99 results on '"Iampietro M"'
Search Results
2. Accelerated heterologous prime-boost adenovirus vector-based SIV vaccine in neonatal rhesus monkeys
- Author
-
Liu J, Li H, Iampietro M, and Barouch DH
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Published
- 2012
- Full Text
- View/download PDF
3. Vaccine protection against acquisition of neutralization-resistant SIV challenges in rhesus monkeys.
- Author
-
Barouch, Dan H, Liu, Jinyan, Li, Hualin, Maxfield, Lori F, Abbink, Peter, Lynch, Diana M, Iampietro, M Justin, SanMiguel, Adam, Seaman, Michael S, Ferrari, Guido, Forthal, Donald N, Ourmanov, Ilnour, Hirsch, Vanessa M, Carville, Angela, Mansfield, Keith G, Stablein, Donald, Pau, Maria G, Schuitemaker, Hanneke, Sadoff, Jerald C, Billings, Erik A, Rao, Mangala, Robb, Merlin L, Kim, Jerome H, Marovich, Mary A, Goudsmit, Jaap, and Michael, Nelson L
- Subjects
AIDS Vaccines: immunology ,Adenoviridae: genetics ,immunology ,Animals ,Antibodies ,Neutralizing: immunology ,Enzyme-Linked Immunosorbent Assay ,Female ,HIV-1: immunology ,Macaca mulatta: immunology ,Male ,Neutralization Tests ,SAIDS Vaccines: immunology ,Simian immunodeficiency virus: immunology ,Viral Vaccines: immunology - Abstract
Preclinical studies of human immunodeficiency virus type 1 (HIV-1) vaccine candidates have typically shown post-infection virological control, but protection against acquisition of infection has previously only been reported against neutralization-sensitive virus challenges. Here we demonstrate vaccine protection against acquisition of fully heterologous, neutralization-resistant simian immunodeficiency virus (SIV) challenges in rhesus monkeys. Adenovirus/poxvirus and adenovirus/adenovirus-vector-based vaccines expressing SIV(SME543) Gag, Pol and Env antigens resulted in an 80% or greater reduction in the per-exposure probability of infection against repetitive, intrarectal SIV(MAC251) challenges in rhesus monkeys. Protection against acquisition of infection showed distinct immunological correlates compared with post-infection virological control and required the inclusion of Env in the vaccine regimen. These data demonstrate the proof-of-concept that optimized HIV-1 vaccine candidates can block acquisition of stringent, heterologous, neutralization-resistant virus challenges in rhesus monkeys.
- Published
- 2012
4. Development of novel replication-defective lymphocytic choriomeningitis virus vectors expressing SIV antigens
- Author
-
Penaloza MacMaster, Pablo, Shields, Jennifer L., Alayo, Quazim A., Cabral, Crystal, Jimenez, Jessica, Mondesir, Jade, Chandrashekar, Abishek, Cabral, Joseph M., Lim, Matthew, Iampietro, M. Justin, Provine, Nicholas M., Bricault, Christine A., Seaman, Michael, Orlinger, Klaus, Aspoeck, Andreas, Fuhrmann, Gerhard, Lilja, Anders E., Monath, Thomas, Mangeat, Bastien, Pinschewer, Daniel D., and Barouch, Dan H.
- Published
- 2017
- Full Text
- View/download PDF
5. Vaccine protection against Zika virus from Brazil
- Author
-
Larocca, Rafael A., Abbink, Peter, Peron, Jean Pierre S., de A. Zanotto, Paolo M., Iampietro, M. Justin, Badamchi-Zadeh, Alexander, Boyd, Michael, Nganga, David, Kirilova, Marinela, Nityanandam, Ramya, Mercado, Noe B., Li, Zhenfeng, Moseley, Edward T., Bricault, Christine A., Borducchi, Erica N., Giglio, Patricia B., Jetton, David, Neubauer, George, Nkolola, Joseph P., Maxfield, Lori F., De La Barrera, Rafael A., Jarman, Richard G., Eckels, Kenneth H., Michael, Nelson L., Thomas, Stephen J., and Barouch, Dan H.
- Subjects
Prevention ,Product development ,Zika virus infection -- Prevention ,Viral vaccines -- Product development - Abstract
Author(s): Rafael A. Larocca [1]; Peter Abbink [1]; Jean Pierre S. Peron [2]; Paolo M. de A. Zanotto [2]; M. Justin Iampietro [1]; Alexander Badamchi-Zadeh [1]; Michael Boyd [1]; David [...]
- Published
- 2016
- Full Text
- View/download PDF
6. Characterization of Humoral and Cellular Immune Responses Elicited by a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine in Healthy Adults (IPCAVD 001)
- Author
-
Barouch, Dan H., Liu, Jinyan, Peter, Lauren, Abbink, Peter, Iampietro, M. Justin, Cheung, Ann, Alter, Galit, Chung, Amy, Dugast, Anne-Sophie, Frahm, Nicole, McElrath, M. Juliana, Wenschuh, Holger, Reimer, Ulf, Seaman, Michael S., Pau, Maria G., Weijtens, Mo, Goudsmit, Jaap, Walsh, Stephen R., Dolin, Raphael, and Baden, Lindsey R.
- Published
- 2013
- Full Text
- View/download PDF
7. Combined HDAC and BET Inhibition Enhances Melanoma Vaccine Immunogenicity and Efficacy
- Author
-
Badamchi-Zadeh, Alexander, primary, Moynihan, Kelly D., additional, Larocca, Rafael A., additional, Aid, Malika, additional, Provine, Nicholas M., additional, Iampietro, M. Justin, additional, Kinnear, Ekaterina, additional, Penaloza-MacMaster, Pablo, additional, Abbink, Peter, additional, Blass, Eryn, additional, Tregoning, John S., additional, Irvine, Darrell J., additional, and Barouch, Dan H., additional
- Published
- 2018
- Full Text
- View/download PDF
8. Immunogenicity and Cross-Reactivity of Rhesus Adenoviral Vectors
- Author
-
Iampietro, M. Justin, primary, Larocca, Rafael A., additional, Provine, Nicholas M., additional, Abbink, Peter, additional, Kang, Zi Han, additional, Bricault, Christine A., additional, and Barouch, Dan H., additional
- Published
- 2018
- Full Text
- View/download PDF
9. Adenovirus serotype 5 vaccine vectors trigger IL-27–dependent inhibitory CD4 + T cell responses that impair CD8 + T cell function
- Author
-
Larocca, Rafael A., primary, Provine, Nicholas M., additional, Aid, Malika, additional, Iampietro, M. Justin, additional, Borducchi, Erica N., additional, Badamchi-Zadeh, Alexander, additional, Abbink, Peter, additional, Ng’ang’a, David, additional, Bricault, Christine A., additional, Blass, Eryn, additional, Penaloza-MacMaster, Pablo, additional, Stephenson, Kathryn E., additional, and Barouch, Dan H., additional
- Published
- 2016
- Full Text
- View/download PDF
10. Adenovirus 5 vaccination elicits IL-27, which impairs CD4 and CD8 T cell functionality (VAC12P.1113)
- Author
-
Larocca, Rafael, primary, Provine, Nicholas, additional, Iampietro, M., additional, Borducchi, Erica, additional, Penaloza-MacMaster, Pablo, additional, Bricault, Christine, additional, Blass, Eryn, additional, Parenteau, Lily, additional, Blackmore, Stephen, additional, Stephenson, Kathryn, additional, and Barouch, Dan, additional
- Published
- 2015
- Full Text
- View/download PDF
11. Construction and Evaluation of Novel Rhesus Monkey Adenovirus Vaccine Vectors
- Author
-
Abbink, Peter, primary, Maxfield, Lori F., additional, Ng'ang'a, David, additional, Borducchi, Erica N., additional, Iampietro, M. Justin, additional, Bricault, Christine A., additional, Teigler, Jeffrey E., additional, Blackmore, Stephen, additional, Parenteau, Lily, additional, Wagh, Kshitij, additional, Handley, Scott A., additional, Zhao, Guoyan, additional, Virgin, Herbert W., additional, Korber, Bette, additional, and Barouch, Dan H., additional
- Published
- 2015
- Full Text
- View/download PDF
12. Inhibition of Breast Cancer Cell Proliferation through Disturbance of the Calcineurin/NFAT Pathway by Human Herpesvirus 6B U54 Tegument Protein
- Author
-
Iampietro, M., primary, Gravel, A., additional, and Flamand, L., additional
- Published
- 2014
- Full Text
- View/download PDF
13. Adenovirus serotype 5 vaccine vectors trigger IL-27-dependent inhibitory CD4+ T cell responses that impair CD8+ T cell function.
