless, new and less toxic therapy strategies have to be developed especially for patients with advanced disease. The activation of human endogenous retroviruses (HERV) is suspected to occur in HL and therefore, HERV might represent interesting target structures. In order to identify transcribed HERV of the HERV-H and HERV-K families in HL we used a reverse transcription-polymerase chain reaction based cloning approach. In addition to unspliced HERV-H and HERV-K transcripts, we detected spliced HERV-K transcripts that matched genomic sequences with the expected splicing-donor and splicing-acceptor sites. Of particular interest was the expression of HERV-K18 related transcripts at the CD48 locus. Our data indicate transcriptional activity of several HERV loci in HL cells.
Collapse
Affiliation(s)
- Marie Barth
- University Clinic and Outpatient Clinic for Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany
| | - Victoria Gröger
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle, Germany
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, 06120, Halle, Germany
| | - Martin Sebastian Staege
- University Clinic and Outpatient Clinic for Pediatrics I, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle, Germany.
| |
Collapse
|
38
|
Bourque G, Burns KH, Gehring M, Gorbunova V, Seluanov A, Hammell M, Imbeault M, Izsvák Z, Levin HL, Macfarlan TS, Mager DL, Feschotte C. Ten things you should know about transposable elements. Genome Biol 2018; 19:199. [PMID: 30454069 PMCID: PMC6240941 DOI: 10.1186/s13059-018-1577-z] [Citation(s) in RCA: 760] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Transposable elements (TEs) are major components of eukaryotic genomes. However, the extent of their impact on genome evolution, function, and disease remain a matter of intense interrogation. The rise of genomics and large-scale functional assays has shed new light on the multi-faceted activities of TEs and implies that they should no longer be marginalized. Here, we introduce the fundamental properties of TEs and their complex interactions with their cellular environment, which are crucial to understanding their impact and manifold consequences for organismal biology. While we draw examples primarily from mammalian systems, the core concepts outlined here are relevant to a broad range of organisms.
Collapse
Affiliation(s)
- Guillaume Bourque
- Department of Human Genetics, McGill University, Montréal, Québec, H3A 0G1, Canada.
- Canadian Center for Computational Genomics, McGill University, Montréal, Québec, H3A 0G1, Canada.
| | - Kathleen H Burns
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mary Gehring
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Molly Hammell
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Michaël Imbeault
- Department of Genetics, University of Cambridge, Cambridge, CB2 3EH, UK
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - Henry L Levin
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Todd S Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, The National Institutes of Health, Bethesda, Maryland, USA
| | - Dixie L Mager
- Terry Fox Laboratory, British Columbia Cancer Agency and Department of Medical Genetics, University of BC, Vancouver, BC, V5Z1L3, Canada
| | - Cédric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
39
|
Grandi N, Tramontano E. Human Endogenous Retroviruses Are Ancient Acquired Elements Still Shaping Innate Immune Responses. Front Immunol 2018; 9:2039. [PMID: 30250470 PMCID: PMC6139349 DOI: 10.3389/fimmu.2018.02039] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/20/2018] [Indexed: 12/25/2022] Open
Abstract
About 8% of our genome is composed of sequences with viral origin, namely human Endogenous Retroviruses (HERVs). HERVs are relics of ancient infections that affected the primates' germ line along the last 100 million of years, and became stable elements at the interface between self and foreign DNA. Intriguingly, HERV co-evolution with the host led to the domestication of activities previously devoted to the retrovirus life cycle, providing novel cellular functions. For example, selected HERV envelope proteins have been coopted for pregnancy-related purposes, and proviral Long Terminal Repeats participate in the transcriptional regulation of various cellular genes. Given the HERV persistence in the host genome and its basal expression in most healthy tissues, it is reasonable that human defenses should prevent HERV-mediated immune activation. Despite this, HERVs and their products (including RNA, cytosolic DNA, and proteins) are still able to modulate and be influenced by the host immune system, fascinatingly suggesting a central role in the evolution and functioning of the human innate immunity. Indeed, HERV sequences had been major contributors in shaping and expanding the interferon network, dispersing inducible genes that have been occasionally domesticated in various mammalian lineages. Also the HERV integration within or near to genes encoding for critical immune factors has been shown to influence their activity, or to be responsible for their polymorphic variation in the human population, such as in the case of an HERV-K(HML10) provirus in the major histocompatibility complex region. In addition, HERV expressed products have been shown to modulate innate immunity effectors, being therefore often related on the one side to inflammatory and autoimmune disorders, while on the other side to the control of excessive immune activation through their immunosuppressive properties. Finally, HERVs have been proposed to establish a protective effect against exogenous infections. The present review summarizes the involvement of HERVs and their products in innate immune responses, describing how their intricate interplay with the first line of human defenses can actively contribute either to the host protection or to his damage, implying a subtle balance between the persistence of HERV expression and the maintenance of a basal immune alert.
