1
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
2
|
Gonzalez MM, Vizoso-Pinto MG, Erra-Balsells R, Gensch T, Cabrerizo FM. In Vitro Effect of 9,9'-Norharmane Dimer against Herpes Simplex Viruses. Int J Mol Sci 2024; 25:4966. [PMID: 38732185 PMCID: PMC11084892 DOI: 10.3390/ijms25094966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Herpes simplex virus (HSV) infections are highly widespread among humans, producing symptoms ranging from ulcerative lesions to severe diseases such as blindness and life-threatening encephalitis. At present, there are no vaccines available, and some existing antiviral treatments can be ineffective or lead to adverse effects. As a result, there is a need for new anti-HSV drugs. In this report, the in vitro anti-HSV effect of 9,9'-norharmane dimer (nHo-dimer), which belongs to the β-carboline (βC) alkaloid family, was evaluated. The dimer exhibited no virucidal properties and did not impede either the attachment or penetration steps of viral particles. The antiviral effect was only exerted under the constant presence of the dimer in the incubation media, and the mechanism of action was found to involve later events of virus infection. Analysis of fluorescence lifetime imaging data showed that the nHo-dimer internalized well into the cells when present in the extracellular incubation medium, with a preferential accumulation into perinuclear organelles including mitochondria. After washing the host cells with fresh medium free of nHo-dimer, the signal decreased, suggesting the partial release of the compound from the cells. This agrees with the observation that the antiviral effect is solely manifested when the alkaloid is consistently present in the incubation media.
Collapse
Affiliation(s)
- María Micaela Gonzalez
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, CC 164 (B7130IWA), Chascomús 7130, Argentina;
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín 1650, Argentina
| | - Maria Guadalupe Vizoso-Pinto
- Max von Pettenkofer Institute, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU, D-80336 Munich, Germany;
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, San Miguel de Tucumán 4000, Argentina
- Laboratorio Central de Cs. Básicas, Facultad de Medicina, Universidad Nacional de Tucumán, Tucumán 4000, Argentina
| | - Rosa Erra-Balsells
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón II, 3er P., Ciudad Universitaria, Buenos Aires 1428, Argentina;
- Centro de Investigación en Hidratos de Carbono (CIHIDECAR), CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Naturales Pabellón II, 3er P. Ciudad Universitaria, Buenos Aires 1428, Argentina
| | - Thomas Gensch
- Institute of Biological Information Processing 1 (IBI-1; Molecular and Cellular Physiology), Forschungszentrum Jülich, Wilhelm-Jonen-Straße, 52428 Jülich, Germany
| | - Franco M. Cabrerizo
- Instituto Tecnológico de Chascomús (CONICET-UNSAM), Av. Intendente Marino Km 8.2, CC 164 (B7130IWA), Chascomús 7130, Argentina;
- Escuela de Bio y Nanotecnologías (UNSAM), San Martín 1650, Argentina
| |
Collapse
|
3
|
Sarfraz M, Nguyen TTT, Wheler C, Köster W, Gerdts V, Dar A. Characterization of Dosage Levels for In Ovo Administration of Innate Immune Stimulants for Prevention of Yolk Sac Infection in Chicks. Vet Sci 2022; 9:vetsci9050203. [PMID: 35622731 PMCID: PMC9142911 DOI: 10.3390/vetsci9050203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Innate immune stimulants, especially toll-like receptor (TLR) ligands and agonists, are the main players in the initiation of innate immunity and have been widely studied as alternatives to antibiotics to control infection. In the present study, we characterized the dosage levels of various innate immune stimulants, including unmethylated cytosine-phosphate-guanosine dinucleotide -containing oligodeoxynucleotides (CpG ODN), polyinosinic-polycytidylic acid (poly I:C), cyclic polyphosphazene 75B (CPZ75B), avian beta-defensin 2 (ABD2), and combinations of these reagents given in ovo. Data derived from a series of animal experiments demonstrated that the in ovo administration of 10–50 µg CpG ODN/embryo (on embryonic day 18) is an effective formulation for control of yolk sac infection (YSI) due to avian pathogenic Escherichia coli (E. coli) in young chicks. Amongst the different combinations of innate immune stimulants, the in ovo administration of CpG ODN 10 µg in combination with 15 µg of poly I:C was the most effective combination, offering 100% protection from YSI. It is expected that the introduction of these reagents to management practices at the hatchery level may serve as a potential replacement for antibiotics for the reduction of early chick mortality (ECM) due to YSI/colibacillosis.
Collapse
|
4
|
Zhang P, Cui Z, Li S. The protective effects of S14G-humanin (HNG) against lipopolysaccharide (LPS)- induced inflammatory response in human dental pulp cells (hDPCs) mediated by the TLR4/MyD88/NF-κB pathway. Bioengineered 2021; 12:7552-7562. [PMID: 34605740 PMCID: PMC8806744 DOI: 10.1080/21655979.2021.1979914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulpitis is reported in large populations of patients and significantly impacts their normal life quality. It is reported that the lipopolysaccharide (LPS) in Gram-negative bacteria induces severe inflammation in dental pulp tissues. S14G-humanin is a derivative of humanin and has been recently confirmed to possess promising anti-inflammatory properties. The current study aims to explore the possibility of treating pulpitis with S14G-humanin. LPS-stimulated dental pulp cells (DPCs) were utilized to simulate an inflammatory state in the progression of pulpitis. We found the elevated expressions and production of interleukin- 6 (IL-6), tumor necrosis factor-α (TNF-α), macrophage chemoattractant protein-1 (MCP-1), matrix metalloproteinase-2 (MMP-2), and matrix metalloproteinase-9 (MMP-9), upregulated Pentraxin 3 (PTX3) and activated oxidative stress in LPS-treated DPCs were all reversed by treatment with 50 and 100 μM S14G-humanin. In addition, the LPS-induced elevated expression levels of toll-like receptor 4 (TLR4) and myeloid differentiation primary response 88 (Myd88), and activation of the IκBα/NF-κB signaling pathway in hDPCs were significantly repressed by treatment with S14G-humanin. Conclusively, we found that S14G-humanin protected LPS-treated hDPCs by inhibiting the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Stomatology, Heji Hospital Affiliated of Changzhi Medical College, Changzhi, Shanxi, China
| | - Zhiqiang Cui
- Department of Stomatology, Heji Hospital Affiliated of Changzhi Medical College, Changzhi, Shanxi, China
| | - Shuai Li
- Department of Stomatology, Heji Hospital Affiliated of Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
5
|
Wijesinghe VN, Farouk IA, Zabidi NZ, Puniyamurti A, Choo WS, Lal SK. Current vaccine approaches and emerging strategies against herpes simplex virus (HSV). Expert Rev Vaccines 2021; 20:1077-1096. [PMID: 34296960 DOI: 10.1080/14760584.2021.1960162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Vaccine development for the disease caused by the herpes simplex virus (HSV) has been challenging over the years and is always in dire need of novel approaches for prevention and cure. To date, the HSV disease remains incurable and challenging to prevent. The disease is extremely widespread due to its high infection rate, resulting in millions of infection cases worldwide.Areas covered: This review first explains the diverse forms of HSV-related disease presentations and reports past vaccine history for the disease. Next, this review examines current and novel HSV vaccine approaches being studied and tested for efficacy and safety as well as vaccines in clinical trial phases I to III. Modern approaches to vaccine design using bioinformatics are described. Finally, we discuss measures to enhance new vaccine development pipelines for HSV.Expert opinion: Modernized approaches using in silico analysis and bioinformatics are emerging methods that exhibit potential for producing vaccines with enhanced targets and formulations. Although not yet fully established for HSV disease, we describe current studies using these approaches for HSV vaccine design to shed light on these methods. In addition, we provide up-to-date requirements of immunogenicity, adjuvant selection, and routes of administration.
Collapse
Affiliation(s)
| | - Isra Ahmad Farouk
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | | | | | - Wee Sim Choo
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | - Sunil Kumar Lal
- School of Science, Monash University, Bandar Sunway, Selangor, Malaysia.,Tropical Medicine & Biology Platform, Monash University, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
6
|
Abstract
Antiviral drugs have traditionally been developed by directly targeting essential viral components. However, this strategy often fails due to the rapid generation of drug-resistant viruses. Recent genome-wide approaches, such as those employing small interfering RNA (siRNA) or clustered regularly interspaced short palindromic repeats (CRISPR) or those using small molecule chemical inhibitors targeting the cellular "kinome," have been used successfully to identify cellular factors that can support virus replication. Since some of these cellular factors are critical for virus replication, but are dispensable for the host, they can serve as novel targets for antiviral drug development. In addition, potentiation of immune responses, regulation of cytokine storms, and modulation of epigenetic changes upon virus infections are also feasible approaches to control infections. Because it is less likely that viruses will mutate to replace missing cellular functions, the chance of generating drug-resistant mutants with host-targeted inhibitor approaches is minimized. However, drug resistance against some host-directed agents can, in fact, occur under certain circumstances, such as long-term selection pressure of a host-directed antiviral agent that can allow the virus the opportunity to adapt to use an alternate host factor or to alter its affinity toward the target that confers resistance. This review describes novel approaches for antiviral drug development with a focus on host-directed therapies and the potential mechanisms that may account for the acquisition of antiviral drug resistance against host-directed agents.
Collapse
|
7
|
Vitali D, Bagri P, Wessels JM, Arora M, Ganugula R, Parikh A, Mandur T, Felker A, Garg S, Kumar MR, Kaushic C. Curcumin Can Decrease Tissue Inflammation and the Severity of HSV-2 Infection in the Female Reproductive Mucosa. Int J Mol Sci 2020; 21:ijms21010337. [PMID: 31947962 PMCID: PMC6982333 DOI: 10.3390/ijms21010337] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
Herpes Simplex Virus Type 2 (HSV-2) is one of the most prevalent sexually transmitted viruses and is a known risk factor for HIV acquisition in the Female Genital Tract (FGT). Previously, we found that curcumin can block HSV-2 infection and abrogate the production of inflammatory cytokines and chemokines by genital epithelial cells in vitro. In this study, we investigated whether curcumin, encapsulated in nanoparticles and delivered by various in vivo routes, could minimize inflammation and prevent or reduce HSV-2 infection in the FGT. Female mice were pre-treated with curcumin nanoparticles through oral, intraperitoneal and intravaginal routes, and then exposed intravaginally to the tissue inflammation stimulant CpG-oligodeoxynucleotide (ODN). Local intravaginal delivery of curcumin nanoparticles, but not intraperitoneal or oral delivery, reduced CpG-mediated inflammatory histopathology and decreased production of pro-inflammatory cytokines Interleukin (IL)-6, Tumor Necrosis Factor Alpha (TNF-α) and Monocyte Chemoattractant Protein-1 (MCP-1) in the FGT. However, curcumin nanoparticles did not demonstrate anti-viral activity nor reduce tissue pathology when administered prior to intravaginal HSV-2 infection. In an alternative approach, intravaginal pre-treatment with crude curcumin or solid dispersion formulations of curcumin demonstrated increased survival and delayed pathology following HSV-2 infection. Our results suggest that curcumin nanoparticle delivery in the vaginal tract could reduce local tissue inflammation. The anti-inflammatory properties of curcumin delivered to the vaginal tract could potentially reduce the severity of HSV-2 infection and decrease the risk of HIV acquisition in the FGT of women.