- Author
-
Larocca, Rafael A., Provine, Nicholas M., Aid, Malika, Iampietro, M. Justin, Borducchi, Erica N., Badamchi-Zadeh, Alexander, Abbink, Peter, Ng'ang'a, David, Bricault, Christine A., Blass, Eryn, Penaloza-MacMaster, Pablo, Stephenson, Kathryn E., and Barouch, Dan H.
- Published
- 2016
- Full Text
- View/download PDF
14. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine (IPCAVD 001)
- Author
-
Baden, Lindsey R., primary, Walsh, Stephen R., additional, Seaman, Michael S., additional, Tucker, Robert P., additional, Krause, Kathleen H., additional, Patel, Alka, additional, Johnson, Jennifer A., additional, Kleinjan, Jane, additional, Yanosick, Katherine E., additional, Perry, James, additional, Zablowsky, Elise, additional, Abbink, Peter, additional, Peter, Lauren, additional, Iampietro, M. Justin, additional, Cheung, Ann, additional, Pau, Maria G., additional, Weijtens, Mo, additional, Goudsmit, Jaap, additional, Swann, Edith, additional, Wolff, Mark, additional, Loblein, Hayley, additional, Dolin, Raphael, additional, and Barouch, Dan H., additional
- Published
- 2012
- Full Text
- View/download PDF
15. Characterization of Humoral and Cellular Immune Responses Elicited by a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine in Healthy Adults (IPCAVD 001)
- Author
-
Barouch, Dan H., primary, Liu, Jinyan, additional, Peter, Lauren, additional, Abbink, Peter, additional, Iampietro, M. Justin, additional, Cheung, Ann, additional, Alter, Galit, additional, Chung, Amy, additional, Dugast, Anne-Sophie, additional, Frahm, Nicole, additional, McElrath, M. Juliana, additional, Wenschuh, Holger, additional, Reimer, Ulf, additional, Seaman, Michael S., additional, Pau, Maria G., additional, Weijtens, Mo, additional, Goudsmit, Jaap, additional, Walsh, Stephen R., additional, Dolin, Raphael, additional, and Baden, Lindsey R., additional
- Published
- 2012
- Full Text
- View/download PDF
16. Vaccination with Adenovirus Serotypes 35, 26, and 48 Elicits Higher Levels of Innate Cytokine Responses than Adenovirus Serotype 5 in Rhesus Monkeys
- Author
-
Teigler, Jeffrey E., primary, Iampietro, M. Justin, additional, and Barouch, Dan H., additional
- Published
- 2012
- Full Text
- View/download PDF
17. Accelerated Heterologous Adenovirus Prime-Boost SIV Vaccine in Neonatal Rhesus Monkeys
- Author
-
Liu, Jinyan, primary, Li, Hualin, additional, Iampietro, M. Justin, additional, and Barouch, Dan H., additional
- Published
- 2012
- Full Text
- View/download PDF
18. Angiopoietin-like 4 promotes terminal chondrogenic differentiation
- Author
-
Mathieu, M., primary, Chuchana, P., additional, Iampietro, M., additional, Noël, D., additional, and Jorgensen, C., additional
- Published
- 2012
- Full Text
- View/download PDF
19. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine (IPCAVD 001).
- Author
-
Baden, Lindsey R., Walsh, Stephen R., Seaman, Michael S., Tucker, Robert P., Krause, Kathleen H., Patel, Alka, Johnson, Jennifer A., Kleinjan, Jane, Yanosick, Katherine E., Perry, James, Zablowsky, Elise, Abbink, Peter, Peter, Lauren, Iampietro, M. Justin, Cheung, Ann, Pau, Maria G., Weijtens, Mo, Goudsmit, Jaap, Swann, Edith, and Wolff, Mark
- Subjects
HIV infections ,THERAPEUTICS ,ADENOVIRUSES ,VIRAL vaccines ,ENZYME-linked immunosorbent assay ,DRUG development ,BLOOD cells ,MEDICAL statistics - Abstract
Background. We report the first-in-human safety and immunogenicity assessment of a prototype Ad26 vector-based human immunodeficiency virus (HIV) vaccine in humans.Methods. Sixty Ad26-seronegative, healthy, HIV-uninfected subjects were enrolled in a randomized, double-blinded, placebo-controlled, dose-escalation phase 1 study. Five groups of 12 subjects received 109–1011 vp of the Ad26-EnvA vaccine (N = 10/group) or placebo (N = 2/group) at weeks 0 and 24 or weeks 0, 4, and 24. Safety and immunogenicity were assessed.Results. Self-limited reactogenicity was observed after the initial immunization at the highest (1011 vp) dose. No product-related SAEs were observed. All subjects who received the Ad26-EnvA vaccine developed Ad26 NAb titers, EnvA-specific enzyme-linked immunosorbent assays (ELISA) titers, and EnvA-specific enzyme-linked immunospot assays (ELISPOT) responses. These responses persisted at week 52. At week 28 in the 109, 1010, 1011 vp 3-dose and the 1010 and 5 × 1010 vp 2-dose groups, geometric mean EnvA ELISA titers were 6113, 12 470, 8545, 3470, and 9655 and mean EnvA ELISPOT responses were 397, 178, 736, 196, and 1311 SFC/106 peripheral blood mononuclear cells, respectively.Conclusion. This Ad26 vectored vaccine was generally safe and immunogenic at all doses tested. Reactogenicity was minimal with doses of 5 × 1010 vp or less. Ad26 is a promising new vaccine vector for HIV-1.Clinical Trials Registration. NCT00618605. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
20. Accelerated heterologous prime-boost adenovirus vector-based SIV vaccine in neonatal rhesus monkeys.
- Author
-
J. Liu, H. Li, Iampietro, M., and Barouch, D. H.
- Subjects
ABSTRACTS ,ADENOVIRUSES - Abstract
An abstract of the conference paper "Accelerated heterologous prime-boost adenovirus vector-based SIV vaccine in neonatal rhesus monkeys," by J. Liu and colleagues are presented.
- Published
- 2012
- Full Text
- View/download PDF
21. Accelerated Heterologous Adenovirus Prime-Boost SIV Vaccine in Neonatal Rhesus Monkeys.
- Author
-
Jinyan Liu, Hualin Li, Iampietro, M. Justin, and Barouch, Dan H.
