201
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
202
|
Wang N, Yao F, Liu D, Jiang H, Xia X, Xiong S. RNA N6-methyladenosine in nonocular and ocular disease. J Cell Physiol 2021; 237:1686-1710. [PMID: 34913163 DOI: 10.1002/jcp.30652] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 10/27/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
N6 -methyladenosine (m6 A), the sixth N methylation of adenylate (A) in RNA, is the most abundant transcriptome modification in eukaryotic messenger RNA (mRNAs). m6 A modification exists in both coding mRNA and noncoding RNAs, and its functions are controlled by methyltransferase, demethylase, and m6 A reading proteins. Methylation modification of m6 A can regulate RNA cleavage, transport, stability, and expression. This review summarizes the enzymes involved in RNA m6 A methylation and the commonly used detection methods. The role of m6 A modification in physiological processes is described, and its impact on tumorigenesis, viral infection, and diabetes is further highlighted. Moreover, up-to-date knowledge of the implications of RNA m6 A modification in ocular diseases such as uveal melanoma and diabetic retinopathy is introduced. Clarifying the mechanism of RNA m6 A methylation will help elucidate the pathogenesis of various diseases, providing options for subsequent treatment.
Collapse
Affiliation(s)
- Nan Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| | - Fei Yao
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| | - Die Liu
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| | - Haibo Jiang
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| | - Siqi Xiong
- Eye Center of Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Opthalmology, Central South University, Changsha, China
| |
Collapse
|
203
|
Kornuta CA, Cheuquepán F, Bidart JE, Soria I, Gammella M, Quattrocchi V, Hecker YP, Moore DP, Romera SA, Marin MS, Zamorano PI, Langellotti CA. TLR activation, immune response and viral protection elicited in cattle by a commercial vaccine against Bovine Herpesvirus-1. Virology 2021; 566:98-105. [PMID: 34896902 DOI: 10.1016/j.virol.2021.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/11/2021] [Accepted: 11/29/2021] [Indexed: 11/29/2022]
Abstract
The innate and acquired immune response induced by a commercial inactivated vaccine against Bovine Herpesvirus-1 (BoHV-1) and protection conferred against the virus were analyzed in cattle. Vaccination induced high levels of BoHV-1 antibodies at 30, 60, and 90 days post-vaccination (dpv). IgG1 and IgG2 isotypes were detected at 90 dpv, as well as virus-neutralizing antibodies. An increase of anti-BoHV-1 IgG1 in nasal swabs was detected 6 days post-challenge in vaccinated animals. After viral challenge, lower virus excretion and lower clinical score were observed in vaccinated as compared to unvaccinated animals, as well as BoHV-1-specific proliferation of lymphocytes and production of IFNγ, TNFα, and IL-4. Downregulation of the expression of endosome Toll-like receptors 8-9 was detected after booster vaccination. This is the first thorough study of the immunity generated by a commercial vaccine against BoHV-1 in cattle.
Collapse
Affiliation(s)
- Claudia Alejandra Kornuta
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Felipe Cheuquepán
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Instituto de Innovación para La Producción Agropecuaria y El Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Juan Esteban Bidart
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ivana Soria
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina
| | - Mariela Gammella
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina
| | - Valeria Quattrocchi
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina
| | - Yanina Paola Hecker
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Instituto de Innovación para La Producción Agropecuaria y El Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Dadin Prando Moore
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Instituto de Innovación para La Producción Agropecuaria y El Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Sonia Alejandra Romera
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Universidad Del Salvador, Buenos Aires, Argentina
| | - Maia Solange Marin
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Instituto de Innovación para La Producción Agropecuaria y El Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Patricia Inés Zamorano
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Universidad Del Salvador, Buenos Aires, Argentina
| | - Cecilia Ana Langellotti
- Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), Hurlingham, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
204
|
Dietary Flavone Baicalein Combinate with Genipin Attenuates Inflammation Stimulated by Lipopolysaccharide in RAW264.7 Cells or Pseudomonas aeruginosa in Mice via Regulating the Expression and Phosphorylation of AKT. Nutrients 2021; 13:nu13124462. [PMID: 34960014 PMCID: PMC8708859 DOI: 10.3390/nu13124462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 01/01/2023] Open
Abstract
Mounting evidence has shown that single-targeted therapy might be inadequate to achieve satisfactory effects. Thus, drug combinations are gaining attention as they can regulate multiple targets to obtain more beneficial effects. Heat shock protein 90 (HSP90) is a molecular chaperone that assists the protein assembly and folding of client proteins and maintains their stability. Interfering with the interaction between HSP90 and its client proteins by inhibiting the latter’s activity may offer a new approach toward combination therapy. The HSP90 client protein AKT plays an important role in the inflammatory response syndrome caused by infections. In this study, the dietary flavone baicalein was identified as a novel inhibitor of HSP90 that targeted the N-terminal ATP binding pocket of HSP90 and hindered the chaperone cycle, resulting in AKT degradation. Combining baicalein with genipin, which was extracted from Gardenia jasminoides, could inhibit the pleckstrin homology domain of AKT, significantly increasing the anti-inflammatory effects both in vitro and in vivo. This synergistic effect was attributed to the reduction in AKT expression and phosphorylation. Thus, elucidating the mechanism underlying this effect will provide a new avenue for the clinical application and development of synergistic anti-inflammatory drugs.