Collapse
Affiliation(s)
- Danielle Vitali
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
| | - Puja Bagri
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
| | - Jocelyn M. Wessels
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
| | - Meenakshi Arora
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (M.A.); (R.G.)
| | - Raghu Ganugula
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (M.A.); (R.G.)
| | - Ankit Parikh
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide 5000, Australia; (A.P.); (S.G.)
| | - Talveer Mandur
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
| | - Allison Felker
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide 5000, Australia; (A.P.); (S.G.)
| | - M.N.V. Ravi Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, College Station, TX 77843, USA; (M.A.); (R.G.)
| | - Charu Kaushic
- Department of Pathology & Molecular Medicine and McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada; (D.V.); (P.B.); (J.M.W.); (T.M.); (A.F.)
- Correspondence: ; Tel.: +1-905-525-9140 (ext. 22988); Fax: +1-905-522-6750
| |
Collapse
|
8
|
Tognarelli EI, Palomino TF, Corrales N, Bueno SM, Kalergis AM, González PA. Herpes Simplex Virus Evasion of Early Host Antiviral Responses. Front Cell Infect Microbiol 2019; 9:127. [PMID: 31114761 PMCID: PMC6503643 DOI: 10.3389/fcimb.2019.00127] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/10/2019] [Indexed: 12/21/2022] Open
Abstract
Herpes simplex viruses type 1 (HSV-1) and type 2 (HSV-2) have co-evolved with humans for thousands of years and are present at a high prevalence in the population worldwide. HSV infections are responsible for several illnesses including skin and mucosal lesions, blindness and even life-threatening encephalitis in both, immunocompetent and immunocompromised individuals of all ages. Therefore, diseases caused by HSVs represent significant public health burdens. Similar to other herpesviruses, HSV-1 and HSV-2 produce lifelong infections in the host by establishing latency in neurons and sporadically reactivating from these cells, eliciting recurrences that are accompanied by viral shedding in both, symptomatic and asymptomatic individuals. The ability of HSVs to persist and recur in otherwise healthy individuals is likely given by the numerous virulence factors that these viruses have evolved to evade host antiviral responses. Here, we review and discuss molecular mechanisms used by HSVs to evade early innate antiviral responses, which are the first lines of defense against these viruses. A comprehensive understanding of how HSVs evade host early antiviral responses could contribute to the development of novel therapies and vaccines to counteract these viruses.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomás F Palomino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Corrales
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Koonpaew S, Teeravechyan S, Frantz PN, Chailangkarn T, Jongkaewwattana A. PEDV and PDCoV Pathogenesis: The Interplay Between Host Innate Immune Responses and Porcine Enteric Coronaviruses. Front Vet Sci 2019; 6:34. [PMID: 30854373 PMCID: PMC6395401 DOI: 10.3389/fvets.2019.00034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/28/2019] [Indexed: 12/24/2022] Open
Abstract
Enteropathogenic porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV), members of the coronavirus family, account for the majority of lethal watery diarrhea in neonatal pigs in the past decade. These two viruses pose significant economic and public health burdens, even as both continue to emerge and reemerge worldwide. The ability to evade, circumvent or subvert the host’s first line of defense, namely the innate immune system, is the key determinant for pathogen virulence, survival, and the establishment of successful infection. Unfortunately, we have only started to unravel the underlying viral mechanisms used to manipulate host innate immune responses. In this review, we gather current knowledge concerning the interplay between these viruses and components of host innate immunity, focusing on type I interferon induction and signaling in particular, and the mechanisms by which virus-encoded gene products antagonize and subvert host innate immune responses. Finally, we provide some perspectives on the advantages gained from a better understanding of host-pathogen interactions. This includes their implications for the future development of PEDV and PDCoV vaccines and how we can further our knowledge of the molecular mechanisms underlying virus pathogenesis, virulence, and host coevolution.
Collapse
Affiliation(s)
- Surapong Koonpaew
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Samaporn Teeravechyan
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Phanramphoei Namprachan Frantz
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Thanathom Chailangkarn
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anan Jongkaewwattana
- Virology and Cell Technology Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| |
Collapse
|
10
|
Dendritic cells in the cornea during Herpes simplex viral infection and inflammation. Surv Ophthalmol 2018; 63:565-578. [DOI: 10.1016/j.survophthal.2017.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/24/2022]
|
11
|
Ahmed-Hassan H, Abdul-Cader MS, Ahmed Sabry M, Hamza E, Sharif S, Nagy E, Abdul-Careem MF. Double-Stranded Ribonucleic Acid-Mediated Antiviral Response Against Low Pathogenic Avian Influenza Virus Infection. Viral Immunol 2018; 31:433-446. [PMID: 29813000 DOI: 10.1089/vim.2017.0142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Toll-like receptor (TLR)3 signaling pathway is known to induce type 1 interferons (IFNs) and proinflammatory mediators leading to antiviral response against many viral infections. Double-stranded ribonucleic acid (dsRNA) has been shown to act as a ligand for TLR3 and, as such, has been a focus as a potential antiviral agent in many host-viral infection models. Yet, its effectiveness and involved mechanisms as a mediator against low pathogenic avian influenza virus (LPAIV) have not been investigated adequately. In this study, we used avian fibroblasts to verify whether dsRNA induces antiviral response against H4N6 LPAIV and clarify whether type 1 IFNs and proinflammatory mediators such as interleukin (IL)-1β are contributing to the dsRNA-mediated antiviral response against H4N6 LPAIV. We found that dsRNA induces antiviral response in avian fibroblasts against H4N6 LPAIV infection. The treatment of avian fibroblasts with dsRNA increases the expressions of TLR3, IFN-α, IFN-β, and IL-1β. We also confirmed that this antiviral response elicited against H4N6 LPAIV infection correlates, but is not attributable to type 1 IFNs or IL-1β. Our findings imply that the TLR3 ligand, dsRNA, can elicit antiviral response in avian fibroblasts against LPAIV infection, highlighting potential value of dsRNA as an antiviral agent against LPAIV infections. However, further investigations are required to determine the potential role of other innate immune mediators or combination of the tested cytokines in the dsRNA-mediated antiviral response against H4N6 LPAIV infection.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada .,2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Mohamed Sarjoon Abdul-Cader
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| | - Maha Ahmed Sabry
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Eman Hamza
- 2 Zoonoses Department, Faculty of Veterinary Medicine, Cairo University , Giza, Egypt
| | - Shayan Sharif
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Eva Nagy
- 3 Department of Pathobiology, University of Guelph , Guelph, Ontario, Canada
| | - Mohamed Faizal Abdul-Careem
- 1 Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, Health Research Innovation Center 2C53, University of Calgary , Calgary, Alberta, Canada
| |
Collapse
|
12
|
Skouboe MK, Knudsen A, Reinert LS, Boularan C, Lioux T, Perouzel E, Thomsen MK, Paludan SR. STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice. PLoS Pathog 2018; 14:e1006976. [PMID: 29608601 PMCID: PMC5897032 DOI: 10.1371/journal.ppat.1006976] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 04/12/2018] [Accepted: 03/17/2018] [Indexed: 01/27/2023] Open
Abstract
In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2’3’-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2’3’-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2’3’-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo. Herpes simplex virus (HSV)-2 is the leading cause of genital ulcers, and HSV-2 infection has also been reported to amplify HIV-transmission. So far, all attempts at making an effective anti-HSV2 vaccine have failed. In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat infections. Although the TLR7 agonist imiquimod has been shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects including local inflammation. Type I interferon (IFN)s are key players in antiviral defense, and it is now known that the DNA sensor cyclic GMP-AMP synthase produces the cyclic di-nucleotide (CDN) 2’3’-cyclic GMP-AMP (cGAMP), which activates the adaptor protein STING to induce IFN expression. In this work we show that natural and non-natural CDNs activate strong type I IFN responses in vivo without stimulating significant expression of genes driven by the transcription factor NF-κB, which induces inflammation. Application of CDNs at epithelial surfaces gave rise to local IFN activity, but only limited systemic responses. Importantly, all tested treatment regimens, strongly reduced replication of HSV-2 in a model for genital herpes, and significantly reduced development of disease. Finally, when comparing to TLR agonists, CDNs showed the best profile with strong IFN response specifically in the epithelial cells and limited induction of inflammation.
Collapse
Affiliation(s)
| | - Alice Knudsen
- Department of Biomedicine, Aarhus University, Denmark
| | | | | | | | | | - Martin K. Thomsen
- Department of Clinical Medicine, Aarhus University, Denmark
- * E-mail: (SRP); (MKT)
| | - Søren R. Paludan
- Department of Biomedicine, Aarhus University, Denmark
- * E-mail: (SRP); (MKT)
| |
Collapse
|
13
|
Bavananthasivam J, Kulkarni RR, Read L, Sharif S. Reduction of Marek's Disease Virus Infection by Toll-Like Receptor Ligands in Chicken Embryo Fibroblast Cells. Viral Immunol 2018; 31:389-396. [PMID: 29570417 DOI: 10.1089/vim.2017.0195] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Evolutionarily conserved pattern recognition receptors, including Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns (PAMPs) that are present in microbes. PAMPs induce several pathways downstream of TLRs that lead to induction of antiviral responses. The objective of this study was to investigate the stimulatory effect of various PAMPs (in the form of TLR ligands) in reducing Marek's disease virus (MDV) infection in chicken embryo fibroblast cells (CEFs). To this end, CEFs were pretreated with Pam3CSK4, Poly(IC), lipopolysaccharide (LPS), and CpG ODN as TLR2, TLR3, TLR4, and TLR21 ligands, respectively for 24 h followed by infection with MDV. The results indicated that pretreatment with Poly(IC) resulted in a robust reduction (by about 81%) of MDV infection in CEFs at 96 h postinfection while a moderate reduction was observed with treatment of Pam3CSK4 (35%), LPS (26%), and CpG ODN (23%) PAMPs. Transcriptional analysis of gene expression in CEFs demonstrated that all TLR ligand treatments and MDV infection significantly increased the expression of type I interferons, interleukin (IL)-1β, interferon regulatory factor 7 (IRF7), interferon induced protein with tetratricopeptide repeats 5 (IFIT5), and myxoma-resistance protein (Mx). Further studies are needed to explore the mechanism by which PAMPs, particularly the TLR3 ligands could reduce MDV infection in CEFs, which may play an important role in controlling the replication of MDV in chicken.