- Subjects
- *
ADENOVIRUS diseases , *LABORATORY monkeys , *VIRAL vaccines , *HIV , *VIRAL diseases in children , *HUMORAL immunity - Abstract
A pediatric human immunodeficiency virus type 1 (HIV-1) vaccine would be desirable to protect infants against HIV-1 transmission from breast-feeding. Such a vaccine would need to induce protective immunity at mucosal surfaces in neonates as soon as possible after birth. Recombinant adenovirus (rAd) vectors have been shown to elicit potent systemic and mucosal virus-specific immune responses in adult nonhuman primates and humans, but these vectors have not previously been comprehensively studied in infants. In this study, we demonstrate that a single injection of rAd26 encoding simian immunodeficiency virus mac239 (SIVmac239) Gag on the day of birth elicited detectable Gag-specific cellular immune responses in rhesus monkeys, but these responses were transient and waned quickly. In contrast, an accelerated heterologous prime-boost regimen involving administration of rAd35 at birth and rAd26 at 4 weeks of life elicited potent and durable Gag-specific cellular and humoral immune responses in neonatal rhesus monkeys, including mucosal responses that remained detectable at 1 year of age. These results suggest the potential of an accelerated heterologous rAd prime-boost regimen as a candidate HIV-1 vaccine for newborns. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. Development of nebulized inhalation delivery for fusion-inhibitory lipopeptides to protect non-human primates against Nipah-Bangladesh infection.
- Author
-
Reynard O, Iampietro M, Dumont C, Le Guellec S, Durand S, Moroso M, Brisebard E, Dhondt KP, Pelissier R, Mathieu C, Cabrera M, Le Pennec D, Amurri L, Alabi C, Cardinaud S, Porotto M, Moscona A, Vecellio L, and Horvat B
- Abstract
Nipah virus (NiV) is a lethal zoonotic paramyxovirus that can be transmitted from person to person through the respiratory route. There are currently no licensed vaccines or therapeutics. A lipopeptide-based fusion inhibitor was developed and previously evaluated for efficacy against the NiV-Malaysia strain. Intraperitoneal administration in hamsters showed superb prophylactic activity and promising efficacy, however the intratracheal delivery mode in non-human primates proved intractable and spurred the development of an aerosolized delivery route that could be clinically applicable. We developed an aerosol delivery system in an artificial respiratory 3D model and optimized the combinations of flow rate and particle size for lung deposition. We characterized the nebulizer device and assessed the safety of lipopeptide nebulization in an African green monkey model that mimics human NiV infection. Three nebulized doses of fusion-inhibitory lipopeptide were administered every 24 h, resulting in peptide deposition across multiple regions of both lungs without causing toxicity or adverse hematological and biochemical effects. In peptide-treated monkeys challenged with a lethal dose of NiV-Bangladesh, animals retained robust levels of T and B-lymphocytes in the blood, infection-induced lethality was significantly delayed, and 2 out of 5 monkeys were protected from NiV infection. The present study establishes the safety and feasibility of the nebulizer delivery method for AGM studies. Future studies will compare delivery methods using next-generation fusion-inhibitory anti-NiV lipopeptides to evaluate the potential role of this aerosol delivery approach in achieving a rapid antiviral response., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: S. Le Guellec is employed by DTF Medical (Saint Etienne, France) and L. Vecellio was employed by DTF Medical from 2001 to 2018 and by Nemera (La Verpilliere, France) from 2018 to 2020. Moscona and M. Porotto are co-founders and have stock options in a start up company, Thylacine Biotherapeutics, set up to develop the lipopeptide antiviral platform. The remaining authors declare no competing interests., (Copyright © 2025 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
23. Multifaceted activation of STING axis upon Nipah and measles virus-induced syncytia formation.
- Author
-
Amurri L, Dumont C, Pelissier R, Reynard O, Mathieu C, Spanier J, Pályi B, Déri D, Karkowski L, Gonzalez C, Skerra J, Kis Z, Kalinke U, Horvat B, and Iampietro M
- Subjects
- Animals, Mice, Measles virology, Measles metabolism, Measles immunology, Humans, Virus Replication physiology, Henipavirus Infections virology, Henipavirus Infections metabolism, Henipavirus Infections immunology, Phosphoproteins metabolism, Nuclear Proteins metabolism, Mice, Inbred C57BL, Measles virus physiology, Giant Cells virology, Giant Cells metabolism, Membrane Proteins metabolism, Nipah Virus physiology
- Abstract
Activation of the DNA-sensing STING axis by RNA viruses plays a role in antiviral response through mechanisms that remain poorly understood. Here, we show that the STING pathway regulates Nipah virus (NiV) replication in vivo in mice. Moreover, we demonstrate that following both NiV and measles virus (MeV) infection, IFNγ-inducible protein 16 (IFI16), an alternative DNA sensor in addition to cGAS, induces the activation of STING, leading to the phosphorylation of NF-κB p65 and the production of IFNβ and interleukin 6. Finally, we found that paramyxovirus-induced syncytia formation is responsible for loss of mitochondrial membrane potential and leakage of mitochondrial DNA in the cytoplasm, the latter of which is further detected by both cGAS and IFI16. These results contribute to improve our understanding about NiV and MeV immunopathogenesis and provide potential paths for alternative therapeutic strategies., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Amurri et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
24. A vaccine targeting antigen-presenting cells through CD40 induces protective immunity against Nipah disease.
- Author
-
Pastor Y, Reynard O, Iampietro M, Surenaud M, Picard F, El Jahrani N, Lefebvre C, Hammoudi A, Dupaty L, Brisebard É, Reynard S, Moureaux É, Moroso M, Durand S, Gonzalez C, Amurri L, Gallouët AS, Marlin R, Baize S, Chevillard E, Raoul H, Hocini H, Centlivre M, Thiébaut R, Horvat B, Godot V, Lévy Y, and Cardinaud S
- Subjects
- Animals, Chlorocebus aethiops, T-Lymphocytes, Antibody Formation, Antigen-Presenting Cells, Virus Replication, Viral Vaccines
- Abstract
Nipah virus (NiV) has been recently ranked by the World Health Organization as being among the top eight emerging pathogens likely to cause major epidemics, whereas no therapeutics or vaccines have yet been approved. We report a method to deliver immunogenic epitopes from NiV through the targeting of the CD40 receptor of antigen-presenting cells by fusing a selected humanized anti-CD40 monoclonal antibody to the Nipah glycoprotein with conserved NiV fusion and nucleocapsid peptides. In the African green monkey model, CD40.NiV induces specific immunoglobulin A (IgA) and IgG as well as cross-neutralizing responses against circulating NiV strains and Hendra virus and T cell responses. Challenge experiments using a NiV-B strain demonstrate the high protective efficacy of the vaccine, with all vaccinated animals surviving and showing no significant clinical signs or virus replication, suggesting that the CD40.NiV vaccine conferred sterilizing immunity. Overall, results obtained with the CD40.NiV vaccine are highly promising in terms of the breadth and efficacy against NiV., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. Setup of Aerosol Delivery System to Prevent Measles Virus Infection in Nonhuman Primate Model.