Collapse
|
205
|
Potential value of pharmacological agents acting on toll-like receptor (TLR) 7 and/or TLR8 in COVID-19. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100068. [PMID: 34870161 PMCID: PMC8562070 DOI: 10.1016/j.crphar.2021.100068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic is an ongoing global pandemic of coronavirus disease 2019 as an atypical type of viral pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many potential pharmacotherapies are currently being investigated against this disease. This article points to and justifies, the importance of investigating the potential therapeutic value of pharmacological agents acting on Toll-like Receptor (TLR) 7 and/or TLR8 as double-edged swords combating COVID-19. Induction of TLR7 and/or TLR8 may be investigated as a strategy to stimulate immunity and may be added to anti-COVID19 vaccines to cope with their current viral escape challenge. TLR7 stimulation may not only help viral clearance through Th1 antiviral responses but may also provide beneficial broncho- and vaso-dilatory, as well as, anti-inflammatory effects. Pharmacological compounds acting as TLR7 and/or TLR8 agonists may be of value if used by frontline healthcare workers with comorbidities who demonstrate mild symptoms of the disease. On the other hand, TLR7 and/or TLR8 antagonists may be used in combination with immune-modulatory/anti-inflammatory drugs in severe cases of the disease, with potential synergistic effects that could also help in reducing the doses of such therapies, and consequently their adverse effects. There is potential value in development of TLR7/8 agonists/antagonists as double-edged swords combating COVID-19. TLR7/8 agonists, as immune-stimulants in mild disease, may also be added to preventive vaccines acting against viral escape. TLR7/8 antagonists may be combined with immune-suppressants in severe cases for potential synergism and minimized toxicity.
Collapse
|
206
|
Xu C, Chen J, Chen X. Host Innate Immunity Against Hepatitis Viruses and Viral Immune Evasion. Front Microbiol 2021; 12:740464. [PMID: 34803956 PMCID: PMC8598044 DOI: 10.3389/fmicb.2021.740464] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Hepatitis viruses are primary causative agents of hepatitis and represent a major source of public health problems in the world. The host innate immune system forms the first line of defense against hepatitis viruses. Hepatitis viruses are sensed by specific pathogen recognition receptors (PRRs) that subsequently trigger the innate immune response and interferon (IFN) production. However, hepatitis viruses evade host immune surveillance via multiple strategies, which help compromise the innate immune response and create a favorable environment for viral replication. Therefore, this article reviews published findings regarding host innate immune sensing and response against hepatitis viruses. Furthermore, we also focus on how hepatitis viruses abrogate the antiviral effects of the host innate immune system.
Collapse
Affiliation(s)
- Chonghui Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jizheng Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xinwen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
207
|
Mabrey FL, Morrell ED, Wurfel MM. TLRs in COVID-19: How they drive immunopathology and the rationale for modulation. Innate Immun 2021; 27:503-513. [PMID: 34806446 PMCID: PMC8762091 DOI: 10.1177/17534259211051364] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is both a viral illness and a disease of immunopathology. Proximal events within the innate immune system drive the balance between deleterious inflammation and viral clearance. We hypothesize that a divergence between the generation of excessive inflammation through over activation of the TLR associated myeloid differentiation primary response (MyD88) pathway relative to the TIR-domain-containing adaptor-inducing IFN-β (TRIF) pathway plays a key role in COVID-19 severity. Both viral elements and damage associated host molecules act as TLR ligands in this process. In this review, we detail the mechanism for this imbalance in COVID-19 based on available evidence, and we discuss how modulation of critical elements may be important in reducing severity of disease.