Collapse
Affiliation(s)
- Jegarubee Bavananthasivam
- Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Raveendra R Kulkarni
- Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Leah Read
- Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| |
Collapse
|
14
|
Ahmed-Hassan H, Abdul-Cader MS, De Silva Senapathi U, Sabry MA, Hamza E, Nagy E, Sharif S, Abdul-Careem MF. Potential mediators of in ovo delivered double stranded (ds) RNA-induced innate response against low pathogenic avian influenza virus infection. Virol J 2018. [PMID: 29530062 PMCID: PMC5848551 DOI: 10.1186/s12985-018-0954-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Toll like receptor (TLR) 3 is a critically important innate pattern recognizing receptor that senses many viral infections. Although, it has been shown that double stranded (ds) RNA can be used for the stimulation of TLR3 signaling pathway in a number of host-viral infection models, it’s effectiveness as an antiviral agent against low pathogenic avian influenza virus (LPAIV) needs further investigation. Methods In this study, first, we delivered TLR3 ligand, dsRNA, in ovo at embryo day (ED)18 since in ovo route is routinely used for vaccination against poultry viral and parasitic infections and infected with H4N6 LPAIV 24-h post-treatment. A subset of in ovo dsRNA treated and control groups were observed for the expressions of TLR3 and type I interferon (IFN)s, mRNA expression of interleukin (IL)-1β and macrophage recruitment coinciding with the time of H4N6 LPAIV infection (24 h post-treatment). Additionally, Day 1 chickens were given dsRNA intra-tracheally along with a control group and a subset of chickens were infected with H4N6 LPAIV 24-h post-treatment whereas the rest of the animals were observed for macrophage and type 1 IFN responses coinciding with the time of viral infection. Results Our results demonstrate that the pre-hatch treatment of eggs with dsRNA reduces H4N6 replication in lungs. Further studies revealed that in ovo delivery of dsRNA increases TLR3 expression, type I IFN production and number of macrophages in addition to mRNA expression of IL-1β in lung 24-h post-treatment. The same level of induction of innate response was not evident in the spleen. Moreover, we discovered that dsRNA elicits antiviral response against LPAIV correlating with type I IFN activity in macrophages in vitro. Post-hatch, we found no difference in H4N6 LPAIV genome loads between dsRNA treated and control chickens although we observed higher macrophage recruitment and IFN-β response coinciding with the time of viral infection. Conclusions Our findings imply that the TLR3 ligand, dsRNA has antiviral activity in ovo and in vitro but not in chickens post-hatch and dsRNA-mediated innate host response is characterized by macrophage recruitment and expressions of TLR3 and type 1 IFNs.
Collapse
Affiliation(s)
- Hanaa Ahmed-Hassan
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.,Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed Sarjoon Abdul-Cader
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Upasama De Silva Senapathi
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Maha Ahmed Sabry
- Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Eman Hamza
- Zoonoses Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Eva Nagy
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Mohamed Faizal Abdul-Careem
- Department of Ecosystem and Public Health, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
15
|
Neurological Disorders Associated with Human Alphaherpesviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1045:85-102. [PMID: 29896664 DOI: 10.1007/978-981-10-7230-7_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Herpes simplex virus (HSV) encephalitis is the most common cause of sporadic fatal encephalitis worldwide, and central nervous system (CNS) involvement is observed in approximately one-third of neonatal HSV infections . In recent years, single-gene inborn errors of innate immunity have been shown to be associated with susceptibility to HSV encephalitis . Temporal lobe abnormalities revealed by magnetic resonance imaging-the most sensitive imaging method for HSV encephalitis-are considered strong evidence for the disease. Detection of HSV DNA in the cerebrospinal fluid by polymerase chain reaction (PCR) is the gold standard for the diagnosis of HSV encephalitis and neonatal meningoencephalitis. Intravenous acyclovir for 14-21 days is the standard treatment in HSV encephalitis. Neurological outcomes in neonates are improved by intravenous high-dose acyclovir for 21 days followed by oral acyclovir suppressive therapy for 6 months. Varicella-zoster virus (VZV) causes a wide range of CNS manifestations. VZV encephalitis typically occurs after primary infection, and reactivation of VZV may cause encephalitis. On the other hand, VZV infection of cerebral arteries produces vasculopathy, which can manifest as ischemic stroke. Vasculopathy can occur after primary infection or reactivation of VZV. PCR detection of VZV DNA in the cerebrospinal fluid can be used for the diagnosis of encephalitis or vasculopathy. Although there are no controlled treatment trials to assess VZV treatments of encephalitis or vasculopathy, intravenous acyclovir is a common treatment.
Collapse
|
16
|
Frequently used bioinformatics tools overestimate the damaging effect of allelic variants. Genes Immun 2017; 20:10-22. [PMID: 29217828 DOI: 10.1038/s41435-017-0002-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 11/08/2022]
Abstract
We selected two sets of naturally occurring human missense allelic variants within innate immune genes. The first set represented eleven non-synonymous variants in six different genes involved in interferon (IFN) induction, present in a cohort of patients suffering from herpes simplex encephalitis (HSE) and the second set represented sixteen allelic variants of the IFNLR1 gene. We recreated the variants in vitro and tested their effect on protein function in a HEK293T cell based assay. We then used an array of 14 available bioinformatics tools to predict the effect of these variants upon protein function. To our surprise two of the most commonly used tools, CADD and SIFT, produced a high rate of false positives, whereas SNPs&GO exhibited the lowest rate of false positives in our test. As the problem in our test in general was false positive variants, inclusion of mutation significance cutoff (MSC) did not improve accuracy.
Collapse
|
17
|
Khan S, Woodruff EM, Trapecar M, Fontaine KA, Ezaki A, Borbet TC, Ott M, Sanjabi S. Dampened antiviral immunity to intravaginal exposure to RNA viral pathogens allows enhanced viral replication. J Exp Med 2016; 213:2913-2929. [PMID: 27852793 PMCID: PMC5154948 DOI: 10.1084/jem.20161289] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/17/2016] [Accepted: 10/17/2016] [Indexed: 11/21/2022] Open
Abstract
Khan et al. demonstrate that the lower female reproductive tract is exceptionally vulnerable to infection by LCMV and Zika virus, as intravaginal exposure to these RNA viral pathogens elicits a dampened antiviral immune response. Understanding the host immune response to vaginal exposure to RNA viruses is required to combat sexual transmission of this class of pathogens. In this study, using lymphocytic choriomeningitis virus (LCMV) and Zika virus (ZIKV) in wild-type mice, we show that these viruses replicate in the vaginal mucosa with minimal induction of antiviral interferon and inflammatory response, causing dampened innate-mediated control of viral replication and a failure to mature local antigen-presenting cells (APCs). Enhancement of innate-mediated inflammation in the vaginal mucosa rescues this phenotype and completely inhibits ZIKV replication. To gain a better understanding of how this dampened innate immune activation in the lower female reproductive tract may also affect adaptive immunity, we modeled CD8 T cell responses using vaginal LCMV infection. We show that the lack of APC maturation in the vaginal mucosa leads to a delay in CD8 T cell activation in the draining lymph node and hinders the timely appearance of effector CD8 T cells in vaginal mucosa, thus further delaying viral control in this tissue. Our study demonstrates that vaginal tissue is exceptionally vulnerable to infection by RNA viruses and provides a conceptual framework for the male to female sexual transmission observed during ZIKV infection.
Collapse
Affiliation(s)
- Shahzada Khan
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158
| | - Erik M Woodruff
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158
| | - Martin Trapecar
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158
| | | | - Ashley Ezaki
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158
| | - Timothy C Borbet
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158
| | - Melanie Ott
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158.,Department of Medicine, University of California, San Francisco, San Francisco, CA 94143
| | - Shomyseh Sanjabi
- Virology and Immunology, Gladstone Institutes, San Francisco, CA 94158 .,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
18
|
Xu Q, Chen Y, Zhao W, Zhang T, Liu C, Qi T, Han Z, Shao Y, Ma D, Liu S. Infection of Goose with Genotype VIId Newcastle Disease Virus of Goose Origin Elicits Strong Immune Responses at Early Stage. Front Microbiol 2016; 7:1587. [PMID: 27757109 PMCID: PMC5047883 DOI: 10.3389/fmicb.2016.01587] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/22/2016] [Indexed: 01/11/2023] Open
Abstract
Newcastle disease (ND), caused by virulent strains of Newcastle disease virus (NDV), is a highly contagious disease of birds that is responsible for heavy economic losses for the poultry industry worldwide. However, little is known about host-virus interactions in waterfowl, goose. In this study, we aim to characterize the host immune response in goose, based on the previous reports on the host response to NDV in chickens. Here, we evaluated viral replication and mRNA expression of 27 immune-related genes in 10 tissues of geese challenged with a genotype VIId NDV strain of goose origin (go/CH/LHLJ/1/06). The virus showed early replication, especially in digestive and immune tissues. The expression profiles showed up-regulation of Toll-like receptor (TLR)1–3, 5, 7, and 15, avian β-defensin (AvBD) 5–7, 10, 12, and 16, cytokines [interleukin (IL)-8, IL-18, IL-1β, and interferon-γ], inducible NO synthase (iNOS), and MHC class I in some tissues of geese in response to NDV. In contrast, NDV infection suppressed expression of AvBD1 in cecal tonsil of geese. Moreover, we observed a highly positive correlation between viral replication and host mRNA expressions of TLR1-5 and 7, AvBD4-6, 10, and 12, all the cytokines measured, MHC class I, FAS ligand, and iNOS, mainly at 72 h post-infection. Taken together, these results demonstrated that NDV infection induces strong innate immune responses and intense inflammatory responses at early stage in goose which may associate with the viral pathogenesis.