- Author
-
Reynard O, Montharu J, Iampietro M, Gonzalez C, Le Guellec S, Horvat B, and Vecellio L
- Subjects
- Animals, Lipopeptides administration & dosage, Humans, Drug Delivery Systems methods, Measles prevention & control, Measles virus, Aerosols, Disease Models, Animal
- Abstract
Measles virus is one of the most contagious airborne human viruses which keeps causing outbreaks in numerous countries over the world despite the existence of an efficient vaccine. Fusion inhibitory lipopeptides were shown to inhibit viral entry into target cells, and their adequate administration into the respiratory tract may provide a novel preventive approach against airborne infections. Aerosol delivery presents the best administration route to deliver such preventive compounds to the upper and lower respiratory tract. This approach offers a conceptually new strategy to protect the population at risk against infection by respiratory viruses, including measles. It is a noninvasive needle-free approach, which may be used when antiviral protection is required, without any medical assistance. In this chapter, we describe the nebulization approach of lipopeptide compounds in nonhuman primates and the subsequent measles virus challenge., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
26. Interplay of Ebola Virus With Immune Cells Leading to Their Death by Diverse Mechanisms.
- Author
-
Iampietro M, Amurri L, Reynard O, and Bukreyev A
- Subjects
- Humans, T-Lymphocytes metabolism, Apoptosis, Cell Death, Ebolavirus, Hemorrhagic Fever, Ebola
- Abstract
Inflammation and cytopenia are commonly observed during Ebola virus (EBOV) infection; however, mechanisms responsible for EBOV-induced cell death remain obscure. While apoptosis and necrosis are already identified as mechanisms of cell death induced by the virus, our study demonstrates that THP-1 monocytes and SupT1 T cells exposed to EBOV undergo pyroptosis and necroptosis, respectively, through a direct contact with EBOV, and also mediate pyroptosis or necroptosis of uninfected bystander cells via indirect effects associated with secreted soluble factors. These results emphasize novel aspects of interactions between EBOV and immune cell populations and provide a better understanding of the immunopathogenesis of EBOV disease., Competing Interests: Potential conflicts of interest. The authors do not have a commercial or other association that might pose a conflict of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
27. SARS-CoV-2 awakens ancient retroviral genes and the expression of proinflammatory HERV-W envelope protein in COVID-19 patients.
- Author
-
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, and Horvat B
- 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., Competing Interests: HP, BC, JB, JP, and NQ receive compensation from GeNeuro-Innovation for their work. PK received consulting fees from GeNeuro SA. Other co-authors do not declare conflict of interest., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
28. Interplay between RNA viruses and cGAS/STING axis in innate immunity.
- Author
-
Amurri L, Horvat B, and Iampietro M
- Subjects
- Humans, Nucleotidyltransferases metabolism, Signal Transduction, Immunity, Innate, RNA Viruses
- Abstract
While the function of cGAS/STING signalling axis in the innate immune response to DNA viruses is well deciphered, increasing evidence demonstrates its significant contribution in the control of RNA virus infections. After the first evidence of cGAS/STING antagonism by flaviviruses, STING activation has been detected following infection by various enveloped RNA viruses. It has been discovered that numerous viral families have implemented advanced strategies to antagonize STING pathway through their evolutionary path. This review summarizes the characterized cGAS/STING escape strategies to date, together with the proposed mechanisms of STING signalling activation perpetrated by RNA viruses and discusses possible therapeutic approaches. Further studies regarding the interaction between RNA viruses and cGAS/STING-mediated immunity could lead to major discoveries important for the understanding of immunopathogenesis and for the treatment of RNA viral infections., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Amurri, Horvat and Iampietro.)
- Published
- 2023
- Full Text
- View/download PDF
29. Early Permissiveness of Central Nervous System Cells to Measles Virus Infection Is Determined by Hyperfusogenicity and Interferon Pressure.
- Author
-
Ferren M, Lalande A, Iampietro M, Canus L, Decimo D, Gerlier D, Porotto M, and Mathieu C
- Subjects
- Animals, Cricetinae, Humans, Brain, Interferons metabolism, Viral Fusion Proteins genetics, Central Nervous System virology, Measles, Measles virus physiology
- Abstract
The cessation of measles virus (MeV) vaccination in more than 40 countries as a consequence of the COVID-19 pandemic is expected to significantly increase deaths due to measles. MeV can infect the central nervous system (CNS) and lead to lethal encephalitis. Substantial part of virus sequences recovered from patients' brain were mutated in the matrix and/or the fusion protein (F). Mutations of the heptad repeat domain located in the C terminal (HRC) part of the F protein were often observed and were associated to hyperfusogenicity. These mutations promote brain invasion as a hallmark of neuroadaptation. Wild-type F allows entry into the brain, followed by limited spreading compared with the massive invasion observed for hyperfusogenic MeV. Taking advantage of our ex vivo models of hamster organotypic brain cultures, we investigated how the hyperfusogenic mutations in the F HRC domain modulate virus distribution in CNS cells. In this study, we also identified the dependence of neural cells susceptibility on both their activation state and destabilization of the virus F protein. Type I interferon (IFN-I) impaired mainly astrocytes and microglial cells permissiveness contrarily to neurons, opening a new way of consideration on the development of treatments against viral encephalitis.
- Published
- 2023
- Full Text
- View/download PDF
30. Mouse Models of Henipavirus Infection.
- Author
-
Iampietro M, Barron S, Duthey A, and Horvat B
- Subjects
- Humans, Animals, Mice, Disease Models, Animal, Henipavirus Infections
- Abstract
The Nipah and Hendra viruses, belonging to henipavirus genus, are recently emerged zoonotic pathogens that cause severe and often fatal, neurologic, and/or respiratory diseases in both humans and various animals. As mice represent a small animal model convenient to study viral infections and provide a well-developed experimental toolbox for analysis in immunovirology, we describe in this chapter a few basic methods used in biosafety 4 level (BSL4) conditions to study henipavirus infection in mice., (© 2023. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
31. Measles Virus-Induced Host Immunity and Mechanisms of Viral Evasion.
- Author
-
Amurri L, Reynard O, Gerlier D, Horvat B, and Iampietro M
- Subjects
- Humans, Interferons, Signal Transduction, Immune Evasion, Immunity, Innate, Measles immunology, Measles virus
- Abstract
The immune system deploys a complex network of cells and signaling pathways to protect host integrity against exogenous threats, including measles virus (MeV). However, throughout its evolutionary path, MeV developed various mechanisms to disrupt and evade immune responses. Despite an available vaccine, MeV remains an important re-emerging pathogen with a continuous increase in prevalence worldwide during the last decade. Considerable knowledge has been accumulated regarding MeV interactions with the innate immune system through two antagonistic aspects: recognition of the virus by cellular sensors and viral ability to inhibit the induction of the interferon cascade. Indeed, while the host could use several innate adaptors to sense MeV infection, the virus is adapted to unsettle defenses by obstructing host cell signaling pathways. Recent works have highlighted a novel aspect of innate immune response directed against MeV unexpectedly involving DNA-related sensing through activation of the cGAS/STING axis, even in the absence of any viral DNA intermediate. In addition, while MeV infection most often causes a mild disease and triggers a lifelong immunity, its tropism for invariant T-cells and memory T and B-cells provokes the elimination of one primary shield and the pre-existing immunity against previously encountered pathogens, known as "immune amnesia".
- Published
- 2022
- Full Text
- View/download PDF
32. Nebulized fusion inhibitory peptide protects cynomolgus macaques from measles virus infection.
- Author
-
Reynard O, Gonzalez C, Dumont C, Iampietro M, Ferren M, Le Guellec S, Laurie L, Mathieu C, Carpentier G, Roseau G, Bovier FT, Zhu Y, Le Pennec D, Montharu J, Addetia A, Greninger AL, Alabi CA, Brisebard E, Moscona A, Vecellio L, Porotto M, and Horvat B
- Subjects
- Animals, Humans, Measles virus, SARS-CoV-2, Viral Fusion Proteins metabolism, Peptides pharmacology, Macaca fascicularis metabolism, COVID-19 prevention & control, Measles prevention & control
- Abstract
Measles is the most contagious airborne viral infection and the leading cause of child death among vaccine-preventable diseases. We show here that aerosolized lipopeptide fusion inhibitor, derived from heptad-repeat regions of the measles virus (MeV) fusion protein, blocks respiratory MeV infection in a non-human primate model, the cynomolgus macaque. We use a custom-designed mesh nebulizer to ensure efficient aerosol delivery of peptide to the respiratory tract and demonstrate the absence of adverse effects and lung pathology in macaques. The nebulized peptide efficiently prevents MeV infection, resulting in the complete absence of MeV RNA, MeV-infected cells, and MeV-specific humoral responses in treated animals. This strategy provides an additional means to fight against respiratory infection in non-vaccinated people, that can be readily translated to human trials. It presents a proof-of-concept for the aerosol delivery of fusion inhibitory peptides to protect against measles and other airborne viruses, including SARS-CoV-2, in case of high-risk exposure., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
33. Hamster organotypic kidney culture model of early-stage SARS-CoV-2 infection highlights a two-step renal susceptibility.
