1
|
Weng S, Zhu R, Wu Y, Xia N, Xu L, Cheng T. Research progress and application prospects of animal models of group B Coxsackievirus infections. Emerg Microbes Infect 2025; 14:2441391. [PMID: 39665300 DOI: 10.1080/22221751.2024.2441391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Group B Coxsackieviruses (CVBs) consist of six serotypes, CVB1 to CVB6, which can clinically affect the heart, brain, liver, pancreas and other organs, causing myocarditis, encephalitis, myelitis, pancreatitis, hand-foot-and-mouth disease (HFMD) and other diseases, and can even lead to death. CVBs are widespread globally and highly contagious. However, there are currently no approved CVB vaccines or effective treatments. The construction and optimization of animal models will aid in the in-depth understanding of CVB infections and its pathogenesis, providing essential tools for the exploration of vaccine development and antiviral therapies. This paper reviews the latest research progress and application prospects of CVB animal models.
Collapse
Affiliation(s)
- Shihan Weng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| | - Rui Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| | - Yuanyuan Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| | - Longfa Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, People's Republic of China
| |
Collapse
|
2
|
Gao J, Yang F, Zhang J, Yang H, Chen W. CircPTPN11 inhibits the replication of Coxsackievirus B5 through regulating the IFN-I pathway by targeting miR-152-3p/SIRPA axis. Virus Res 2024; 350:199508. [PMID: 39647532 PMCID: PMC11699211 DOI: 10.1016/j.virusres.2024.199508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Coxsackievirus B5 (CVB5) is a major pathogen responsible for hand-foot-mouth disease, herpangina, and even severe death. The mechanisms underlying CVB5-induced diseases are not fully elucidated, and no specific antiviral treatments are currently available. Circular RNAs (circRNAs), a closed-loop molecular structure, have been reported to be involved in virus infectious diseases. However, their roles and mechanisms in CVB5 infection remain largely unknown. In this study, we identify that CircPTPN11 is significantly upregulated following CVB5 infection in RD cells. Characteristic analysis reveals that the expression of CircPTPN11 is both time- and dose-dependent upon CVB5 infection and is specific to intestinal tissue. Moreover, CircPTPN11 inhibits CVB5 replication by activating IRF3 in the type-I interferon (IFN-I) pathway. Further underneath mechanism shows that CircPTPN11 indirectly regulates CVB5 replication by sponging miR-152-3p, and miR-152-3p influences CVB5 replication by interacting with the gene coding for signal regulatory protein alpha (SIRPA). In conclusion, this study suggests that CircPTPN11 targets SIRPA by sponging miR-152-3p, thereby inhibiting the replication and proliferation of CVB5. These findings provide a molecular target for the diagnosis and treatment of CVB5 infection.
Collapse
Affiliation(s)
- Jingru Gao
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Fan Yang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan Province, China; Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, Yunnan Province, China
| | - Jihong Zhang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Heng Yang
- College of Agriculture and Life Sciences, Kunming University, Kunming, China.
| | - Wei Chen
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan Province, China.