Collapse
Affiliation(s)
- Qianqian Xu
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Yuqiu Chen
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Wenjun Zhao
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Tingting Zhang
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Chenggang Liu
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Tianming Qi
- College of Animal Science and Technology, Northeast Agricultural UniversityHarbin, China; Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural SciencesHarbin, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences Harbin, China
| | - Yuhao Shao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences Harbin, China
| | - Deying Ma
- College of Animal Science and Technology, Northeast Agricultural University Harbin, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences Harbin, China
| |
Collapse
|
19
|
Zhou L, Li JL, Zhou Y, Liu JB, Zhuang K, Gao JF, Liu S, Sang M, Wu JG, Ho WZ. Induction of interferon-λ contributes to TLR3 and RIG-I activation-mediated inhibition of herpes simplex virus type 2 replication in human cervical epithelial cells. Mol Hum Reprod 2015; 21:917-29. [PMID: 26502803 PMCID: PMC4664393 DOI: 10.1093/molehr/gav058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 09/27/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
STUDY HYPOTHESIS Is it possible to immunologically activate human cervical epithelial cells to produce antiviral factors that inhibit herpes simplex virus type 2 (HSV-2) replication? STUDY FINDING Our results indicate that human cervical epithelial cells possess a functional TLR3/RIG-I signaling system, the activation of which can mount an Interferon-λ (IFN-λ)-mediated anti-HSV-2 response. WHAT IS KNOWN ALREADY There is limited information about the role of cervical epithelial cells in genital innate immunity against HSV-2 infection. STUDY DESIGN, SAMPLES/MATERIALS, METHODS We examined the expression of toll-like receptors (TLRs) and retinoic acid-inducible I (RIG-I) in End1/E6E7 cells by real-time PCR. The IFN-λ induced by TLR3 and RIG-I activation of End1/E6E7 cells was also examined by real-time PCR and ELISA. HSV-2 infection of End1/E6E7 cells was evaluated by the real-time PCR detection of HSV-2 gD expression. The antibody to IL-10Rβ was used to determine whether IFN-λ contributes to TLR3/RIG-I mediated HSV-2 inhibition. Expression of interferon regulatory factor 3 (IRF3), IRF7, IFN-stimulated gene 56 (ISG56), 2'-5'-oligoadenylate synthetase I (OAS-1) and myxovirus resistance A (MxA) were determined by the real-time PCR and western blot. End1/E6E7 cells were transfected with shRNA to knockdown the IRF3, IRF7 or RIG-I expression. Student's t-test and post Newman-Keuls test were used to analyze stabilized differences in the immunological parameters above between TLR3/RIG-I-activated cells and control cells. MAIN RESULTS AND THE ROLE OF CHANCE Human cervical epithelial cells expressed functional TLR3 and RIG-I, which could be activated by poly I:C and 5'ppp double-strand RNAs (5'ppp dsRNA), resulting in the induction of endogenous interferon lambda (IFN-λ). The induced IFN-λ contributed to TLR3/RIG-I-mediated inhibition of HSV-2 replication in human cervical epithelial cells, as an antibody to IL-10Rβ, an IFN-λ receptor subunit, could compromise TLR3/RIG-I-mediated inhibition of HSV-2. Further studies showed that TLR3/RIG-I signaling in the cervical epithelial cells by dsRNA induced the expression of the IFN-stimulated genes (ISGs), ISG56, 2'-5'-oligoadenylate synthetase I (OAS-1) and myxovirus resistance A (MxA), the key antiviral elements in the IFN signaling pathway. In addition, we observed that the topical treatment of genital mucosa with poly I:C could protect mice from genital HSV-2 infection. LIMITATIONS, REASONS FOR CAUTION Future prospective studies with primary cells and suitable animal models are needed in order to confirm these outcomes. WIDER IMPLICATIONS OF THE FINDINGS The findings provide direct and compelling evidence that there is intracellular expression and regulation of IFN-λ in human cervical epithelial cells, which may have a key role in the innate genital protection against viral infections. LARGE SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This work was supported by the National Natural Science Foundation of China (81301428 to L.Z. and 81271334 to W.-Z.H.), the Fundamental Research Funds for the Central Universities (2042015kf0188 to L.Z.), the China Postdoctoral Science Foundation (2013M531745 to L.Z.), the Development Program of China ('973', 2012CB518900 to W.-Z.H.) from the Ministry of Science and Technology of the People's Republic of China, grants (DA12815 and DA022177 to W.-Z.H.) from the National Institute on Drug Abuse (NIDA) and the open project of Hubei Key Laboratory of Wudang Local Chinese Medicine Research (WDCM005 to M.S.). The authors declare no competing financial interests.
Collapse
Affiliation(s)
- Li Zhou
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Jie-Liang Li
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Yu Zhou
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jin-Biao Liu
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Ke Zhuang
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Jian-Feng Gao
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China
| | - Shi Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Ming Sang
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China Present address: College of Basic Medical Sciences, Central Laboratory of the Fourth Affiliated Hospital in Xiangyang, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 44200, China
| | - Jian-Guo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430071, China
| | - Wen-Zhe Ho
- Animal Biosafety Level III Laboratory at the Center for Animal Experiment, Wuhan University, Wuhan 430071, China Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
20
|
Evasion of early antiviral responses by herpes simplex viruses. Mediators Inflamm 2015; 2015:593757. [PMID: 25918478 PMCID: PMC4396904 DOI: 10.1155/2015/593757] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/10/2015] [Indexed: 02/06/2023] Open
Abstract
Besides overcoming physical constraints, such as extreme temperatures, reduced humidity, elevated pressure, and natural predators, human pathogens further need to overcome an arsenal of antimicrobial components evolved by the host to limit infection, replication and optimally, reinfection. Herpes simplex virus-1 (HSV-1) and herpes simplex virus-2 (HSV-2) infect humans at a high frequency and persist within the host for life by establishing latency in neurons. To gain access to these cells, herpes simplex viruses (HSVs) must replicate and block immediate host antiviral responses elicited by epithelial cells and innate immune components early after infection. During these processes, infected and noninfected neighboring cells, as well as tissue-resident and patrolling immune cells, will sense viral components and cell-associated danger signals and secrete soluble mediators. While type-I interferons aim at limiting virus spread, cytokines and chemokines will modulate resident and incoming immune cells. In this paper, we discuss recent findings relative to the early steps taking place during HSV infection and replication. Further, we discuss how HSVs evade detection by host cells and the molecular mechanisms evolved by these viruses to circumvent early antiviral mechanisms, ultimately leading to neuron infection and the establishment of latency.
Collapse
|
21
|
|
22
|
Uyangaa E, Patil AM, Eo SK. Prophylactic and therapeutic modulation of innate and adaptive immunity against mucosal infection of herpes simplex virus. Immune Netw 2014; 14:187-200. [PMID: 25177251 PMCID: PMC4148489 DOI: 10.4110/in.2014.14.4.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/28/2014] [Accepted: 08/04/2014] [Indexed: 12/01/2022] Open
Abstract
Herpes simplex virus types 1 and 2 (HSV-1 and HSV-2) are the most common cause of genital ulceration in humans worldwide. Typically, HSV-1 and 2 infections via mucosal route result in a lifelong latent infection after peripheral replication in mucosal tissues, thereby providing potential transmission to neighbor hosts in response to reactivation. To break the transmission cycle, immunoprophylactics and therapeutic strategies must be focused on prevention of infection or reduction of infectivity at mucosal sites. Currently, our understanding of the immune responses against mucosal infection of HSV remains intricate and involves a balance between innate signaling pathways and the adaptive immune responses. Numerous studies have demonstrated that HSV mucosal infection induces type I interferons (IFN) via recognition of Toll-like receptors (TLRs) and activates multiple immune cell populations, including NK cells, conventional dendritic cells (DCs), and plasmacytoid DCs. This innate immune response is required not only for the early control of viral replication at mucosal sites, but also for establishing adaptive immune responses against HSV antigens. Although the contribution of humoral immune response is controversial, CD4(+) Th1 T cells producing IFN-γ are believed to play an important role in eradicating virus from the hosts. In addition, the recent experimental successes of immunoprophylactic and therapeutic compounds that enhance resistance and/or reduce viral burden at mucosal sites have accumulated. This review focuses on attempts to modulate innate and adaptive immunity against HSV mucosal infection for the development of prophylactic and therapeutic strategies. Notably, cells involved in innate immune regulations appear to shape adaptive immune responses. Thus, we summarized the current evidence of various immune mediators in response to mucosal HSV infection, focusing on the importance of innate immune responses.
Collapse
Affiliation(s)
- Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| |
Collapse
|
23
|
Lacroix-Lamandé S, Guesdon W, Drouet F, Potiron L, Lantier L, Laurent F. The gut flora is required for the control of intestinal infection by poly(I:C) administration in neonates. Gut Microbes 2014; 5:533-40. [PMID: 24918602 DOI: 10.4161/gmic.29154] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We found that immunostimulation of the intestinal immune system of neonatal mice by poly(I:C) injection decreased intestinal infection by the parasite Cryptosporidium parvum. We showed that the presence of dendritic cells and the cooperation of mutually dependent cytokines, such as IL-12p40, and type I and type II IFNs, were involved in the mechanism of protection induced by poly(I:C). This protection is dependent not only on TLR3-TRIF signaling, but also on the activation of the TLR5-MyD88 pathway by gut microbiota. These results raise the possibility that flagellated intestinal commensal bacteria may, in the presence of natural or synthetic agonists of TLR3, provide synergy between the TRIF and MyD88 signaling pathways, thereby favoring the development of mucosal defenses. In this addendum, we summarize these recent findings and discuss their implications for neonatal infections and immunomodulatory strategies.
Collapse
Affiliation(s)
- Sonia Lacroix-Lamandé
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| | - William Guesdon
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| | - Françoise Drouet
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| | - Laurent Potiron
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| | - Louis Lantier
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| | - Fabrice Laurent
- INRA Val de Loire; UMR 1282 Infectiologie et Santé Publique, F-37380; Nouzilly, France; Université François Rabelais; UMR 1282 Infectiologie et Santé Publique, F-37000 ; Tours, France
| |
Collapse
|
24
|
Buitendijk M, Eszterhas SK, Howell AL. Toll-like receptor agonists are potent inhibitors of human immunodeficiency virus-type 1 replication in peripheral blood mononuclear cells. AIDS Res Hum Retroviruses 2014; 30:457-67. [PMID: 24328502 DOI: 10.1089/aid.2013.0199] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Innate immune responses to microbial pathogens are initiated following the binding of ligand to specific pattern recognition receptors. Each pattern recognition receptor, which includes members of the Toll-like receptor (TLR) family, is specific for a particular type of pathogen associated molecular pattern ensuring that the organism can respond rapidly to a wide range of pathogens including bacteria, viruses, and fungi. We studied the extent to which agonists to endosomal TLR could induce anti-HIV-1 activity in peripheral blood mononuclear cells (PBMCs). When agonists to TLR3, TLR7, TLR8 and TLR9 were added prior to infection with HIV-1, they significantly reduced infection of peripheral blood mononuclear cells. Interestingly, agonists to TLR8 and TLR9 were highly effective at blocking HIV replication even when added as late as 48 h or 72 h, respectively, after HIV-1 infection, indicating that the anti-viral effect was durable and long lasting. Analysis of the induction of anti-viral genes after agonist activation of TLR indicated that all of the agonists induced expression of the type I interferons and interferon stimulated genes, although to variable levels that depended on the agonist used. Interestingly, only the agonist to TLR9, ODN2395 DNA, induced expression of type II interferon and the anti-HIV proteins Apobec3G and SAMHD1. By blocking TLR activity using an inhibitor to the MyD88 adaptor protein, we demonstrated that, at least for TLR8 and TLR9, the anti-HIV activity was not entirely mediated by TLR activation, but likely by the activation of additional anti-viral sensors in HIV target cells. These findings suggest that agonists to the endosomal TLR function to induce expression of anti-HIV molecules by both TLR-mediated and non-TLR-mediated mechanisms. Moreover, the non-TLR-mediated mechanisms induced by these agonists could potentially be exploited to block HIV-1 replication in recently HIV-exposed individuals.