- Author
-
Shyfrin SR, Ferren M, Perrin-Cocon L, Espi M, Charmetant X, Brailly M, Decimo D, Iampietro M, Canus L, Horvat B, Lotteau V, Vidalain PO, Thaunat O, and Mathieu C
- Abstract
Kidney pathology is frequently reported in patients hospitalized with COVID-19, the pandemic disease caused by the Severe acute respiratory coronavirus 2 (SARS-CoV-2). However, due to a lack of suitable study models, the events occurring in the kidney during the earliest stages of infection remain unknown. We have developed hamster organotypic kidney cultures (OKCs) to study the early stages of direct renal infection. OKCs maintained key renal structures in their native three-dimensional arrangement. SARS-CoV-2 productively replicated in hamster OKCs, initially targeting endothelial cells and later disseminating into proximal tubules. We observed a delayed interferon response, markers of necroptosis and pyroptosis, and an early repression of pro-inflammatory cytokines transcription followed by a strong later upregulation. While it remains an open question whether an active replication of SARS-CoV-2 takes place in the kidneys of COVID-19 patients with AKI, our model provides new insights into the kinetics of SARS-CoV-2 kidney infection and can serve as a powerful tool for studying kidney infection by other pathogens and testing the renal toxicity of drugs., Competing Interests: Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article., (© The Author(s) 2022.)
- Published
- 2022
- Full Text
- View/download PDF
34. [The cGAS/STING signaling pathway controls infections by measles and Nipah viruses].
- Author
-
Iampietro M, Amurri L, and Horvat B
- Subjects
- Humans, Nucleotidyltransferases, Signal Transduction, Measles epidemiology, Nipah Virus metabolism
- Published
- 2022
- Full Text
- View/download PDF
35. Nipah virus W protein harnesses nuclear 14-3-3 to inhibit NF-κB-induced proinflammatory response.
- Author
-
Enchéry F, Dumont C, Iampietro M, Pelissier R, Aurine N, Bloyet LM, Carbonnelle C, Mathieu C, Journo C, Gerlier D, and Horvat B
- Subjects
- Animals, Cell Line, Cell Nucleus immunology, Chlorocebus aethiops, HEK293 Cells, HeLa Cells, Humans, NF-kappa B, Nipah Virus genetics, Immunity, Innate physiology, Nipah Virus physiology, Signal Transduction immunology, Viral Proteins metabolism
- Abstract
Nipah virus (NiV) is a highly pathogenic emerging bat-borne Henipavirus that has caused numerous outbreaks with public health concerns. It is able to inhibit the host innate immune response. Since the NF-κB pathway plays a crucial role in the innate antiviral response as a major transcriptional regulator of inflammation, we postulated its implication in the still poorly understood NiV immunopathogenesis. We report here that NiV inhibits the canonical NF-κB pathway via its nonstructural W protein. Translocation of the W protein into the nucleus causes nuclear accumulation of the cellular scaffold protein 14-3-3 in both African green monkey and human cells infected by NiV. Excess of 14-3-3 in the nucleus was associated with a reduction of NF-κB p65 subunit phosphorylation and of its nuclear accumulation. Importantly, W-S449A substitution impairs the binding of the W protein to 14-3-3 and the subsequent suppression of NF-κB signaling, thus restoring the production of proinflammatory cytokines. Our data suggest that the W protein increases the steady-state level of 14-3-3 in the nucleus and consequently enhances 14-3-3-mediated negative feedback on the NF-κB pathway. These findings provide a mechanistic model of W-mediated disruption of the host inflammatory response, which could contribute to the high severity of NiV infection., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
36. Hamster organotypic modeling of SARS-CoV-2 lung and brainstem infection.
- Author
-
Ferren M, Favède V, Decimo D, Iampietro M, Lieberman NAP, Weickert JL, Pelissier R, Mazelier M, Terrier O, Moscona A, Porotto M, Greninger AL, Messaddeq N, Horvat B, and Mathieu C
- Subjects
- Adenosine Monophosphate analogs & derivatives, Adenosine Monophosphate pharmacology, Alanine analogs & derivatives, Alanine pharmacology, Alveolar Epithelial Cells virology, Animals, Antiviral Agents pharmacology, Brain Stem cytology, Brain Stem immunology, Brain Stem pathology, Cricetinae, Immunity, Innate, Inflammation, Lung cytology, Lung immunology, Lung pathology, Neurons virology, Organ Culture Techniques, Regulated Cell Death, SARS-CoV-2 drug effects, Viral Tropism, Brain Stem virology, Lung virology, Models, Biological, SARS-CoV-2 pathogenicity
- Abstract
SARS-CoV-2 has caused a global pandemic of COVID-19 since its emergence in December 2019. The infection causes a severe acute respiratory syndrome and may also spread to central nervous system leading to neurological sequelae. We have developed and characterized two new organotypic cultures from hamster brainstem and lung tissues that offer a unique opportunity to study the early steps of viral infection and screening antivirals. These models are not dedicated to investigate how the virus reaches the brain. However, they allow validating the early tropism of the virus in the lungs and demonstrating that SARS-CoV-2 could infect the brainstem and the cerebellum, mainly by targeting granular neurons. Viral infection induces specific interferon and innate immune responses with patterns specific to each organ, along with cell death by apoptosis, necroptosis, and pyroptosis. Overall, our data illustrate the potential of rapid modeling of complex tissue-level interactions during infection by a newly emerged virus., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
37. Hydroxyurea Use Associated with Nonverbal and Executive Skills in Sickle Cell Anemia.
- Author
-
Tarazi RA, Patrick KE, Iampietro M, and Apollonsky N
- Subjects
- Child, Child, Preschool, Executive Function, Fetal Hemoglobin, Hemoglobin, Sickle, Humans, Infant, Infant, Newborn, Anemia, Sickle Cell drug therapy, Hydroxyurea therapeutic use
- Abstract
Objective: Hydroxyurea (HU) is used in children with sickle cell disease (SCD) to increase fetal hemoglobin (HF), contributing to a decrease in physical symptoms and potential protection against cerebral microvasculopathy. There has been minimal investigation into the association between HU use and cognition in this population. This study examined the relationship between HU status and cognition in children with SCD., Methods: Thirty-seven children with SCD HbSS or HbS/β0 thalassaemia (sickle cell anemia; SCA) ages 4:0-11 years with no history of overt stroke or chronic transfusion completed a neuropsychological test battery. Other medical, laboratory, and demographic data were obtained. Neuropsychological function across 3 domains (verbal, nonverbal, and attention/executive) was compared for children on HU (n = 9) to those not taking HU (n = 28)., Results: Children on HU performed significantly better than children not taking HU on standardized measures of attention/executive functioning and nonverbal skills. Performance on verbal measures was similar between groups., Conclusions: These results suggest that treatment with HU may not only reduce physical symptoms, but may also provide potential benefit to cognition in children with SCA, particularly in regard to attention/executive functioning and nonverbal skills. Replication with larger samples and longitudinal studies are warranted., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Society of Pediatric Psychology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