Collapse
Affiliation(s)
- Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Enzo Tramontano
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| |
Collapse
|
40
|
Wang X, Huang J, Zhu F. Human Endogenous Retroviral Envelope Protein Syncytin-1 and Inflammatory Abnormalities in Neuropsychological Diseases. Front Psychiatry 2018; 9:422. [PMID: 30245643 PMCID: PMC6137383 DOI: 10.3389/fpsyt.2018.00422] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/17/2018] [Indexed: 12/16/2022] Open
Abstract
Human endogenous retroviruses (HERVs) comprise approximately 8% of the human genome. Recent studies have considered HERVs as potential pathogenic factors. The majority of HERV genes are mutated and not capable of encoding functional proteins; regardless, some HERV genes, such as HERV-W envelope (env) glycoprotein, are known to have intact open reading frames. The HERV-W element on 7q21.2, which encodes a protein referred to as Syncytin-1, participates in human placental morphogenesis and can activate a pro-inflammatory and autoimmune cascade. Neuropsychological disorders are typically linked to inflammatory abnormalities. In this study, we review that Syncytin-1 has been increasingly involved in the development of neuropsychological disorders, such as schizophrenia and multiple sclerosis (MS). This study also presents inflammation imbalances in schizophrenia and MS. More importantly, we discuss the potential role and molecular mechanisms by which Syncytin-1 regulates inflammatory abnormalities in neuropsychological diseases. In summary, Syncytin-1 activity may represent a novel molecular pathogenic mechanism in neuropyschological diseases, such as schizophrenia and MS.
Collapse
Affiliation(s)
- Xiuling Wang
- Department of Medical Microbiology, School of MedicineWuhan University, Wuhan, China
- Department of Medical Laboratory, The Central Hospital of WuhanHuazhong University of Science and Technology, Wuhan, China
| | - Jin Huang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of WuhanHuazhong University of Science and Technology, Wuhan, China
| | - Fan Zhu
- Department of Medical Microbiology, School of MedicineWuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and ImmunologyWuhan University, Wuhan, China
| |
Collapse
|
41
|
Tartaglione AM, Cipriani C, Chiarotti F, Perrone B, Balestrieri E, Matteucci C, Sinibaldi-Vallebona P, Calamandrei G, Ricceri L. Early Behavioral Alterations and Increased Expression of Endogenous Retroviruses Are Inherited Across Generations in Mice Prenatally Exposed to Valproic Acid. Mol Neurobiol 2018; 56:3736-3750. [DOI: 10.1007/s12035-018-1328-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022]
|
42
|
Curtin F, Bernard C, Levet S, Perron H, Porchet H, Médina J, Malpass S, Lloyd D, Simpson R. A new therapeutic approach for type 1 diabetes: Rationale for GNbAC1, an anti-HERV-W-Env monoclonal antibody. Diabetes Obes Metab 2018; 20:2075-2084. [PMID: 29749030 DOI: 10.1111/dom.13357] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022]
Abstract
We describe a newly identified therapeutic target for type 1 diabetes (T1D): an envelope protein of endogenous retroviral origin, human endogenous retrovirus W envelope (HERV-W-Env). HERV-W-Env was found to be detected in the blood of ~60% of patients with T1D and is expressed in acinar pancreatic cells of 75% of patients with T1D at post mortem examination. Preclinical experiments showed that this protein displays direct cytotoxicity on human β-islet cells. In vivo HERV-W-Env impairs the insulin and glucose metabolism in transgenic mice expressing HERV-W-Env. GNbAC1, an IgG4 monoclonal antibody, has been developed to specifically target HERV-W-Env and to neutralize the effect of HERV-W-Env in vitro and in vivo. GNbAC1 is currently in clinical development for multiple sclerosis and > 300 subjects have been administered with GNbAC1 so far. GNbAC1 is now being tested in T1D in the RAINBOW-T1D study, which is a randomized placebo-controlled study with the objective of showing the safety and pharmacodynamic response of GNbAC1 in patients who have had T1D with a maximum of 4 years' duration. GNbAC1 is being tested vs placebo at the dose of 6 mg/kg in 60 patients during six repeated administrations for 6 months; a 6-month open-label extension will follow. The primary endpoint is to assess safety, and secondary endpoints are the pharmacodynamic responses to GNbAC1. GNbAC1 targeting HERV-W-Env is currently in clinical development in T1D, with the first safety and pharmacodynamic study. If the study results are positive, this may open the door to the development of an innovative non-immunomodulatory disease-modifying treatment for T1D.