Collapse
Affiliation(s)
- Maarten Buitendijk
- Department of Veterans Affairs, Veterans Health Administration, Biomedical Laboratory Research and Development, White River Junction, Vermont
- Department of Physiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Susan K. Eszterhas
- Department of Veterans Affairs, Veterans Health Administration, Biomedical Laboratory Research and Development, White River Junction, Vermont
- Department of Microbiology/Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Alexandra L. Howell
- Department of Veterans Affairs, Veterans Health Administration, Biomedical Laboratory Research and Development, White River Junction, Vermont
- Department of Microbiology/Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| |
Collapse
|
25
|
Zhang Y, Lin A, Zhang C, Tian Z, Zhang J. Phosphorothioate-modified CpG oligodeoxynucleotide (CpG ODN) induces apoptosis of human hepatocellular carcinoma cells independent of TLR9. Cancer Immunol Immunother 2014; 63:357-67. [PMID: 24452201 PMCID: PMC11029435 DOI: 10.1007/s00262-014-1518-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 01/05/2014] [Indexed: 12/13/2022]
Abstract
Toll-like receptors (TLRs) expressed on cancer cells are closely associated with tumor development. In this study, we investigated the biological functions of the TLR9 ligand, CpG oligodeoxynucleotide (CpG ODN), on TLR9 expressed in the cytoplasm of hepatocellular carcinoma (HCC) cells. In vitro, human HCC cell lines were transfected with phosphorothioate-modified oligodeoxynucleotides TLR9 agonist OND M362 and its negative control ODN M362 ctrl, which inhibited the proliferation of HCC cells by inducing apoptosis without altering the cell cycle. Interestingly, ODN M362 and ODN M362 Ctrl displayed a similar proapoptotic effect on HCC, possibly related to phosphorothioate modification of the structure of CpG ODN. Although both of them resulted in the upregulation of the TLR9 receptor, their effect on HCC apoptosis was independent of TLR9. They also upregulated inflammatory cytokines, but did not activate the NF-κB signaling pathway. Finally, the activities of ODN M362 and ODN M362 Ctrl were demonstrated in nude mice inoculated with HCC cells. These findings suggest that the phosphorothioate-modified TLR9 agonist ODN M362, and its control, elicit antitumor activity in HCC cells and may serve as a novel therapeutic target for HCC therapy.
Collapse
Affiliation(s)
- Yuyi Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan, 250012 China
| | - Ang Lin
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan, 250012 China
| | - Cai Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan, 250012 China
| | - Zhigang Tian
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan, 250012 China
| | - Jian Zhang
- Institute of Immunopharmacology and Immunotherapy, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua West Road, Jinan, 250012 China
| |
Collapse
|
26
|
Triantafilou K, Eryilmazlar D, Triantafilou M. Herpes simplex virus 2-induced activation in vaginal cells involves Toll-like receptors 2 and 9 and DNA sensors DAI and IFI16. Am J Obstet Gynecol 2014; 210:122.e1-122.e10. [PMID: 24080302 DOI: 10.1016/j.ajog.2013.09.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/14/2013] [Accepted: 09/24/2013] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The pathway by which herpes simplex virus 2 (HSV2) triggers the innate immune system in the urogenital system has not as yet been fully elucidated. In this study, we aimed to determine which pattern recognition receptors (PRRs) recognize HSV2 in primary vaginal epithelial cells. Once we deciphered the receptors involved, we aimed to target them to immunomodulate innate responses as a prophylactic or therapeutic intervention for early HSV2 infection. STUDY DESIGN To determine which PRRs are involved, receptor silencing as well as confocal microscopy was utilized. For immunomodulation, PRR agonists were utilized to induce a strong, local response to limit the infection, and we used 2 quantitative methods, flow cytometry and plaque assays, to determine their effect on HSV2 replication. RESULTS Our results show that HSV2 is detected by a plethora of PRRs: Toll-like receptors (TLR) 2 as well as deoxyribonucleic acid (DNA) sensors TLR9, DNA-dependent activator of interferon regulatory factors, and to a lesser extent interferon-inducible 16, which trigger cytokine secretion to protect the host. Using PRR agonists, such as lipoproteins, CpG DNA, and cyclic dinucleotides, we could significantly limit HSV2 replication. CONCLUSION Different PRRs are strategically placed in different cell locations to detect virus invasion. Use of agonists that target and activate these PRRs appeared to be effective in preventing primary HSV2 infection in vaginal cells and could provide new insights in defense against HSV2 urogenital infections.
Collapse
Affiliation(s)
- Kathy Triantafilou
- Institute of Infection and Immunity, School of Medicine, Cardiff University, University Hospital of Wales, Cardiff, Wales, UK
| | - Dilan Eryilmazlar
- Institute of Infection and Immunity, School of Medicine, Cardiff University, University Hospital of Wales, Cardiff, Wales, UK
| | - Martha Triantafilou
- Institute of Infection and Immunity, School of Medicine, Cardiff University, University Hospital of Wales, Cardiff, Wales, UK.
| |
Collapse
|
27
|
Lantier L, Drouet F, Guesdon W, Mancassola R, Metton C, Lo-Man R, Werts C, Laurent F, Lacroix-Lamandé S. Poly(I:C)-induced protection of neonatal mice against intestinal Cryptosporidium parvum infection requires an additional TLR5 signal provided by the gut flora. J Infect Dis 2013; 209:457-67. [PMID: 24014881 DOI: 10.1093/infdis/jit432] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The neonatal intestinal immune system is still undergoing development at birth, leading to a higher susceptibility to mucosal infections. In this study, we investigated the effect of poly(I:C) on controlling enteric infection by the protozoan Cryptosporidium parvum in neonatal mice. After poly(I:C) administration, a rapid reduction in parasite burden was observed and proved to be dependent on CD11c(+) cells and TLR3/TRIF signaling. Protection against C. parvum required additional signals provided by the gut flora through TLR5 and MyD88 signaling. This cooperation gave rise to higher levels of expression of critical mutually dependent cytokines such as interleukin 12p40 and type 1 and type 2 interferons, the last 2 being known to play a key role in the elimination of infected enterocytes. Our findings demonstrate in neonatal mice how gut flora synergizes with poly(I:C) to elicit protective intestinal immunity against an intracellular pathogen.
Collapse
Affiliation(s)
- Louis Lantier
- INRA Val de Loire, UMR1282 Infectiologie et Santé Publique, Nouzilly
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mian MF, Ahmed AN, Rad M, Babaian A, Bowdish D, Ashkar AA. Length of dsRNA (poly I:C) drives distinct innate immune responses, depending on the cell type. J Leukoc Biol 2013; 94:1025-36. [PMID: 23911868 DOI: 10.1189/jlb.0312125] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Poly I:C, a synthetic dsRNA analogue, has been used extensively for decades to study innate responses in vivo and in different cell types. We have found substantial variability while using poly I:C from different sources. In this study we found that poly I:C from 2 commercial sources induced sharply opposite responses in myeloid and fibroblasts, depending on the length of the poly I:C. Although short poly I:C (≈ 1-1.5 kb) induced greater amounts of TNF-α, IL-8, and IFN-β and a stronger antiviral response in myeloid cells, it was a poor inducer in fibroblasts. By contrast, long poly I:C (>5 kb) preferentially elicited higher cytokine and antiviral responses in fibroblasts and showed diminished responses in myeloid cells. Poly I:C activated NF-κB and STAT-1 signaling in a length- and cell-type-dependent fashion. Mechanistically, short poly I:C was better internalized in the myeloid cells and long poly I:C in the fibroblasts. Finally, long poly I:C required SR-A, whereas short poly I:C required RIG-I and Raftlin. We provide evidence that the length of dsRNA drives distinct innate responses in different cell lineages. These findings may augment in selecting the appropriate poly I:C type to design cell-type-specific potent adjuvants for vaccines against infectious diseases or cancers.
Collapse
Affiliation(s)
- M Firoz Mian
- 2.Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4K1, Canada.
| | | | | | | | | | | |
Collapse
|
29
|
Synthetic immunostimulatory oligonucleotides in experimental and clinical practice. Pharmacol Rep 2013; 64:1003-10. [PMID: 23238459 DOI: 10.1016/s1734-1140(12)70899-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 05/22/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND Oligonucleotides belong to a class of macromolecules with great potential for research and various therapeutic applications. Their mechanisms of action are extremely diverse, although they are rather homogeneous in composition. Single-stranded oligodeoxynucleotides are not only inhibitors of gene expression, but their CpG sequence motifs may activate the innate immune response. Recent progress made in preclinical and clinical testing, as well as the case of the most recently discovered RNA interference technology, will help to overcome efficacy problems of the previous approaches of the 'standard therapy' of such diseases as tumors and various infections. METHODS The aim of this article is to present various therapeutic aspects of oligonucleotides, and to review the most significant therapeutic applications of synthetic oligonucleotides. This paper presents a comprehensive review of current literature on various therapeutic properties of synthetic oligonucleotides. CONCLUSIONS The available results gathered from preclinical and clinical studies suggest that TLR9-targeted therapy of oligonucleotides can stimulate both innate and adaptive immunity. It also appears that CpG ODNs are generally safe, although moderate adverse effects, based on a backbone-related mechanism have been reported. The presented studies demonstrate that adjuvant CpG ODN can unify an immune response that leads to enhanced antigen-specific Ab formation. CpG ODN may therefore provide a unique approach to enhancing the efficacy of immunization, including the strengthening of antitumor immunity.
Collapse
|
30
|
Slavica L, Nordström I, Karlsson MN, Valadi H, Kacerovsky M, Jacobsson B, Eriksson K. TLR3 impairment in human newborns. J Leukoc Biol 2013; 94:1003-11. [PMID: 23901120 DOI: 10.1189/jlb.1212617] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Newborns are highly susceptible to viral infections. We hypothesized that this susceptibility could be due to a dysregulated expression of innate virus-sensing receptors, i.e., TLR3, TLR7, TLR8, and TLR9 and the cytosolic receptors retinoic acid-inducible gene I, melanoma differentiation-associated protein 5, protein kinase R, and IFN-γ-inducible protein 16. Cord blood mononuclear cells (CBMCs) expressed mRNA for all these receptors except for TLR3. In peripheral blood mononuclear cells (PBMCs), TLR3 mRNA was preferentially expressed in cytotoxic cells, particularly CD56(dim) NK cells. Cord NK cells in contrast showed low TLR3 mRNA expression and lacked TLR3 protein expression. Cord NK cells did not produce IFN-γ in response to polyinosinic-polycytidylic acid [poly(I:C)], whereas strong IFN-γ production was observed in poly(I:C)-stimulated adult NK cells. Cord NK cells had poor cytotoxic function that was only marginally enhanced by exposure to the TLR3 ligand poly(I:C). Opposite to NK cells from adults, their cytotoxicity was not improved by herpes simplex virus (HSV) exposure and they were unable to kill HSV-infected cells. There were no differences in the TLR3 mRNA levels among men, women, and pregnant women, implying that TLR3 is not under sex hormone control. However, decidual NK cells expressed low levels of TLR3 mRNA, which was attributed to their CD56(bright) phenotype. Our data show that cord blood NK cells have deficient TLR3 expression associated with an inability to respond to poly(I:C) and HSV activation and to kill HSV-infected cells. This might explain why newborns are particularly sensitive to neonatal HSV infections.