| |
Collapse
|
3
|
Farghaly TA, Abbas EMH, Abd-Elghaffar HS, Elsayed MA, Elnaggar DH, El-Sayed AF, Abd-Elshafy DN, Mohamed SF. Synthesis, characterization, molecular docking, pharmacokinetics, and molecular dynamics of new bis-thiazoles based on bis-thiosemicarbazone as anti-coxsackievirus. Sci Rep 2024; 14:29378. [PMID: 39592765 PMCID: PMC11599599 DOI: 10.1038/s41598-024-80753-z] [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: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
It was known that the majority of viral infections start off as cutaneous eruptions, which heal on their own in most cases. The prognosis is dependent on the state of immunologic surveillance, just like in other infectious disorders. Therefore, those who are immunosuppressed are more in danger. But recently it's becoming increasingly clear that eruptions that were once thought to be benign diseases can really cause problems and even death, even in immunocompetent patients. Hence, in this article, our goal was to identify possible potential antiviral candidates. We have synthesized a series of bis-thiazole derivatives via the reaction of bis-thiosemicarbazone derivative 3 with hydrazonoyl chlorides and haloketones in an effort to examine their potential antiviral properties and interactions with the main protease of Coxsackievirus B. Spectroscopic methods and elemental analysis were used to corroborate the structures of the novel bis-thiazole derivatives. The most potent derivative, bis-thiazole derivative 7a, was found to have the strongest antiviral activity against Coxsackievirus B (Cox B). Further investigation into its mode of action indicated that compound 7a has a dual activity that inhibits viral adsorption and replication. The efficacy of many compounds against Coxsackievirus adenovirus targets was assessed using molecular docking. The findings revealed that compounds 7a, 7c, 11b and 11c have high binding energies, efficiently engaging the active sites of essential Cox B virus proteins such as the Coxsackievirus adenovirus receptor (CAR), 3C-protease, and RNA-dependent RNA polymerase (RdRp). These interactions involved a variety of chemical bonding types, indicating that these substances can inhibit enzyme activity while also exhibiting substantial antiviral effects involving viral replication and adsorption. Furthermore, the computational ADMET study of these compounds indicated conformance to Lipinski's criteria, implying positive physicochemical properties. Furthermore, MD simulations demonstrated stable complexes of 7a and 11b with Coxsackievirus adenovirus receptor (CAR), 3C-protease, and RNA dependent RNA polymerase (RdRp) with RMSD (0.1-0.30, 0.20-0.30, and 0.20-0.35 nm), RMSF (0.1-0.5 nm), and SASA (80-105, 140-150, and 220-235). These outcomes further reinforce the potential of these compounds in current antiviral drug development endeavors. The collective findings underscore the potential of these compounds as candidates for antiviral therapies against Coxsackievirus adenovirus.
Collapse
Affiliation(s)
- Thoraya A Farghaly
- Department of Chemistry, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Eman M H Abbas
- Department of Chemistry, Natural and Microbial Products, National Research Centre, Dokki, Cairo, Egypt
| | - Heba S Abd-Elghaffar
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Mohamed A Elsayed
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Cairo, Egypt
| | - Dina H Elnaggar
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Cairo, Egypt.
| | - Ahmed F El-Sayed
- Microbial Genetics Department, Biotechnology Research Institute, National Research Centre, Giza, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Dina N Abd-Elshafy
- Water Pollution Research Department, National Research Centre, Dokki, 12622, Cairo, Egypt
- Research Group Immune- and Bio-markers for Infection, Centre of Excellence for Advanced Science, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Salwa F Mohamed
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Cairo, Egypt.
| |
Collapse
|
4
|
Kordbacheh R, Ashley M, Cutts WD, Keyzer TE, Chatterjee S, Altman TJ, Alexander NG, Sparer TE, Kim BJ, Sin J. Common Chemical Plasticizer Di(2-Ethhylhexyl) Phthalate Exposure Exacerbates Coxsackievirus B3 Infection. Viruses 2024; 16:1821. [PMID: 39772131 PMCID: PMC11680387 DOI: 10.3390/v16121821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Di(2-ethhylhexyl) phthalate (DEHP) is a common plastic rubberizer. DEHP leaches from plastic matrices and is under increasing scrutiny as numerous studies have linked it to negative human health manifestations. Coxsackievirus B3 (CVB) is a human pathogen that typically causes subclinical infections but can sometimes cause severe diseases such as pancreatitis, myocarditis, and meningoencephalitis. Though CVB infections are common, severe illness is relatively rare, and it is unclear what factors mediate disease severity. In this study, we sought to determine the effects that DEHP has on CVB infection in a variety of human cell types to evaluate whether this plastic-derived pollutant could represent a proviral environmental factor. METHODS HeLa cervical cancer cells, human induced pluripotent stem cell-derived brain-like endothelial cells (iBECs), and Caco-2 colon carcinoma cells were exposed to 40 µg/mL DEHP for 24 h prior to infecting with enhanced green fluorescent protein (EGFP)-expressing CVB. The severity of the infection was evaluated via fluorescence microscopy and flow cytometry-based viral EGFP detection, viral plaque assay on tissue culture media, and Western blotting to detect VP1 viral capsid protein. Interferon-associated proteins such as interferon regulatory factor (IRF) 3, IRF7, interferon-induced transmembrane (IFITM) 2, and IFITM3 were measured by Western blotting. The roles of IFITM2 and IFITM3 in the context of CVB infection were evaluated via siRNA silencing. RESULTS We found that DEHP drastically increased CVB infection in each of the cell types we tested, and, while the cellular processes underlying DEHP's proviral properties were not entirely clear, we observed that DEHP may subvert CVB-induced interferon signaling and elevate levels of IFITMs, which appeared to bolster CVB infection. CONCLUSIONS DEHP may represent a major environmental factor associated with the severity of CVB infection. Further understanding of how DEHP exacerbates infection may better elucidate its potential role as a proviral environmental factor.