Collapse
Affiliation(s)
- Francois Curtin
- GeNeuro SA, Plan-les-Ouates, Switzerland
- Division of Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | | - Hervé Perron
- GeNeuro SA, Plan-les-Ouates, Switzerland
- Geneuro-Innovation, Lyon, France
- Laboratory of Immune Deficiencies, Faculty of Medicine Laënnec, University of Lyon, Lyon, France
| | - Hervé Porchet
- GeNeuro SA, Plan-les-Ouates, Switzerland
- Department of Pharmacology, University of Pretoria, Pretoria, South Africa
| | | | - Sam Malpass
- Southern Star Research Pty Ltd, Gordon, Australia
| | - David Lloyd
- Southern Star Research Pty Ltd, Gordon, Australia
| | | |
Collapse
|
43
|
Morris G, Maes M, Murdjeva M, Puri BK. Do Human Endogenous Retroviruses Contribute to Multiple Sclerosis, and if So, How? Mol Neurobiol 2018; 56:2590-2605. [PMID: 30047100 PMCID: PMC6459794 DOI: 10.1007/s12035-018-1255-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022]
Abstract
The gammaretroviral human endogenous retrovirus (HERV) families MRSV/HERV-W and HERV-H (including the closely related HERV-Fc1) are associated with an increased risk of multiple sclerosis (MS). Complete HERV sequences betray their endogenous retroviral origin, with open reading frames in gag, pro, pol and env being flanked by two long terminal repeats containing promoter and enhancer sequences with the capacity to regulate HERV transactivation and the activity of host genes in spite of endogenous epigenetic repression mechanisms. HERV virions, RNA, cDNA, Gag and Env, and antibodies to HERV transcriptional products, have variously been found in the blood and/or brain and/or cerebrospinal fluid of MS patients, with the HERV expression level being associated with disease status. Furthermore, some HERV-associated single nucleotide polymorphisms (SNPs), such as rs662139 T/C in a 3-kb region of Xq22.3 containing a HERV-W env locus, and rs391745, upstream of the HERV-Fc1 locus on the X chromosome, are associated with MS susceptibility, while a negative association has been reported with SNPs in the tripartite motif-containing (TRIM) protein-encoding genes TRIM5 and TRIM22. Factors affecting HERV transcription include immune activation and inflammation, since HERV promoter regions possess binding sites for related transcription factors; oxidative stress, with oxidation of guanine to 8-oxoguanine and conversion of cytosine to 5-hydroxymethylcytosine preventing binding of methyl groups transferred by DNA methyltransferases; oxidative stress also inhibits the activity of deacetylases, thereby favouring the acetylation of histone lysine residues favouring gene expression; interferon beta; natalizumab treatment; impaired epigenetic regulation; and the sex of patients.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Marianna Murdjeva
- Department of Microbiology and Immunology, Medical University, Plovdiv, Bulgaria
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
44
|
Kremer D, Küry P, Hartung HP. ECTRIMS/ACTRIMS 2017: Closing in on neurorepair in progressive multiple sclerosis. Mult Scler 2018; 24:696-700. [PMID: 29629605 DOI: 10.1177/1352458518768770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND While there is now a multitude of potent medications for relapsing-remitting multiple sclerosis (RRMS), effective therapies targeting neurodegeneration in progressive multiple sclerosis types are still lacking. Stimulation of neurorepair in this disease remains a pathogenetically defined treatment goal. However, therapeutic progress is slowed by the still inadequate tool set to capture "regeneration/repair" in MS and to define appropriate outcomes in clinical trials. OBJECTIVES In this review, we discuss studies investigating promising regenerative agents for progressive MS which were recently presented during the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS)/Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) 2017 meeting in Paris.