Collapse
Affiliation(s)
- Lucija Slavica
- 1.University of Gothenburg, Guldhedsgatan 10A, Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
31
|
Cherpes TL, Harvey SAK, Phillips JM, Vicetti Miguel RD, Melan MA, Quispe Calla NE, Hendricks RL. Use of transcriptional profiling to delineate the initial response of mice to intravaginal herpes simplex virus type 2 infection. Viral Immunol 2013; 26:172-9. [PMID: 23638732 DOI: 10.1089/vim.2012.0093] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Intravaginal (ivag) infection of mice with herpes simplex virus type 2 (HSV-2) causes genital tissue damage, quickly followed by development of fatal encephalopathy. To delineate initial host responses generated by HSV-2 infection, here oligonucleotide microarrays compared gene expression in vaginal tissue from uninfected mice and mice 1, 2, 3, 4, 5, 6, or 7 days after ivag infection with 10(4) pfu HSV-2. While comparison of mRNA expression in uninfected and HSV-infected vaginal tissue detected few changes during the first 2 days post infection (dpi), there were 156 genes whose expression was first significantly altered 3 dpi that remained significantly modified at all later time points examined. These 156 genes were significantly enriched in canonical pathways associated with interferon (IFN) signaling, activation of IFN elements by intracellular pattern recognition receptors, and antiviral immunity induced by cytosolic RIG-like receptors. Evaluation of this gene set with the National Center for Biotechnology Information Gene and INTERFEROME databases corroborated pathway analysis, as function of most (53%) were linked to IFN-mediated host immunity. In the final set of experiments, ivag administration of the Toll-like receptor 3 agonist polyinosinic: polycytidylic acid (poly I:C) 24 h before ivag HSV-2 infection reduced the incidence of genital pathology and encephalopathy, while these poly I:C-treated mice were subsequently protected from ocular HSV-2 challenge lethal to uninfected controls. The latter results imply that the exuberant antiviral immunity produced in our experimental model is simply formed too late to prevent viral replication and dissemination, and that poly I:C-induced formation of an antiviral state protecting against primary ivag infection also permits development of HSV-specific protective immunity.
Collapse
Affiliation(s)
- Thomas L Cherpes
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Gibbert K, Schlaak JF, Yang D, Dittmer U. IFN-α subtypes: distinct biological activities in anti-viral therapy. Br J Pharmacol 2013; 168:1048-58. [PMID: 23072338 PMCID: PMC3594665 DOI: 10.1111/bph.12010] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/15/2012] [Accepted: 09/07/2012] [Indexed: 12/12/2022] Open
Abstract
During most viral infections, the immediate host response is characterized by an induction of type I IFN. These cytokines have various biological activities, including anti-viral, anti-proliferative and immunomodulatory effects. After induction, they bind to their IFN-α/β receptor, which leads to downstream signalling resulting in the expression of numerous different IFN-stimulated genes. These genes encode anti-viral proteins that directly inhibit viral replication as well as modulate immune function. Thus, the induction of type I IFN is a very powerful tool for the host to fight virus infections. Many viruses evade this response by various strategies like the direct suppression of IFN induction or inhibition of the IFN signalling pathway. Therefore, the therapeutic application of exogenous type I IFN or molecules that induce strong IFN responses should be of great potential for future immunotherapies against viral infections. Type I IFN is currently used as a treatment in chronic hepatitis B and C virus infection, but as yet is not widely utilized for other viral infections. One reason for this restricted clinical use is that type I IFN belongs to a multigene family that includes 13 different IFN-α subtypes and IFN-β, whose individual anti-viral and immunomodulatory properties have so far not been investigated in detail to improve IFN therapy against viral infections in humans. In this review, we summarize the recent achievements in defining the distinct biological functions of type I IFN subtypes in cell culture and in animal models of viral infection as well as their clinical usage in chronic hepatitis virus infections.
Collapse
Affiliation(s)
- K Gibbert
- Department of Virology, University Hospital Essen, Essen, Germany.
| | | | | | | |
Collapse
|
33
|
Learning from the messengers: innate sensing of viruses and cytokine regulation of immunity - clues for treatments and vaccines. Viruses 2013; 5:470-527. [PMID: 23435233 PMCID: PMC3640511 DOI: 10.3390/v5020470] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 12/14/2022] Open
Abstract
Virus infections are a major global public health concern, and only via substantial knowledge of virus pathogenesis and antiviral immune responses can we develop and improve medical treatments, and preventive and therapeutic vaccines. Innate immunity and the shaping of efficient early immune responses are essential for control of viral infections. In order to trigger an efficient antiviral defense, the host senses the invading microbe via pattern recognition receptors (PRRs), recognizing distinct conserved pathogen-associated molecular patterns (PAMPs). The innate sensing of the invading virus results in intracellular signal transduction and subsequent production of interferons (IFNs) and proinflammatory cytokines. Cytokines, including IFNs and chemokines, are vital molecules of antiviral defense regulating cell activation, differentiation of cells, and, not least, exerting direct antiviral effects. Cytokines shape and modulate the immune response and IFNs are principle antiviral mediators initiating antiviral response through induction of antiviral proteins. In the present review, I describe and discuss the current knowledge on early virus–host interactions, focusing on early recognition of virus infection and the resulting expression of type I and type III IFNs, proinflammatory cytokines, and intracellular antiviral mediators. In addition, the review elucidates how targeted stimulation of innate sensors, such as toll-like receptors (TLRs) and intracellular RNA and DNA sensors, may be used therapeutically. Moreover, I present and discuss data showing how current antimicrobial therapies, including antibiotics and antiviral medication, may interfere with, or improve, immune response.
Collapse
|
34
|
Roth K, Ferreira VH, Kaushic C. HSV-2 vaccine: current state and insights into development of a vaccine that targets genital mucosal protection. Microb Pathog 2012; 58:45-54. [PMID: 23159485 DOI: 10.1016/j.micpath.2012.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/05/2012] [Accepted: 11/06/2012] [Indexed: 11/19/2022]
Abstract
HSV-2 is one of the most prevalent sexually transmitted infections that result in significant morbidity and financial burden on health systems around the world. Recurrent and asymptomatic re-activation accompanied by viral shedding is common among sero-positive individuals, leading to relatively high efficiency of transmission. Prophylactic HSV-2 vaccines are the best and cheapest option to address the problems associated with HSV-2 infections globally. However, despite persistent efforts, the search for an efficacious vaccine for HSV-2 remains elusive. In this review, the current state of HSV-2 vaccines and the outcome of past human trials are examined. Furthermore, we discuss the evidence and strategies from experimental mouse models that have been successful in inducing protective immunity in the genital tract against HSV-2, following immunization. Future vaccination strategies that focus on induction of robust mucosal immunity in the genital tract may hold the key for a successful vaccine against HSV-2.
Collapse
Affiliation(s)
- Kristy Roth
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote Center for Learning and Discovery, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
35
|
Boivin N, Menasria R, Piret J, Boivin G. Modulation of TLR9 response in a mouse model of herpes simplex virus encephalitis. Antiviral Res 2012; 96:414-21. [PMID: 23043942 DOI: 10.1016/j.antiviral.2012.09.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/24/2012] [Accepted: 09/27/2012] [Indexed: 01/20/2023]
Abstract
We evaluated the effects of agonists and antagonist of toll-like receptor (TLR) 9 in comparison with a TLR3 agonist in a mouse model of herpes simplex virus type 1 (HSV-1) encephalitis (HSE). BALB/c mice received a single intranasal dose of either a TLR3 agonist (polyinosinic:polycytidylic acid; PIC), TLR9 agonists (oligodeoxynucleotides (ODNs) 1585, 1826 or 2395) or a TLR9 antagonist (ODN 2088), 1 day before and, for selected groups, 3 days after infection with HSV-1. Mice that received the pre-treatment with vehicle, PIC, ODNs 1585, 1826, 2395 and 2088 before infection had survival rates of 25%, 65%, 55%, 40%, 55% and 30%, respectively (P<0.05 for PIC and ODNs 1585 and 2395 versus vehicle). Infected mice subsequently treated with vehicle, ODNs 2395 and 2088 had survival rates of 9%, 0% and 30%, respectively (P<0.05, ODN 2088 versus other groups). The pre-treatment of mice with ODN 2395 reduced both the viral load (P<0.05 at day 5) and the production of CCL2, IL-6 and CCL5 at days 3, 4 and 5 (P<0.05 for IL-6 at day 3 and P<0.05 for CCL2 and CCL5 at day 4). Treatment of infected mice with ODN 2088 reduced the production of the same cytokines (P=0.07 for CCL2 and P=0.09 for IL-6 at day 5). Pre-treatment of mice with TLR9 agonists before infection reduces brain viral load and cytokine levels resulting in increased HSE survival rates. On the other hand, TLR9 antagonists can be helpful to control the inflammatory response that could be detrimental after infection.
Collapse
Affiliation(s)
- Nicolas Boivin
- Research Center in Infectious Diseases, CHUQ-CHUL and Laval University, Quebec City, QC, Canada.
| | | | | | | |
Collapse
|
36
|
Zhou Y, Guo M, Wang X, Li J, Wang Y, Ye L, Dai M, Zhou L, Persidsky Y, Ho W. TLR3 activation efficiency by high or low molecular mass poly I:C. Innate Immun 2012; 19:184-92. [PMID: 23035017 PMCID: PMC3942089 DOI: 10.1177/1753425912459975] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Toll-like receptor 3 (TLR3) plays a critical role in initiating type I IFN-mediated innate immunity against viral infections. TLR3 recognizes various forms of double stranded (ds) RNA, including viral dsRNA and a synthetic mimic of dsRNA, poly I:C, which has been used extensively as a TLR3 ligand to induce antiviral immunity. The activation efficiency of TLR3 by poly I:C is influenced by various factors, including size of the ligands, delivery methods and cell types. In this study, we examined the stimulatory effect of two commercially-available poly I:Cs [high molecular mass (HMM) and low molecular mass (LMM)] on TLR3 activation in various human cell types by determining the induction of type I and type III IFNs, as well as the antiviral effect. We demonstrated that the direct addition of both HMM- and LMM-poly I:C to the cultures of primary macrophages or a neuroplastoma cell line could activate TLR3. However, the transfection of poly I:C was necessary to induce TLR3 activation in other cell types studied. In all the cell lines tested, the efficiency of TLR3 activation by HMM-poly I:C was significantly higher than that by LMM-poly I:C. These observations indicate the importance and necessity of developing effective TLR3 ligands for antiviral therapy.
Collapse
Affiliation(s)
- Yu Zhou
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ming Guo
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Xu Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jielang Li
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yizhong Wang
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Li Ye
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ming Dai
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Li Zhou
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Wenzhe Ho
- Center for Animal Experiment/Animal Biosafety Level III Laboratory and State Key Laboratory of Virology, Wuhan University School of Medicine, Wuhan, Hubei, People's Republic of China
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
37
|
Berry CM, Hertzog PJ, Mangan NE. Interferons as biomarkers and effectors: lessons learned from animal models. Biomark Med 2012; 6:159-76. [PMID: 22448790 DOI: 10.2217/bmm.12.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interferons (IFNs) comprise type I, II and III families with multiple subtypes. Via transcription of IFN-stimulated genes (ISGs), IFNs can exert multiple biological effects on the cell. In infectious and chronic inflammatory diseases, the IFNs and their ISG sets can be potentially utilized as biomarkers of disease outcome. Animal models allow investigations into disease pathogenesis and gene knockout models have proved cause and effect relationships of molecules related to the IFN response. Sets of IFN subtypes and their ISG products provide immunological signature patterns for different viral and other diseases. In this article, we give an overview of IFNs in several virus infection models and autoimmune diseases of medical relevance. Lessons learned from animal models inform us of IFN system parameters as indicators of disease outcome and whether clinical research is warranted. Moreover, validated IFN biomarkers for prognosis enhance our understanding of therapeutic and vaccine development.