38. Activation of cGAS/STING pathway upon paramyxovirus infection.
- Author
-
Iampietro M, Dumont C, Mathieu C, Spanier J, Robert J, Charpenay A, Dupichaud S, Dhondt KP, Aurine N, Pelissier R, Ferren M, Mély S, Gerlier D, Kalinke U, and Horvat B
- Abstract
During inflammatory diseases, cancer, and infection, the cGAS/STING pathway is known to recognize foreign or self-DNA in the cytosol and activate an innate immune response. Here, we report that negative-strand RNA paramyxoviruses, Nipah virus (NiV), and measles virus (MeV), can also trigger the cGAS/STING axis. Although mice deficient for MyD88, TRIF, and MAVS still moderately control NiV infection when compared with wild-type mice, additional STING deficiency resulted in 100% lethality, suggesting synergistic roles of these pathways in host protection. Moreover, deletion of cGAS or STING resulted in decreased type I interferon production with enhanced paramyxoviral infection in both human and murine cells. Finally, the phosphorylation and ubiquitination of STING, observed during viral infections, confirmed the activation of cGAS/STING pathway by NiV and MeV. Our data suggest that cGAS/STING activation is critical in controlling paramyxovirus infection and possibly represents attractive targets to develop countermeasures against severe disease induced by these pathogens., Competing Interests: The authors declare no competing interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
39. Quercetin Blocks Ebola Virus Infection by Counteracting the VP24 Interferon-Inhibitory Function.
- Author
-
Fanunza E, Iampietro M, Distinto S, Corona A, Quartu M, Maccioni E, Horvat B, and Tramontano E
- Subjects
- Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Humans, Viral Proteins antagonists & inhibitors, Ebolavirus drug effects, Hemorrhagic Fever, Ebola drug therapy, Interferons, Quercetin pharmacology
- Abstract
Ebola virus (EBOV) is among the most devastating pathogens causing fatal hemorrhagic fever in humans. The epidemics from 2013 to 2016 resulted in more than 11,000 deaths, and another outbreak is currently ongoing. Since there is no FDA-approved drug so far to fight EBOV infection, there is an urgent need to focus on drug discovery. Considering the tight correlation between the high EBOV virulence and its ability to suppress the type I interferon (IFN-I) system, identifying molecules targeting viral protein VP24, one of the main virulence determinants blocking the IFN response, is a promising novel anti-EBOV therapy approach. Hence, in the effort to find novel EBOV inhibitors, a screening of a small set of flavonoids was performed; it showed that quercetin and wogonin can suppress the VP24 effect on IFN-I signaling inhibition. The mechanism of action of the most active compound, quercetin, showing a half-maximal inhibitory concentration (IC
50 ) of 7.4 μM, was characterized to significantly restore the IFN-I signaling cascade, blocked by VP24, by directly interfering with the VP24 binding to karyopherin-α and thus restoring P-STAT1 nuclear transport and IFN gene transcription. Quercetin significantly blocked viral infection, specifically targeting EBOV VP24 anti-IFN-I function. Overall, quercetin is the first identified inhibitor of the EBOV VP24 anti-IFN function, representing a molecule interacting with a viral binding site that is very promising for further drug development aiming to block EBOV infection at the early steps., (Copyright © 2020 American Society for Microbiology.)- Published
- 2020
- Full Text
- View/download PDF
40. Control of Nipah Virus Infection in Mice by the Host Adaptors Mitochondrial Antiviral Signaling Protein (MAVS) and Myeloid Differentiation Primary Response 88 (MyD88).
- Author
-
Iampietro M, Aurine N, Dhondt KP, Dumont C, Pelissier R, Spanier J, Vallve A, Raoul H, Kalinke U, and Horvat B
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, DEAD Box Protein 58 genetics, DEAD Box Protein 58 metabolism, Gene Expression Regulation immunology, Interferon Type I genetics, Interferon Type I metabolism, Interferon-beta genetics, Interferon-beta metabolism, Mice, Mice, Knockout, Mice, Transgenic, Myeloid Differentiation Factor 88 genetics, Toll-Like Receptors genetics, Toll-Like Receptors metabolism, Adaptor Proteins, Signal Transducing metabolism, Henipavirus Infections immunology, Henipavirus Infections virology, Myeloid Differentiation Factor 88 metabolism, Nipah Virus
- Abstract
Interferon (IFN) type I plays a critical role in the protection of mice from lethal Nipah virus (NiV) infection, but mechanisms responsible for IFN-I induction remain unknown. In the current study, we demonstrated the critical role of the mitochondrial antiviral signaling protein signaling pathway in IFN-I production and NiV replication in murine embryonic fibroblasts in vitro, and the redundant but essential roles of both mitochondrial antiviral signaling protein and myeloid differentiation primary response 88 adaptors, but not toll/interleukin-1 receptor/resistance [TIR] domain-containing adaptor-inducing IFN-β (TRIF), in the control of NiV infection in mice. These results reveal potential novel targets for antiviral intervention and help in understanding NiV immunopathogenesis., (© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
41. Ebola virus-mediated T-lymphocyte depletion is the result of an abortive infection.
- Author
-
Younan P, Santos RI, Ramanathan P, Iampietro M, Nishida A, Dutta M, Ammosova T, Meyer M, Katze MG, Popov VL, Nekhai S, and Bukreyev A
- Subjects
- Animals, Antigens, Viral biosynthesis, Antigens, Viral genetics, Autophagy physiology, CD4-Positive T-Lymphocytes immunology, Cell Line, Chlorocebus aethiops, Endoplasmic Reticulum Stress physiology, HEK293 Cells, Host-Pathogen Interactions, Humans, Indoles pharmacology, Jurkat Cells, Protein Phosphatase 1 antagonists & inhibitors, RNA Interference, RNA, Small Interfering genetics, RNA, Viral biosynthesis, RNA, Viral genetics, Transcription Factors metabolism, Urea analogs & derivatives, Urea pharmacology, Vero Cells, Viral Proteins metabolism, CD4-Positive T-Lymphocytes virology, Ebolavirus immunology, Hemorrhagic Fever, Ebola immunology, Lymphopenia immunology, Virus Replication physiology
- Abstract
Ebola virus (EBOV) infections are characterized by a pronounced lymphopenia that is highly correlative with fatalities. However, the mechanisms leading to T-cell depletion remain largely unknown. Here, we demonstrate that both viral mRNAs and antigens are detectable in CD4+ T cells despite the absence of productive infection. A protein phosphatase 1 inhibitor, 1E7-03, and siRNA-mediated suppression of viral antigens were used to demonstrate de novo synthesis of viral RNAs and antigens in CD4+ T cells, respectively. Cell-to-cell fusion of permissive Huh7 cells with non-permissive Jurkat T cells impaired productive EBOV infection suggesting the presence of a cellular restriction factor. We determined that viral transcription is partially impaired in the fusion T cells. Lastly, we demonstrate that exposure of T cells to EBOV resulted in autophagy through activation of ER-stress related pathways. These data indicate that exposure of T cells to EBOV results in an abortive infection, which likely contributes to the lymphopenia observed during EBOV infections., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
42. Recent advances in the understanding of Nipah virus immunopathogenesis and anti-viral approaches.
- Author
-
Pelissier R, Iampietro M, and Horvat B
- Subjects
- Animals, Bangladesh, Humans, India, Nipah Virus immunology, Adaptive Immunity, Henipavirus Infections immunology, Immunity, Innate, Nipah Virus pathogenicity
- Abstract
Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged at the end of last century as a human pathogen capable of causing severe acute respiratory infection and encephalitis. Although NiV provokes serious diseases in numerous mammalian species, the infection seems to be asymptomatic in NiV natural hosts, the fruit bats, which provide a continuous virus source for further outbreaks. Consecutive human-to-human transmission has been frequently observed during outbreaks in Bangladesh and India. NiV was shown to interfere with the innate immune response and interferon type I signaling, restraining the anti-viral response and permitting viral spread. Studies of adaptive immunity in infected patients and animal models have suggested an unbalanced immune response during NiV infection. Here, we summarize some of the recent studies of NiV pathogenesis and NiV-induced modulation of both innate and adaptive immune responses, as well as the development of novel prophylactic and therapeutic approaches, necessary to control this highly lethal emerging infection., Competing Interests: No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed., (Copyright: © 2019 Pelissier R et al.)