Collapse
Affiliation(s)
- David Kremer
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
45
|
Küry P, Nath A, Créange A, Dolei A, Marche P, Gold J, Giovannoni G, Hartung HP, Perron H. Human Endogenous Retroviruses in Neurological Diseases. Trends Mol Med 2018; 24:379-394. [PMID: 29551251 PMCID: PMC7185488 DOI: 10.1016/j.molmed.2018.02.007] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022]
Abstract
The causes of multiple sclerosis and amyotrophic lateral sclerosis have long remained elusive. A new category of pathogenic components, normally dormant within human genomes, has been identified: human endogenous retroviruses (HERVs). These represent ∼8% of the human genome, and environmental factors have reproducibly been shown to trigger their expression. The resulting production of envelope (Env) proteins from HERV-W and HERV-K appears to engage pathophysiological pathways leading to the pathognomonic features of MS and ALS, respectively. Pathogenic HERV elements may thus provide a missing link in understanding these complex diseases. Moreover, their neutralization may represent a promising strategy to establish novel and more powerful therapeutic approaches.
Collapse
Affiliation(s)
- Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Avindra Nath
- Section of infections of the Nervous System, National Institute of Neurological Diseases and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Alain Créange
- Service de Neurologie, Groupe Hospitalier Henri Mondor, Assistance Publique Hopitaux de Paris (APHP), Université Paris Est, Créteil, France
| | - Antonina Dolei
- Department of Virology, University of Sassari, Sassari, Italy
| | - Patrice Marche
- Institute for Advanced Biosciences (IAB), University of Grenoble-Alpes, La Tronche, France; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1209, La Tronche, France
| | - Julian Gold
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University, London, UK; The Albion Centre, Prince of Wales Hospital, Sydney, Australia
| | - Gavin Giovannoni
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University, London, UK
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| | - Hervé Perron
- Geneuro, Plan les Ouates, Geneva, Switzerland; University of Lyon, Lyon, France
| |
Collapse
|
46
|
Grandi N, Tramontano E. HERV Envelope Proteins: Physiological Role and Pathogenic Potential in Cancer and Autoimmunity. Front Microbiol 2018; 9:462. [PMID: 29593697 PMCID: PMC5861771 DOI: 10.3389/fmicb.2018.00462] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Human endogenous retroviruses (HERVs) are relics of ancient infections accounting for about the 8% of our genome. Despite their persistence in human DNA led to the accumulation of mutations, HERVs are still contributing to the human transcriptome, and a growing number of findings suggests that their expression products may have a role in various diseases. Among HERV products, the envelope proteins (Env) are currently highly investigated for their pathogenic properties, which could likely be participating to several disorders with complex etiology, particularly in the contexts of autoimmunity and cancer. In fact, HERV Env proteins have been shown, on the one side, to trigger both innate and adaptive immunity, prompting inflammatory, cytotoxic and apoptotic reactions; and, on the other side, to prevent the immune response activation, presenting immunosuppressive properties and acting as immune downregulators. In addition, HERV Env proteins have been shown to induce abnormal cell-cell fusion, possibly contributing to tumor development and metastasizing processes. Remarkably, even highly defective HERV env genes and alternative env splicing variants can provide further mechanisms of pathogenesis. A well-known example is the HERV-K(HML2) env gene that, depending on the presence or the absence of a 292-bp deletion, can originate two proteins of different length (Np9 and Rec) proposed to have oncogenic properties. The understanding of their involvement in complex pathological disorders made HERV Env proteins potential targets for therapeutic interventions. Of note, a monoclonal antibody directed against a HERV-W Env is currently under clinical trial as therapeutic approach for multiple sclerosis, representing the first HERV-based treatment. The present review will focus on the current knowledge of the HERV Env expression, summarizing its role in human physiology and its possible pathogenic effects in various cancer and autoimmune disorders. It moreover analyzes HERV Env possible exploitation for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Enzo Tramontano
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cagliari, Italy
| |