Collapse
Affiliation(s)
- Cassandra M Berry
- Centre for Innate Immunity & Infectious Diseases, Monash Institute of Medical Research, Monash University, Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
38
|
Li YG, Siripanyaphinyo U, Tumkosit U, Noranate N, A-Nuegoonpipat A, Pan Y, Kameoka M, Kurosu T, Ikuta K, Takeda N, Anantapreecha S. Poly (I:C), an agonist of toll-like receptor-3, inhibits replication of the Chikungunya virus in BEAS-2B cells. Virol J 2012; 9:114. [PMID: 22698190 PMCID: PMC3490739 DOI: 10.1186/1743-422x-9-114] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 06/01/2012] [Indexed: 11/29/2022] Open
Abstract
Background Double-stranded RNA (dsRNA) and its mimic, polyinosinic acid: polycytidylic acid [Poly (I:C)], are recognized by toll-like receptor 3 (TLR3) and induce interferon (IFN)-β in many cell types. Poly (I:C) is the most potent IFN inducer. In in vivo mouse studies, intraperitoneal injection of Poly (I:C) elicited IFN-α/β production and natural killer (NK) cells activation. The TLR3 pathway is suggested to contribute to innate immune responses against many viruses, including influenza virus, respiratory syncytial virus, herpes simplex virus 2, and murine cytomegalovirus. In Chikungunya virus (CHIKV) infection, the viruses are cleared within 7–10 days postinfection before adaptive immune responses emerge. The innate immune response is important for CHIKV clearance. Results The effects of Poly (I:C) on the replication of CHIKV in human bronchial epithelial cells, BEAS-2B, were studied. Poly (I:C) suppressed cytopathic effects (CPE) induced by CHIKV infection in BEAS-2B cells in the presence of Poly (I:C) and inhibited the replication of CHIKV in the cells. The virus titers of Poly (I:C)-treated cells were much lower compared with those of untreated cells. CHIKV infection and Poly (I:C) treatment of BEAS-2B cells induced the production of IFN-β and increased the expression of anti-viral genes, including IFN-α, IFN-β, MxA, and OAS. Both Poly (I:C) and CHIKV infection upregulate the expression of TLR3 in BEAS-2B cells. Conclusions CHIKV is sensitive to innate immune response induced by Poly (I:C). The inhibition of CHIKV replication by Poly (I:C) may be through the induction of TLR3, which triggers the production of IFNs and other anti-viral genes. The innate immune response is important to clear CHIKV in infected cells.
Collapse
Affiliation(s)
- Yong-Gang Li
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Drannik AG, Nag K, Yao XD, Henrick BM, Sallenave JM, Rosenthal KL. Trappin-2/elafin modulate innate immune responses of human endometrial epithelial cells to PolyI:C. PLoS One 2012; 7:e35866. [PMID: 22545145 PMCID: PMC3335805 DOI: 10.1371/journal.pone.0035866] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 03/27/2012] [Indexed: 12/24/2022] Open
Abstract
Background Upon viral recognition, innate and adaptive antiviral immune responses are initiated by genital epithelial cells (ECs) to eradicate or contain viral infection. Such responses, however, are often accompanied by inflammation that contributes to acquisition and progression of sexually transmitted infections (STIs). Hence, interventions/factors enhancing antiviral protection while reducing inflammation may prove beneficial in controlling the spread of STIs. Serine antiprotease trappin-2 (Tr) and its cleaved form, elafin (E), are alarm antimicrobials secreted by multiple cells, including genital epithelia. Methodology and Principal Findings We investigated whether and how each Tr and E (Tr/E) contribute to antiviral defenses against a synthetic mimic of viral dsRNA, polyinosine-polycytidylic acid (polyI∶C) and vesicular stomatitis virus. We show that delivery of a replication-deficient adenovector expressing Tr gene (Ad/Tr) to human endometrial epithelial cells, HEC-1A, resulted in secretion of functional Tr, whereas both Tr/E were detected in response to polyI∶C. Moreover, Tr/E were found to significantly reduce viral replication by either acting directly on virus or through enhancing polyI∶C-driven antiviral protection. The latter was associated with reduced levels of pro-inflammatory factors IL-8, IL-6, TNFα, lowered expression of RIG-I, MDA5 and attenuated NF-κB activation. Interestingly, enhanced polyI∶C-driven antiviral protection of HEC-Ad/Tr cells was partially mediated through IRF3 activation, but not associated with higher induction of IFNβ, suggesting multiple antiviral mechanisms of Tr/E and the involvement of alternative factors or pathways. Conclusions and Significance This is the first evidence of both Tr/E altering viral binding/entry, innate recognition and mounting of antiviral and inflammatory responses in genital ECs that could have significant implications for homeostasis of the female genital tract.
Collapse
Affiliation(s)
- Anna G. Drannik
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Kakon Nag
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Xiao-Dan Yao
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Bethany M. Henrick
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Michel Sallenave
- Unité de Défense Innée et Inflammation, Institut Pasteur, Paris, France
- Unité U874 INSERM, Paris, France
- Université Paris 7-Denis Diderot, Paris, France
| | - Kenneth L. Rosenthal
- Department of Pathology & Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
40
|
Arav-Boger R, Wojcik GL, Duggal P, Ingersoll RG, Beaty T, Pass RF, Yolken RH. Polymorphisms in Toll-like receptor genes influence antibody responses to cytomegalovirus glycoprotein B vaccine. BMC Res Notes 2012; 5:140. [PMID: 22414065 PMCID: PMC3317442 DOI: 10.1186/1756-0500-5-140] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 03/13/2012] [Indexed: 01/27/2023] Open
Abstract
Background Congenital Cytomegalovirus (CMV) infection is an important medical problem that has yet no current solution. A clinical trial of CMV glycoprotein B (gB) vaccine in young women showed promising efficacy. Improved understanding of the basis for prevention of CMV infection is essential for developing improved vaccines. Results We genotyped 142 women previously vaccinated with three doses of CMV gB for single nucleotide polymorphisms (SNPs) in TLR 1-4, 6, 7, 9, and 10, and their associated intracellular signaling genes. SNPs in the platelet-derived growth factor receptor (PDGFRA) and integrins were also selected based on their role in binding gB. Specific SNPs in TLR7 and IKBKE (inhibitor of nuclear factor kappa-B kinase subunit epsilon) were associated with antibody responses to gB vaccine. Homozygous carriers of the minor allele at four SNPs in TLR7 showed higher vaccination-induced antibody responses to gB compared to heterozygotes or homozygotes for the common allele. SNP rs1953090 in IKBKE was associated with changes in antibody level from second to third dose of vaccine; homozygotes for the minor allele exhibited lower antibody responses while homozygotes for the major allele showed increased responses over time. Conclusions These data contribute to our understanding of the immunogenetic mechanisms underlying variations in the immune response to CMV vaccine.
Collapse
Affiliation(s)
- Ravit Arav-Boger
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins Hospital, Baltimore, Maryland 21287-4933, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Rose WA, McGowin CL, Spagnuolo RA, Eaves-Pyles TD, Popov VL, Pyles RB. Commensal bacteria modulate innate immune responses of vaginal epithelial cell multilayer cultures. PLoS One 2012; 7:e32728. [PMID: 22412914 PMCID: PMC3296736 DOI: 10.1371/journal.pone.0032728] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 02/01/2012] [Indexed: 12/29/2022] Open
Abstract
The human vaginal microbiome plays a critical but poorly defined role in reproductive health. Vaginal microbiome alterations are associated with increased susceptibility to sexually-transmitted infections (STI) possibly due to related changes in innate defense responses from epithelial cells. Study of the impact of commensal bacteria on the vaginal mucosal surface has been hindered by current vaginal epithelial cell (VEC) culture systems that lack an appropriate interface between the apical surface of stratified squamous epithelium and the air-filled vaginal lumen. Therefore we developed a reproducible multilayer VEC culture system with an apical (luminal) air-interface that supported colonization with selected commensal bacteria. Multilayer VEC developed tight-junctions and other hallmarks of the vaginal mucosa including predictable proinflammatory cytokine secretion following TLR stimulation. Colonization of multilayers by common vaginal commensals including Lactobacillus crispatus, L. jensenii, and L. rhamnosus led to intimate associations with the VEC exclusively on the apical surface. Vaginal commensals did not trigger cytokine secretion but Staphylococcus epidermidis, a skin commensal, was inflammatory. Lactobacilli reduced cytokine secretion in an isolate-specific fashion following TLR stimulation. This tempering of inflammation offers a potential explanation for increased susceptibility to STI in the absence of common commensals and has implications for testing of potential STI preventatives.
Collapse
Affiliation(s)
- William A. Rose
- Department of Microbiology and Immunology, University of Texas Medical Branch, Glaveston, Texas, United States of America
| | - Chris L. McGowin
- Department of Pathology, University of Texas Medical Branch, Glaveston, Texas, United States of America
| | - Rae Ann Spagnuolo
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Tonyia D. Eaves-Pyles
- Department of Microbiology and Immunology, University of Texas Medical Branch, Glaveston, Texas, United States of America
| | - Vsevolod L. Popov
- Department of Pathology, University of Texas Medical Branch, Glaveston, Texas, United States of America
| | - Richard B. Pyles
- Department of Microbiology and Immunology, University of Texas Medical Branch, Glaveston, Texas, United States of America
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
42
|
A nonreplicating subunit vaccine protects mice against lethal Ebola virus challenge. Proc Natl Acad Sci U S A 2011; 108:20695-700. [PMID: 22143779 DOI: 10.1073/pnas.1117715108] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Ebola hemorrhagic fever is an acute and often deadly disease caused by Ebola virus (EBOV). The possible intentional use of this virus against human populations has led to design of vaccines that could be incorporated into a national stockpile for biological threat reduction. We have evaluated the immunogenicity and efficacy of an EBOV vaccine candidate in which the viral surface glycoprotein is biomanufactured as a fusion to a monoclonal antibody that recognizes an epitope in glycoprotein, resulting in the production of Ebola immune complexes (EICs). Although antigen-antibody immune complexes are known to be efficiently processed and presented to immune effector cells, we found that codelivery of the EIC with Toll-like receptor agonists elicited a more robust antibody response in mice than did EIC alone. Among the compounds tested, polyinosinic:polycytidylic acid (PIC, a Toll-like receptor 3 agonist) was highly effective as an adjuvant agent. After vaccinating mice with EIC plus PIC, 80% of the animals were protected against a lethal challenge with live EBOV (30,000 LD(50) of mouse adapted virus). Surviving animals showed a mixed Th1/Th2 response to the antigen, suggesting this may be important for protection. Survival after vaccination with EIC plus PIC was statistically equivalent to that achieved with an alternative viral vector vaccine candidate reported in the literature. Because nonreplicating subunit vaccines offer the possibility of formulation for cost-effective, long-term storage in biothreat reduction repositories, EIC is an attractive option for public health defense measures.