- Published
- 2019
- Full Text
- View/download PDF
43. Antibody-Mediated Protective Mechanisms Induced by a Trivalent Parainfluenza Virus-Vectored Ebolavirus Vaccine.
- Author
-
Kimble JB, Malherbe DC, Meyer M, Gunn BM, Karim MM, Ilinykh PA, Iampietro M, Mohamed KS, Negi S, Gilchuk P, Huang K, Wolf YI, Braun W, Crowe JE, Alter G, and Bukreyev A
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Cell Line, Drug Combinations, Epitopes immunology, Female, Ferrets, Genetic Vectors, Glycoproteins immunology, Guinea Pigs, Hemorrhagic Fever, Ebola virology, Parainfluenza Virus 3, Human genetics, Viral Vaccines genetics, Ebola Vaccines genetics, Ebolavirus genetics, Viral Envelope Proteins immunology
- Abstract
Ebolaviruses Zaire (EBOV), Bundibugyo (BDBV), and Sudan (SUDV) cause human disease with high case fatality rates. Experimental monovalent vaccines, which all utilize the sole envelope glycoprotein (GP), do not protect against heterologous ebolaviruses. Human parainfluenza virus type 3-vectored vaccines offer benefits, including needle-free administration and induction of mucosal responses in the respiratory tract. Multiple approaches were taken to induce broad protection against the three ebolaviruses. While GP consensus-based antigens failed to elicit neutralizing antibodies, polyvalent vaccine immunization induced neutralizing responses to all three ebolaviruses and protected animals from death and disease caused by EBOV, SUDV, and BDBV. As immunization with a cocktail of antigenically related antigens can skew the responses and change the epitope hierarchy, we performed comparative analysis of antibody repertoire and Fc-mediated protective mechanisms in animals immunized with monovalent versus polyvalent vaccines. Compared to sera from guinea pigs receiving the monovalent vaccines, sera from guinea pigs receiving the trivalent vaccine bound and neutralized EBOV and SUDV at equivalent levels and BDBV at only a slightly reduced level. Peptide microarrays revealed a preponderance of binding to amino acids 389 to 403, 397 to 415, and 477 to 493, representing three linear epitopes in the mucin-like domain known to induce a protective antibody response. Competition binding assays with monoclonal antibodies isolated from human ebolavirus infection survivors demonstrated that the immune sera block the binding of antibodies specific for the GP glycan cap, the GP1-GP2 interface, the mucin-like domain, and the membrane-proximal external region. Thus, administration of a cocktail of three ebolavirus vaccines induces a desirable broad antibody response, without skewing of the response toward preferential recognition of a single virus. IMPORTANCE The symptoms of the disease caused by the ebolaviruses Ebola, Bundibugyo, and Sudan are similar, and their areas of endemicity overlap. However, because of the limited antigenic relatedness of the ebolavirus glycoprotein (GP) used in all candidate vaccines against these viruses, they protect only against homologous and not against heterologous ebolaviruses. Therefore, a broadly specific pan-ebolavirus vaccine is required, and this might be achieved by administration of a cocktail of vaccines. The effects of cocktail administration of ebolavirus vaccines on the antibody repertoire remain unknown. Here, an in-depth analysis of the antibody responses to administration of a cocktail of human parainfluenza virus type 3-vectored vaccines against individual ebolaviruses was performed, which included analysis of binding to GP, neutralization of individual ebolaviruses, epitope specificity, Fc-mediated functions, and protection against the three ebolaviruses. The results demonstrated potent and balanced responses against individual ebolaviruses and no significant reduction of the responses compared to that induced by individual vaccines., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
44. Disruption of Phosphatidylserine Synthesis or Trafficking Reduces Infectivity of Ebola Virus.
- Author
-
Younan P, Iampietro M, Santos RI, Ramanathan P, Popov VL, and Bukreyev A
- Subjects
- Animals, Biological Transport, CDPdiacylglycerol-Serine O-Phosphatidyltransferase metabolism, Chlorocebus aethiops, Vero Cells, Virion physiology, Virus Release, Virus Replication, Ebolavirus pathogenicity, Phosphatidylserines physiology
- Abstract
The outer leaflet of the viral membrane of Ebola virus (EBOV) virions is enriched with phosphatidylserine (PtdSer), which is thought to play a central role in viral tropism, entry, and virus-associated immune evasion. We investigated the effects of inhibiting synthesis and/or export of PtdSer to the cell surface of infected cells on viral infectivity. Knockdown of both PtdSer synthase enzymes, PTDSS1 and PTDSS2, effectively decreased viral production. Decreased PtdSer expression resulted in an accumulation of virions at the plasma membrane and adjacent of intracellular organelles, suggesting that virion budding is impaired. The addition of inhibitors that block normal cellular trafficking of PtdSer to the plasma membrane resulted in a similar accumulation of virions and reduced viral replication. These findings demonstrate that plasma membrane-associated PtdSer is required for efficient EBOV budding, increasing EBOV infectivity, and could constitute a potential therapeutic target for the development of future countermeasures against EBOV.
- Published
- 2018
- Full Text
- View/download PDF
45. Ebola Virus Shed Glycoprotein Triggers Differentiation, Infection, and Death of Monocytes Through Toll-Like Receptor 4 Activation.
- Author
-
Iampietro M, Santos RI, Lubaki NM, and Bukreyev A
- Subjects
- Cell Death immunology, Cell Differentiation immunology, Ebola Vaccines immunology, Ebolavirus immunology, Glycoproteins immunology, Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola metabolism, Hemorrhagic Fever, Ebola virology, Host-Pathogen Interactions immunology, Host-Pathogen Interactions physiology, Humans, Monocytes immunology, Monocytes physiology, Monocytes virology, THP-1 Cells metabolism, THP-1 Cells physiology, THP-1 Cells virology, Viral Envelope Proteins metabolism, Cell Death physiology, Cell Differentiation physiology, Ebolavirus metabolism, Glycoproteins metabolism, Monocytes metabolism, Toll-Like Receptor 4 metabolism
- Abstract
A better understanding of the mechanisms used by Ebola virus to disable the host immune system and spread the infection are of great importance for development of new therapeutic strategies. We demonstrate that treatment of monocytic cells with Ebola virus shed glycoprotein (GP) promotes their differentiation resulting in increased infection and cell death. The effects were inhibited by blocking Toll-like receptor 4 pathway. In addition, high levels of shed GP were detected in supernatants of cells treated with Ebola vaccines. This study highlights the role of shed GP in Ebola pathogenesis and also in adverse effects associated with Ebola vaccines.