Collapse
|
47
|
Wang X, Liu Z, Wang P, Li S, Zeng J, Tu X, Yan Q, Xiao Z, Pan M, Zhu F. Syncytin-1, an endogenous retroviral protein, triggers the activation of CRP via TLR3 signal cascade in glial cells. Brain Behav Immun 2018; 67:324-334. [PMID: 28928004 DOI: 10.1016/j.bbi.2017.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/08/2017] [Accepted: 09/15/2017] [Indexed: 12/17/2022] Open
Abstract
Schizophrenia is a devastating psychiatric disorder that impacts on social functioning and quality of life, and there is accumulating evidence that inflammation is a potential pathogenic mechanism of schizophrenia. However, the mechanism of inflammation possibly occurred in schizophrenia has not been well understood. The endogenous retroviral protein syncytin-1 and inflammatory marker CRP are both abnormally expressed in schizophrenia patients. CRP is one of the markers of bacterial infection generally. Less clear is whether virus or viral protein can trigger the activation of CRP. Here, we detected a robust increase of the levels of syncytin-1 and CRP in schizophrenia patients, and displayed a positive correlation and marked consistency between expressions of syncytin-1 and CRP in schizophrenia patients. Furthermore, overexpression of syncytin-1 significantly elevated the levels of CRP, TLR3, and IL-6 in both human microglia and astrocytes. TLR3 deficiency impaired the expressions of CRP and IL-6 induced by syncytin-1. Importantly, we observed a cellular co-localization and a direct interaction between syncytin-1 and TLR3. Additionally, knockdown of IL-6 inhibited the syncytin-1-induced CRP expression. Thus, the totality of these results showed that viral protein syncytin-1 could trigger the activation of CRP, which might explain the elevated CRP in sterile inflammation and exhibit a novel mechanism for regulation of inflammation by syncytin-1 in schizophrenia.
Collapse
Affiliation(s)
- Xiuling Wang
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, PR China; Department of Medical Laboratory, Central Hospital of Wuhan, Wuhan 430014, PR China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei, PR China
| | - Peigang Wang
- Department of Global Health Institute, Wuhan University, Wuhan 430071, PR China
| | - Shan Li
- Department of Integrated Medicine, Dongfeng Hospital, Hubei University of Medicine, Hubei 442000, PR China
| | - Jie Zeng
- Jiangsu Hengrui Medicine Co., Ltd, No.7 Kunlunshan Road, Lianyungang Eco & Tech Development Zone, Jiangsu Province, Jiangsu 222000, PR China
| | - Xiaoning Tu
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, PR China
| | - Qiujin Yan
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, PR China
| | - Zheman Xiao
- Department of Neurology, Institution of Neuropsychiatry Research, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei, PR China
| | - Mengxian Pan
- Department of Pathophysiology, School of Medicine, Wuhan University, Wuhan 430071, PR China
| | - Fan Zhu
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, PR China; Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
48
|
Arru G, Sechi E, Mariotto S, Farinazzo A, Mancinelli C, Alberti D, Ferrari S, Gajofatto A, Capra R, Monaco S, Deiana GA, Caggiu E, Mameli G, Sechi LA, Sechi GP. Antibody response against HERV-W env surface peptides differentiates multiple sclerosis and neuromyelitis optica spectrum disorder. Mult Scler J Exp Transl Clin 2017; 3:2055217317742425. [PMID: 29204291 PMCID: PMC5703109 DOI: 10.1177/2055217317742425] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/21/2017] [Indexed: 11/16/2022] Open
Abstract
Background A specific humoral immune response against HERV-W envelope surface (env-su) glycoprotein antigens has been reported in serum of patients with multiple sclerosis (MS). However, it has not been evaluated to date in patients with neuromyelitis optica spectrum disorder (NMOSD). Objective The objective of this paper is to investigate whether antibody (Ab) response against HERV-W env-su antigenic peptides differs between NMOSD and MS. Methods Serum samples were collected from 36 patients with NMOSD, 36 patients with MS and 36 healthy control individuals (HCs). An indirect ELISA was set up to detect specific Abs against HERV-W env-su peptides. Results Our data showed that two antigenic peptides, particularly HERV-Wenv93–108 and HERV-Wenv248–262, were statistically significantly present only in serum of MS compared to NMOSD and HCs. Thus, the specific humoral immune response against HERV-W env-su glycoprotein antigens found in MS is widely missing in NMOSD. Conclusion Increased circulating serum levels of these HERV-W Abs may be suitable as additional biomarkers to better differentiate MS from NMOSD.