Collapse
Affiliation(s)
- Ramina Kordbacheh
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Madelyn Ashley
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - William D. Cutts
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Taryn E. Keyzer
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Shruti Chatterjee
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Tyler J. Altman
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Natalie G. Alexander
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Timothy E. Sparer
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA;
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA; (R.K.); (M.A.); (W.D.C.); (T.E.K.); (S.C.); (T.J.A.); (N.G.A.); (B.J.K.)
| |
Collapse
|
5
|
Tammaro C, Plavec Z, Myllymäki L, Mitchell C, Consalvi S, Biava M, Ciogli A, Domanska A, Leppilampi V, Buckner C, Manetto S, Sciò P, Coluccia A, Laajala M, Dondio GM, Bigogno C, Marjomäki V, Butcher SJ, Poce G. SAR Analysis of Novel Coxsackie virus A9 Capsid Binders. J Med Chem 2024; 67:17144-17161. [PMID: 39292620 DOI: 10.1021/acs.jmedchem.4c00701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Enterovirus infections are common in humans, yet there are no approved antiviral treatments. In this study we concentrated on inhibition of one of the Enterovirus B (EV-B), namely Coxsackievirus A9 (CVA9), using a combination of medicinal chemistry, virus inhibition assays, structure determination from cryogenic electron microscopy and molecular modeling, to determine the structure activity relationships for a promising class of novel N-phenylbenzylamines. Of the new 29 compounds synthesized, 10 had half maximal effective concentration (EC50) values between 0.64-10.46 μM, and of these, 7 had 50% cytotoxicity concentration (CC50) values higher than 200 μM. In addition, this new series of compounds showed promising physicochemical properties and act through capsid stabilization, preventing capsid expansion and subsequent release of the genome.
Collapse
Affiliation(s)
- Chiara Tammaro
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Zlatka Plavec
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Laura Myllymäki
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, 40500 Jyväskylä, Finland
| | - Cristopher Mitchell
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Sara Consalvi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Mariangela Biava
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessia Ciogli
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Aušra Domanska
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Valtteri Leppilampi
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Cienna Buckner
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Simone Manetto
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Pietro Sciò
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonio Coluccia
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| | - Mira Laajala
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, 40500 Jyväskylä, Finland
| | | | | | - Varpu Marjomäki
- Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, 40500 Jyväskylä, Finland
| | - Sarah J Butcher
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Bioscience Research Programme, & Helsinki Institute of Life Sciences-Institute of Biotechnology, University of Helsinki, FIN-00014 Helsinki, Finland
| | - Giovanna Poce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
6
|
Volobueva AS, Fedorchenko TG, Lipunova GN, Valova MS, Sbarzaglia VA, Gladkikh AS, Kanaeva OI, Tolstykh NA, Gorshkov AN, Zarubaev VV. Leucoverdazyls as Novel Potent Inhibitors of Enterovirus Replication. Pathogens 2024; 13:410. [PMID: 38787262 PMCID: PMC11123948 DOI: 10.3390/pathogens13050410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Enteroviruses (EV) are important pathogens causing human disease with various clinical manifestations. To date, treatment of enteroviral infections is mainly supportive since no vaccination or antiviral drugs are approved for their prevention or treatment. Here, we describe the antiviral properties and mechanisms of action of leucoverdazyls-novel heterocyclic compounds with antioxidant potential. The lead compound, 1a, demonstrated low cytotoxicity along with high antioxidant and virus-inhibiting activity. A viral strain resistant to 1a was selected, and the development of resistance was shown to be accompanied by mutation of virus-specific non-structural protein 2C. This resistant virus had lower fitness when grown in cell culture. Taken together, our results demonstrate high antiviral potential of leucoverdazyls as novel inhibitors of enterovirus replication and support previous evidence of an important role of 2C proteins in EV replication.