- Published
- 2018
- Full Text
- View/download PDF
46. Role of Transmembrane Protein 16F in the Incorporation of Phosphatidylserine Into Budding Ebola Virus Virions.
- Author
-
Younan P, Iampietro M, Santos RI, Ramanathan P, Popov VL, and Bukreyev A
- Abstract
Viral apoptotic mimicry, which is defined by exposure of phosphatidylserine (PtdSer) into the outer leaflet of budding enveloped viruses, increases viral tropism, infectivity and promotes immune evasion. Here, we report that the calcium (Ca2+)-dependent scramblase, transmembrane protein 16F (TMEM16F), is responsible for the incorporation of PtdSer into virion membranes during Ebola virus infection. Infection of Huh7 cells with Ebola virus resulted in a pronounced increase in plasma membrane-associated PtdSer, which was demonstrated to be dependent on TMEM16F function. Analysis of virions using imaging flow cytometry revealed that short hairpin RNA-mediated down-regulation of TMEM16F function directly reduced virion-associated PtdSer. Taken together, these studies demonstrate that TMEM16F is a central cellular factor in the exposure of PtdSer in the outer leaflet of viral membranes.
- Published
- 2018
- Full Text
- View/download PDF
47. Disabling of lymphocyte immune response by Ebola virus.
- Author
-
Younan P, Iampietro M, and Bukreyev A
- Subjects
- Hemorrhagic Fever, Ebola immunology, Hemorrhagic Fever, Ebola virology, Humans, Ebolavirus immunology, Hemorrhagic Fever, Ebola complications, Lymphocytes immunology, Lymphopenia etiology
- Published
- 2018
- Full Text
- View/download PDF
48. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm.
- Author
-
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, and Bukreyev A
- Subjects
- Animals, Blotting, Western, CD4-Positive T-Lymphocytes virology, Cell Line, Chemokines analysis, Culture Media, Cytokines biosynthesis, Cytokines blood, Gene Expression Profiling, Hepatitis A Virus Cellular Receptor 1 deficiency, Hepatitis A Virus Cellular Receptor 1 genetics, Host-Pathogen Interactions, Mice, Mice, Knockout, Phosphatidylserines metabolism, Receptors, Virus, Signal Transduction, T-Lymphocyte Subsets immunology, Virus Replication, CD4-Positive T-Lymphocytes immunology, Cytokines immunology, Ebolavirus physiology, Hepatitis A Virus Cellular Receptor 1 metabolism, Virus Attachment
- Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1
-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD. IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis.", (Copyright © 2017 Younan et al.)- Published
- 2017
- Full Text
- View/download PDF
49. Ebola virus glycoprotein directly triggers T lymphocyte death despite of the lack of infection.
- Author
-
Iampietro M, Younan P, Nishida A, Dutta M, Lubaki NM, Santos RI, Koup RA, Katze MG, and Bukreyev A
- Subjects
- CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes virology, Cell Death, Cells, Cultured, Ebolavirus genetics, Hemorrhagic Fever, Ebola genetics, Hemorrhagic Fever, Ebola metabolism, Hemorrhagic Fever, Ebola virology, Host-Pathogen Interactions, Humans, Protein Binding, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Viral Envelope Proteins genetics, CD4-Positive T-Lymphocytes cytology, Ebolavirus metabolism, Hemorrhagic Fever, Ebola physiopathology, Viral Envelope Proteins metabolism
- Abstract
Fatal outcomes of Ebola virus (EBOV) infections are typically preceded by a 'sepsis-like' syndrome and lymphopenia despite T cells being resistant to Ebola infection. The mechanisms that lead to T lymphocytes death remain largely unknown; however, the degree of lymphopenia is highly correlative with fatalities. Here we investigated whether the addition of EBOV or its envelope glycoprotein (GP) to isolated primary human CD4+ T cells induced cell death. We observed a significant decrease in cell viability in a GP-dependent manner, which is suggestive of a direct role of GP in T cell death. Using immunoprecipitation assays and flow cytometry, we demonstrate that EBOV directly binds to CD4+ T cells through interaction of GP with TLR4. Transcriptome analysis revealed that the addition of EBOV to CD4+ T cells results in the significant upregulation of pathways associated with interferon signaling, pattern recognition receptors and intracellular activation of NFκB signaling pathway. Both transcriptome analysis and specific inhibitors allowed identification of apoptosis and necrosis as mechanisms associated with the observed T cell death following exposure to EBOV. The addition of the TLR4 inhibitor CLI-095 significantly reduced CD4+ T cell death induced by GP. EBOV stimulation of primary CD4+ T cells resulted in a significant increase in secreted TNFα; inhibition of TNFα-mediated signaling events significantly reduced T cell death while inhibitors of both necrosis and apoptosis similarly reduced EBOV-induced T cell death. Lastly, we show that stimulation with EBOV or GP augments monocyte maturation as determined by an overall increase in expression levels of markers of differentiation. Subsequently, the increased rates of cellular differentiation resulted in higher rates of infection further contributing to T cell death. These results demonstrate that GP directly subverts the host's immune response by increasing the susceptibility of monocytes to EBOV infection and triggering lymphopenia through direct and indirect mechanisms.
- Published
- 2017
- Full Text
- View/download PDF
50. The Ebola Interferon Inhibiting Domains Attenuate and Dysregulate Cell-Mediated Immune Responses.
- Author
-
Lubaki NM, Younan P, Santos RI, Meyer M, Iampietro M, Koup RA, and Bukreyev A
- Subjects
- B-Lymphocytes immunology, Blotting, Western, CD4-Positive T-Lymphocytes immunology, Cell Separation, Dendritic Cells immunology, Ebolavirus immunology, Flow Cytometry, Humans, Leukocytes, Mononuclear virology, Microscopy, Confocal, Protein Domains immunology, Hemorrhagic Fever, Ebola immunology, Lymphocyte Activation immunology, Viral Proteins immunology, Viral Regulatory and Accessory Proteins immunology
- Abstract
Ebola virus (EBOV) infections are characterized by deficient T-lymphocyte responses, T-lymphocyte apoptosis and lymphopenia. We previously showed that disabling of interferon-inhibiting domains (IIDs) in the VP24 and VP35 proteins effectively unblocks maturation of dendritic cells (DCs) and increases the secretion of cytokines and chemokines. Here, we investigated the role of IIDs in adaptive and innate cell-mediated responses using recombinant viruses carrying point mutations, which disabled IIDs in VP24 (EBOV/VP24m), VP35 (EBOV/VP35m) or both (EBOV/VP35m/VP24m). Peripheral blood mononuclear cells (PBMCs) from cytomegalovirus (CMV)-seropositive donors were inoculated with the panel of viruses and stimulated with CMV pp65 peptides. Disabling of the VP35 IID resulted in increased proliferation and higher percentages of CD4+ T cells secreting IFNγ and/or TNFα. To address the role of aberrant DC maturation in the IID-mediated suppression of T cell responses, CMV-stimulated DCs were infected with the panel of viruses and co-cultured with autologous T-lymphocytes. Infection with EBOV/VP35m infection resulted in a significant increase, as compared to wt EBOV, in proliferating CD4+ cells secreting IFNγ, TNFα and IL-2. Experiments with expanded CMV-specific T cells demonstrated their increased activation following co-cultivation with CMV-pulsed DCs pre-infected with EBOV/VP24m, EBOV/VP35m and EBOV/VP35m/VP24m, as compared to wt EBOV. Both IIDs were found to block phosphorylation of TCR complex-associated adaptors and downstream signaling molecules. Next, we examined the effects of IIDs on the function of B cells in infected PBMC. Infection with EBOV/VP35m and EBOV/VP35m/VP24m resulted in significant increases in the percentages of phenotypically distinct B-cell subsets and plasma cells, as compared to wt EBOV, suggesting inhibition of B cell function and differentiation by VP35 IID. Finally, infection with EBOV/VP35m increased activation of NK cells, as compared to wt EBOV. These results demonstrate a global suppression of cell-mediated responses by EBOV IIDs and identify the role of DCs in suppression of T-cell responses., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.