Collapse
Affiliation(s)
- Giannina Arru
- Department of Clinical and Experimental Medicine, University of Sassari, Italy
| | - Elia Sechi
- Department of Clinical and Experimental Medicine, University of Sassari, Italy
| | - Sara Mariotto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Alessia Farinazzo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Chiara Mancinelli
- Multiple Sclerosis Centre, Spedali Civili of Brescia, Montichiari, Italy
| | - Daniela Alberti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Sergio Ferrari
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Alberto Gajofatto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Ruggero Capra
- Multiple Sclerosis Centre, Spedali Civili of Brescia, Montichiari, Italy
| | - Salvatore Monaco
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | - Giovanni A Deiana
- Department of Clinical and Experimental Medicine, University of Sassari, Italy
| | - Elisa Caggiu
- Department of Biomedical Sciences, University of Sassari, Italy
| | - Giuseppe Mameli
- Department of Biomedical Sciences, University of Sassari, Italy
| | | | - Gian Pietro Sechi
- Department of Clinical and Experimental Medicine, University of Sassari, Italy
| |
Collapse
|
49
|
Levet S, Medina J, Joanou J, Demolder A, Queruel N, Réant K, Normand M, Seffals M, Dimier J, Germi R, Piofczyk T, Portoukalian J, Touraine JL, Perron H. An ancestral retroviral protein identified as a therapeutic target in type-1 diabetes. JCI Insight 2017; 2:94387. [PMID: 28878130 DOI: 10.1172/jci.insight.94387] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/25/2017] [Indexed: 12/16/2022] Open
Abstract
Human endogenous retroviruses (HERVs), remnants of ancestral viral genomic insertions, are known to represent 8% of the human genome and are associated with several pathologies. In particular, the envelope protein of HERV-W family (HERV-W-Env) has been involved in multiple sclerosis pathogenesis. Investigations to detect HERV-W-Env in a few other autoimmune diseases were negative, except in type-1 diabetes (T1D). In patients suffering from T1D, HERV-W-Env protein was detected in 70% of sera, and its corresponding RNA was detected in 57% of peripheral blood mononuclear cells. While studies on human Langerhans islets evidenced the inhibition of insulin secretion by HERV-W-Env, this endogenous protein was found to be expressed by acinar cells in 75% of human T1D pancreata. An extensive immunohistological analysis further revealed a significant correlation between HERV-W-Env expression and macrophage infiltrates in the exocrine part of human pancreata. Such findings were corroborated by in vivo studies on transgenic mice expressing HERV-W-env gene, which displayed hyperglycemia and decreased levels of insulin, along with immune cell infiltrates in their pancreas. Altogether, these results strongly suggest an involvement of HERV-W-Env in T1D pathogenesis. They also provide potentially novel therapeutic perspectives, since unveiling a pathogenic target in T1D.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Julie Dimier
- IBS, UMR 5075 CEA-CNRS-Université Grenoble-Alpes, Grenoble, France
| | - Raphaële Germi
- IBS, UMR 5075 CEA-CNRS-Université Grenoble-Alpes, Grenoble, France.,Department of Virology, Grenoble University Hospital, Grenoble, France
| | | | | | | | - Hervé Perron
- GeNeuro Innovation, Lyon, France.,Laboratoire des déficits immunitaires, University of Lyon, France.,GeNeuro SA, Plan-les-Ouates, Geneva, Switzerland
| |
Collapse
|
50
|
Retroviral envelope proteins: Involvement in neuropathogenesis. J Neurol Sci 2017; 380:151-163. [DOI: 10.1016/j.jns.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/23/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023]
|