Collapse
Affiliation(s)
| | - Tatyana G. Fedorchenko
- Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22/20 S. Kovalevskoi St., Yekaterinburg 620108, Russia
| | - Galina N. Lipunova
- Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22/20 S. Kovalevskoi St., Yekaterinburg 620108, Russia
| | - Marina S. Valova
- Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22/20 S. Kovalevskoi St., Yekaterinburg 620108, Russia
| | | | - Anna S. Gladkikh
- St. Petersburg Pasteur Institute, 14 Mira St., St. Petersburg 197101, Russia
| | - Olga I. Kanaeva
- St. Petersburg Pasteur Institute, 14 Mira St., St. Petersburg 197101, Russia
| | - Natalia A. Tolstykh
- St. Petersburg Pasteur Institute, 14 Mira St., St. Petersburg 197101, Russia
| | - Andrey N. Gorshkov
- Smorodintsev Influenza Research Institute, 15/17 Prof. Popova St., St. Petersburg 197376, Russia
| | | |
Collapse
|
7
|
Caetano CCS, Azamor T, Meyer NM, Onwubueke C, Calabrese CM, Calabrese LH, Visperas A, Piuzzi NS, Husni ME, Foo SS, Chen W. Mechanistic insights into bone remodelling dysregulation by human viral pathogens. Nat Microbiol 2024; 9:322-335. [PMID: 38316931 PMCID: PMC11045166 DOI: 10.1038/s41564-023-01586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
Bone-related diseases (osteopathologies) associated with human virus infections have increased around the globe. Recent findings have highlighted the intricate interplay between viral infection, the host immune system and the bone remodelling process. Viral infections can disrupt bone homeostasis, contributing to conditions such as arthritis and soft tissue calcifications. Osteopathologies can occur after arbovirus infections such as chikungunya virus, dengue virus and Zika virus, as well as respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 and enteroviruses such as Coxsackievirus B. Here we explore how human viruses dysregulate bone homeostasis, detailing viral factors, molecular mechanisms, host immune response changes and bone remodelling that ultimately result in osteopathologies. We highlight model systems and technologies to advance mechanistic understanding of viral-mediated bone alterations. Finally, we propose potential prophylactic and therapeutic strategies, introduce 'osteovirology' as a research field highlighting the underestimated roles of viruses in bone-related diseases, and discuss research avenues for further investigation.
Collapse
Affiliation(s)
- Camila C S Caetano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tamiris Azamor
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nikki M Meyer
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chineme Onwubueke
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cassandra M Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Leonard H Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Anabelle Visperas
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Nicolas S Piuzzi
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - M Elaine Husni
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Suan-Sin Foo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Weiqiang Chen
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
8
|
Lemos JRN, Hirani K, von Herrath M. Immunological and virological triggers of type 1 diabetes: insights and implications. Front Immunol 2024; 14:1326711. [PMID: 38239343 PMCID: PMC10794398 DOI: 10.3389/fimmu.2023.1326711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
Type 1 diabetes (T1D) is caused by an autoimmune process which culminates in the destruction of insulin-producing beta cells in the pancreas. It is widely believed that a complex and multifactorial interplay between genetic and environmental factors, such as viruses, play a crucial role in the development of the disease. Research over the past few decades has shown that there is not one single viral culprit, nor one single genetic pathway, causing the disease. Rather, viral infections, most notably enteroviruses (EV), appear to accelerate the autoimmune process leading to T1D and are often seen as a precipitator of clinical diagnosis. In support of this hypothesis, the use of anti-viral drugs has recently shown efficacy in preserving beta cell function after onset of diabetes. In this review, we will discuss the various pathways that viral infections utilize to accelerate the development of T1D. There are three key mechanisms linking viral infections to beta-cell death: One is modulated by the direct infection of islets by viruses, resulting in their impaired function, another occurs in a more indirect fashion, by modulating the immune system, and the third is caused by heightened stress on the beta-cell by interferon-mediated increase of insulin resistance. The first two aspects are surprisingly difficult to study, in the case of the former, because there are still many questions about how viruses might persist for longer time periods. In the latter, indirect/immune case, viruses might impact immunity as a hit-and-run scenario, meaning that many or all direct viral footprints quickly vanish, while changes imprinted upon the immune system and the anti-islet autoimmune response persist. Given the fact that viruses are often associated with the precipitation of clinical autoimmunity, there are concerns regarding the impact of the recent global coronavirus-2019 (COVID-19) pandemic on the development of autoimmune disease. The long-term effects of COVID-19 infection on T1D will therefore be discussed, including the increased development of new cases of T1D. Understanding the interplay between viral infections and autoimmunity is crucial for advancing our knowledge in this field and developing targeted therapeutic interventions. In this review we will examine the intricate relationship between viral infections and autoimmunity and discuss potential considerations for prevention and treatment strategies.