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW The early stage of HIV-1 infection is when the virus is most vulnerable, and should therefore offer the best opportunity for therapeutic interventions. This review addresses the recent progress in the understanding of innate immune responses against HIV-1 with focus on the potential targets for prevention of viral acquisition, replication and dissemination. RECENT FINDINGS Research indicates that the host-derived factor trappin-2/elafin is protective against HIV, whereas semen-derived enhancer of viral infection and defensins 5 and 6 enhance viral transmission. Further, studies suggest that stimulation of TLR4 and inhibition of TLR7-9 pathways may be HIV suppressive. The regulation and function of viral restriction factors tetherin and APOBEC3G have been investigated and a molecule mimicking the premature uncoating achieved by TRIM5α, PF74, has been identified. Chloroquine has been shown to inhibit plasmacytoid dendritic cell activation and suppress negative modulators of T-cell responses. Blockade of HMBG1 has been found to restore natural-killer-cell-mediated killing of infected dendritic cells, normally suppressed by HIV-1. Interestingly, when used as adjuvants, EAT-2 and heat shock protein gp96 reportedly enhance innate immune responses. SUMMARY Several targets for innate immunity-mediated therapeutics have been identified. Nonetheless, more research is required to unveil their underlying mechanisms and interactions before testing these molecules in clinical trials.
Collapse
|
44
|
Abdul-Careem MF, Firoz Mian M, Gillgrass AE, Chenoweth MJ, Barra NG, Chan T, Al-Garawi AA, Chew MV, Yue G, Roojen NV, Xing Z, Ashkar AA. FimH, a TLR4 ligand, induces innate antiviral responses in the lung leading to protection against lethal influenza infection in mice. Antiviral Res 2011; 92:346-55. [DOI: 10.1016/j.antiviral.2011.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 09/03/2011] [Accepted: 09/07/2011] [Indexed: 10/17/2022]
|
45
|
Melchjorsen J. Sensing herpes: more than toll. Rev Med Virol 2011; 22:106-21. [PMID: 22020814 DOI: 10.1002/rmv.716] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 09/12/2011] [Accepted: 09/14/2011] [Indexed: 12/21/2022]
Abstract
To launch an effective antiviral immune response, cells must recognize the virus, activate a cytokine response, and initiate inflammatory processes. Herpes simplex virus 1 (HSV-1) and HSV-2 are nuclear-replicating viruses composed of a double-stranded DNA genome plus glycoproteins that are incorporated into a lipid bilayer envelope that surrounds an icosahedral capsid. Several novel receptors that mediate innate recognition of HSV and that activate the innate immune response have been identified in recent years. The host-virus interactions that lead to type I interferon (IFN), type III IFN, and cytokine production include cellular recognition of viral envelope and structural proteins, recognition of viral genomic DNA and recognition of virus-derived double-stranded RNAs. Such RNAs can interact with cellular pattern-recognition receptors, including Toll-like receptors and a number of cytoplasmic and nuclear receptors for virus DNA and virus-derived RNAs. In this review, I present a systematic overview of innate cellular recognition of HSV infection that leads to immune activation, and I discuss the implications of the known cell-host interactions. In addition, I discuss the use of innate stimulation to improve anti-HSV treatment and vaccine response and I discuss future research aims.
Collapse
Affiliation(s)
- Jesper Melchjorsen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
46
|
Yao XD, Rosenthal KL. Herpes simplex virus type 2 virion host shutoff protein suppresses innate dsRNA antiviral pathways in human vaginal epithelial cells. J Gen Virol 2011; 92:1981-1993. [PMID: 21632561 DOI: 10.1099/vir.0.030296-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Viruses that establish persistent infections have evolved numerous strategies to evade host innate antiviral responses. We functionally assessed the role of herpes simplex virus type 2 (HSV-2) virion host shutoff (vhs) protein on innate immune sensing pathways in human vaginal epithelial cells (VK2 ECs). Infection of cells with wild-type (WT) HSV-2 significantly decreased expression of innate immune sensors of viral infection, Toll-like receptor (TLR)2, TLR3, retinoic acid inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (Mda-5), relative to cells infected with a mutant that lacks vhs (vhsB) or mock-infected cells. Transfection with HSV-2 vhs similarly decreased expression of TLR2, TLR3, RIG-I and Mda-5, which was also confirmed in human embryonic kidney (HEK) 293 cells. vhsB infection of VK2 cells caused robust increases in the active form of interferon regulatory factor (IRF)3 and its translocation to the nucleus compared with the WT. Additionally, IRF3 activation by Sendai virus and polyinosinic : polycytidylic acid-induced stimulation of beta interferon (IFN-β) was significantly inhibited in vhs-transfected cells. Overall, our findings provide the first evidence that HSV-2 vhs plays roles in selectively inhibiting TLR3 and RIG-I/Mda-5, as well as TLR2-mediated antiviral pathways for sensing dsRNA and effectively suppresses IFN-β antiviral responses in human vaginal ECs.
Collapse
Affiliation(s)
- Xiao-Dan Yao
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Kenneth Lee Rosenthal
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
47
|
Wick DA, Webb JR. A novel, broad spectrum therapeutic HPV vaccine targeting the E7 proteins of HPV16, 18, 31, 45 and 52 that elicits potent E7-specific CD8T cell immunity and regression of large, established, E7-expressing TC-1 tumors. Vaccine 2011; 29:7857-66. [PMID: 21816200 DOI: 10.1016/j.vaccine.2011.07.090] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 07/08/2011] [Accepted: 07/20/2011] [Indexed: 12/21/2022]
Abstract
Persistent infection by high risk genotypes of human papillomavirus (HPV) is the cause of cervical cancer, which remains one of the most common cancers among women worldwide. In addition, there is a growing appreciation that high risk HPVs are associated with a number of other cancers including anogenital cancers as well as a subset of head and neck cancers. Recently, prophylactic HPV vaccines targeting the two most prevalent high risk HPVs (HPV16 and HPV18) have been deployed in large-scale vaccination campaigns. However, the extent to which these prophylactic vaccines confer protection against other high risk HPV genotypes is largely unknown and prophylactic vaccines have been shown to be ineffective against pre-existing infection. Thus there continues to be an urgent need for effective therapeutic vaccines against HPV. The E7 protein of HPV16 has been widely studied as a target for therapeutic vaccines in HPV-associated cancer settings because HPV16 is the most prevalent of the high risk HPV genotypes. However, HPV16 accounts for only about 50% of cervical cancers and there are at least 15 other high risk HPVs that are known to be oncogenic. We have developed a novel, broad-spectrum, therapeutic vaccine (Pentarix) directed at the E7 proteins from five of the most prevalent high-risk genotypes of HPV worldwide (HPV16, 18, 31, 45 and 52) that together account for more than 80% of all HPV-associated cancers. Pentarix is a recombinant protein-based vaccine that elicits strong, multi-genotype specific CD8 T cell immunity when administered to mice in combination with adjuvants comprised of agonists of the TLR3 or TLR9 family of innate immune receptors. Furthermore, large, established E7-expressing TC-1 tumors undergo rapid and complete regression after therapeutic vaccination of mice with Pentarix. Together, these data suggest that Pentarix may be of clinical value for patients with E7-positive, HPV-associated precancerous lesions or malignant disease.
Collapse
Affiliation(s)
- Darin A Wick
- British Columbia Cancer Agency, Trev and Joyce Deeley Research Centre, 2410 Lee Avenue, Victoria, Canada
| | | |
Collapse
|
48
|
Tweaking Innate Immunity: the Promise of Innate Immunologicals As Anti-infectives. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2011; 17:307-14. [PMID: 18382644 DOI: 10.1155/2006/195957] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
New and exciting insights into the importance of the innate immune system are revolutionizing our understanding of immune defense against infections, pathogenesis, and the treatment and prevention of infectious diseases. The innate immune system uses multiple families of germline-encoded pattern recognition receptors (PRRs) to detect infection and trigger a variety of antimicrobial defense mechanisms. PRRs are evolutionarily highly conserved and serve to detect infection by recognizing pathogen-associated molecular patterns that are unique to microorganisms and essential for their survival. Toll-like receptors (TLRs) are transmembrane signalling receptors that activate gene expression programs that result in the production of proinflammatory cytokines and chemokines, type I interferons and antimicrobial factors. Furthermore, TLR activation facilitates and guides activation of adaptive immune responses through the activation of dendritic cells. TLRs are localized on the cell surface and in endosomal/lysosomal compartments, where they detect bacterial and viral infections. In contrast, nucleotide-binding oligomerization domain proteins and RNA helicases are located in the cell cytoplasm, where they serve as intracellular PRRs to detect cytoplasmic infections, particularly viruses. Due to their ability to enhance innate immune responses, novel strategies to use ligands, synthetic agonists or antagonists of PRRs (also known as 'innate immunologicals') can be used as stand-alone agents to provide immediate protection or treatment against bacterial, viral or parasitic infections. Furthermore, the newly appreciated importance of innate immunity in initiating and shaping adaptive immune responses is contributing to our understanding of vaccine adjuvants and promises to lead to improved next-generation vaccines.
Collapse
|
49
|
Karpala AJ, Lowenthal JW, Bean AGD. Identifying innate immune pathways of the chicken may lead to new antiviral therapies. Vet Immunol Immunopathol 2011; 148:100-9. [PMID: 21715024 DOI: 10.1016/j.vetimm.2011.05.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 03/25/2011] [Accepted: 05/30/2011] [Indexed: 12/23/2022]
Abstract
Zoonotic viruses, such as highly pathogenic avian influenza (HPAI), present a significant threat to both the poultry industry and public health. The present method of controlling avian influenza (AI) relies on good farming practice with limited use of vaccination in some countries. However, new ways to control disease outbreaks might be possible with additional knowledge of the natural host response to virus. Moreover, manipulation of the innate immune system in mammals improves the outcomes following viral infection. A similar approach might be applied to the chicken, nevertheless, a greater knowledge of the chicken innate immune system is required. This review outlines important mammalian antiviral mechanisms that have been modulated to strengthen viral immunity and highlights the potential application of these strategies in the chicken, especially in regards, to AI.
Collapse
Affiliation(s)
- Adam J Karpala
- CSIRO, Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia.
| | | | | |
Collapse
|
50
|
Han JY, Schleiss MR. It is time to examine the role of host cytokine response in neonatal herpes simplex virus infection. Future Virol 2011. [DOI: 10.2217/fvl.11.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
| | - Mark R Schleiss
- Department of Pediatrics & Center for Infectious Diseases & Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota, 420 Delaware Street SE, MMC 296, Minneapolis, MN 55455, USA
| |
Collapse
|