Collapse
Affiliation(s)
- Joana R. N. Lemos
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL, United States
| | - Khemraj Hirani
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Matthias von Herrath
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
9
|
Tan YW, Teo FMS, Ler SG, Alli-Shaik A, Nyo M, Chong CY, Tan NWH, Wang RYL, Gunaratne J, Chu JJH. Potential relevance of salivary legumain for the clinical diagnostic of hand, foot, and mouth disease. J Med Virol 2023; 95:e29243. [PMID: 38009231 DOI: 10.1002/jmv.29243] [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: 06/21/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/28/2023]
Abstract
The fight against hand, foot, and mouth disease (HFMD) remains an arduous challenge without existing point-of-care (POC) diagnostic platforms for accurate diagnosis and prompt case quarantine. Hence, the purpose of this salivary biomarker discovery study is to set the fundamentals for the realization of POC diagnostics for HFMD. Whole salivary proteome profiling was performed on the saliva obtained from children with HFMD and healthy children, using a reductive dimethylation chemical labeling method coupled with high-resolution mass spectrometry-based quantitative proteomics technology. We identified 19 upregulated (fold change = 1.5-5.8) and 51 downregulated proteins (fold change = 0.1-0.6) in the saliva samples of HFMD patients in comparison to that of healthy volunteers. Four upregulated protein candidates were selected for dot blot-based validation assay, based on novelty as biomarkers and exclusions in oral diseases and cancers. Salivary legumain was validated in the Singapore (n = 43 healthy, 28 HFMD cases) and Taiwan (n = 60 healthy, 47 HFMD cases) cohorts with an area under the receiver operating characteristic curve of 0.7583 and 0.8028, respectively. This study demonstrates the feasibility of a broad-spectrum HFMD POC diagnostic test based on legumain, a virus-specific host systemic signature, in saliva.
Collapse
Affiliation(s)
- Yong Wah Tan
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Fiona Mei Shan Teo
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siok Ghee Ler
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Asfa Alli-Shaik
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Min Nyo
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chia Yin Chong
- Infectious Disease Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Natalie Woon Hui Tan
- Infectious Disease Service, Department of Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Robert Y L Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Kidney Research Center and Department of Nephrology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial and Children's Hospital, Linkou, Taiwan
| | - Jayantha Gunaratne
- Translational Biomedical Proteomics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Fu Y, Zi R, Xiong S. Infection by exosome-carried Coxsackievirus B3 induces immune escape resulting in an aggravated pathogenesis. Microbes Infect 2023; 25:105148. [PMID: 37156458 DOI: 10.1016/j.micinf.2023.105148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
Increasing evidence has shown that extracellular vesicles or exosomes released from virus-infected cells contain viral particles, genomes, or other pathogenic factors that move to neighbor cells, contributing to virus dissemination and productive infection. Our recent study demonstrated that exosomes carrying CVB3 virions exhibited greater infection efficiency than free virions because they accessed various entry routes, overcoming restrictions to viral tropism. However, the pathogenicity of exosomes carried CVB3 and their effect on immunological properties have not yet been completely explained. In the current study, we sought to explore whether exosomes exert their effect on the CVB3-induced pathogenesis or evade the immune attack. Our results showed that exosomes-carried CVB3 could effectively infect viral receptor-negative immune cells in vivo, resulting in inducing immune system loss. Importantly, the exosomes-carried CVB3 had the ability to escape the neutralizing antibodies activity resulting in inducing the severe onset of myocarditis. Using the genetically engineered mouse with deficiency of exosomes, we observed that the exosomes-carried CVB3 reinforced an aggravated pathogenesis. By understanding how exosomes promote the course of viral disease, clinical applications of exosomes can be developed.
Collapse
Affiliation(s)
- Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ruidong Zi
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
11
|
Root-Bernstein R, Chiles K, Huber J, Ziehl A, Turke M, Pietrowicz M. Clostridia and Enteroviruses as Synergistic Triggers of Type 1 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24098336. [PMID: 37176044 PMCID: PMC10179352 DOI: 10.3390/ijms24098336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
What triggers type 1 diabetes mellitus (T1DM)? One common assumption is that triggers are individual microbes that mimic autoantibody targets such as insulin (INS). However, most microbes highly associated with T1DM pathogenesis, such as coxsackieviruses (COX), lack INS mimicry and have failed to induce T1DM in animal models. Using proteomic similarity search techniques, we found that COX actually mimicked the INS receptor (INSR). Clostridia were the best mimics of INS. Clostridia antibodies cross-reacted with INS in ELISA experiments, confirming mimicry. COX antibodies cross-reacted with INSR. Clostridia antibodies further bound to COX antibodies as idiotype-anti-idiotype pairs conserving INS-INSR complementarity. Ultraviolet spectrometry studies demonstrated that INS-like Clostridia peptides bound to INSR-like COX peptides. These complementary peptides were also recognized as antigens by T cell receptor sequences derived from T1DM patients. Finally, most sera from T1DM patients bound strongly to inactivated Clostridium sporogenes, while most sera from healthy individuals did not; T1DM sera also exhibited evidence of anti-idiotype antibodies against idiotypic INS, glutamic acid decarboxylase, and protein tyrosine phosphatase non-receptor (islet antigen-2) antibodies. These results suggest that T1DM is triggered by combined enterovirus-Clostridium (and possibly combined Epstein-Barr-virus-Streptococcal) infections, and the probable rate of such co-infections approximates the rate of new T1DM diagnoses.
Collapse
Affiliation(s)
| | - Kaylie Chiles
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Jack Huber
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Alison Ziehl
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Miah Turke
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Maja Pietrowicz
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
12
|
Tammaro C, Guida M, Appetecchia F, Biava M, Consalvi S, Poce G. Direct-Acting Antivirals and Host-Targeting Approaches against Enterovirus B Infections: Recent Advances. Pharmaceuticals (Basel) 2023; 16:203. [PMID: 37259352 PMCID: PMC9966857 DOI: 10.3390/ph16020203] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 04/03/2025] Open
Abstract
Enterovirus B (EV-B)-related diseases, which can be life threatening in high-risk populations, have been recognized as a serious health problem, but their clinical treatment is largely supportive, and no selective antivirals are available on the market. As their clinical relevance has become more serious, efforts in the field of anti-EV-B inhibitors have greatly increased and many potential antivirals with very high selectivity indexes and promising in vitro activities have been discovered. The scope of this review encompasses recent advances in the discovery of new compounds with anti-viral activity against EV-B, as well as further progress in repurposing drugs to treat these infections. Current progress and future perspectives in drug discovery against EV-Bs are briefly discussed and existing gaps are spotlighted.
Collapse
Affiliation(s)
| | | | | | | | - Sara Consalvi
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro, 5, 00185 Rome, Italy
| | - Giovanna Poce
- Department of Chemistry and Technologies of Drug, Sapienza University of Rome, Piazzale A. Moro, 5, 00185 Rome, Italy
| |
Collapse
|
13
|
Hand-Foot-and-Mouth Disease-Associated Enterovirus and the Development of Multivalent HFMD Vaccines. Int J Mol Sci 2022; 24:ijms24010169. [PMID: 36613612 PMCID: PMC9820767 DOI: 10.3390/ijms24010169] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Hand-foot-and-mouth disease (HFMD) is an infectious disease of children caused by more than 20 types of enteroviruses, with most cases recovering spontaneously within approximately one week. Severe HFMD in individual children develops rapidly, leading to death, and is associated with other complications such as viral myocarditis and type I diabetes mellitus. The approval and marketing of three inactivated EV-A71 vaccines in China in 2016 have provided a powerful tool to curb the HFMD epidemic but are limited in cross-protecting against other HFMD-associated enteroviruses. This review focuses on the epidemiological analysis of HFMD-associated enteroviruses since the inactivated EV-A71 vaccine has been marketed, collates the progress in the development of multivalent enteroviruses vaccines in different technical routes reported in recent studies, and discusses issues that need to be investigated for safe and effective HFMD multivalent vaccines.
Collapse
|