Collapse
Affiliation(s)
- F Linzee Mabrey
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| | - Eric D Morrell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, 7284University of Washington, USA
| |
Collapse
|
208
|
Orús-Alcalde A, Lu TM, Børve A, Hejnol A. The evolution of the metazoan Toll receptor family and its expression during protostome development. BMC Ecol Evol 2021; 21:208. [PMID: 34809567 PMCID: PMC8609888 DOI: 10.1186/s12862-021-01927-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 10/21/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Toll-like receptors (TLRs) play a crucial role in immunity and development. They contain leucine-rich repeat domains, one transmembrane domain, and one Toll/IL-1 receptor domain. TLRs have been classified into V-type/scc and P-type/mcc TLRs, based on differences in the leucine-rich repeat domain region. Although TLRs are widespread in animals, detailed phylogenetic studies of this gene family are lacking. Here we aim to uncover TLR evolution by conducting a survey and a phylogenetic analysis in species across Bilateria. To discriminate between their role in development and immunity we furthermore analyzed stage-specific transcriptomes of the ecdysozoans Priapulus caudatus and Hypsibius exemplaris, and the spiralians Crassostrea gigas and Terebratalia transversa. RESULTS We detected a low number of TLRs in ecdysozoan species, and multiple independent radiations within the Spiralia. V-type/scc and P-type/mcc type-receptors are present in cnidarians, protostomes and deuterostomes, and therefore they emerged early in TLR evolution, followed by a loss in xenacoelomorphs. Our phylogenetic analysis shows that TLRs cluster into three major clades: clade α is present in cnidarians, ecdysozoans, and spiralians; clade β in deuterostomes, ecdysozoans, and spiralians; and clade γ is only found in spiralians. Our stage-specific transcriptome and in situ hybridization analyses show that TLRs are expressed during development in all species analyzed, which indicates a broad role of TLRs during animal development. CONCLUSIONS Our findings suggest that a clade α TLR gene (TLR-Ca) and a clade β/γ TLR gene (TLR-Cβ/γ) were already present in the cnidarian-bilaterian common ancestor. However, although TLR-Ca was conserved in cnidarians, TLR-Cβ/γ was lost during the early evolution of these taxa. Moreover, TLR-Cβ/γ duplicated to generate TLR-Cβ and TLR-Cγ in the lineage to the last common protostome-deuterostome ancestor. TLR-Ca, TLR-Cβ and TLR-Cγ further expanded generating the three major TLR clades. While all three clades radiated in several spiralian lineages, specific TLRs clades have been presumably lost in other lineages. Furthermore, the expression of the majority of these genes during protostome ontogeny suggests a likely role in development.
Collapse
Affiliation(s)
- Andrea Orús-Alcalde
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Tsai-Ming Lu
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Aina Børve
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Andreas Hejnol
- Sars International Centre for Marine Molecular Biology, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway.
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
| |
Collapse
|
209
|
Glitscher M, Hildt E. Endosomal Cholesterol in Viral Infections - A Common Denominator? Front Physiol 2021; 12:750544. [PMID: 34858206 PMCID: PMC8632007 DOI: 10.3389/fphys.2021.750544] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
Cholesterol has gained tremendous attention as an essential lipid in the life cycle of virtually all viruses. These seem to have developed manifold strategies to modulate the cholesterol metabolism to the side of lipid uptake and de novo synthesis. In turn, affecting the cholesterol homeostasis has emerged as novel broad-spectrum antiviral strategy. On the other hand, the innate immune system is similarly regulated by the lipid and stimulated by its derivatives. This certainly requires attention in the design of antiviral strategies aiming to decrease cellular cholesterol, as evidence accumulates that withdrawal of cholesterol hampers innate immunity. Secondly, there are exceptions to the rule of the abovementioned virus-induced metabolic shift toward cholesterol anabolism. It therefore is of interest to dissect underlying regulatory mechanisms, which we aimed for in this minireview. We further collected evidence for intracellular cholesterol concentrations being less important in viral life cycles as compared to the spatial distribution of the lipid. Various routes of cholesterol trafficking were found to be hijacked in viral infections with respect to organelle-endosome contact sites mediating cholesterol shuttling. Thus, re-distribution of cellular cholesterol in the context of viral infections requires more attention in ongoing research. As a final aim, a pan-antiviral treatment could be found just within the transport and re-adjustment of local cholesterol concentrations. Thus, we aimed to emphasize the importance of the regulatory roles the endosomal system fulfils herein and hope to stimulate research in this field.
Collapse
Affiliation(s)
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
210
|
Foot-and-Mouth Disease Virus VP3 Protein Acts as a Critical Proinflammatory Factor by Promoting Toll-Like Receptor 4-Mediated Signaling. J Virol 2021; 95:e0112021. [PMID: 34524915 PMCID: PMC8577349 DOI: 10.1128/jvi.01120-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Foot-and-mouth disease virus (FMDV) infection in cloven-hoofed animals causes severe inflammatory symptoms, including blisters on the oral mucosa, hoof, and breast; however, the molecular mechanism underlying the inflammatory response is unclear. In this study, we provide the first evidence that the FMDV protein VP3 activates lipopolysaccharide-triggered Toll-like receptor 4 (TLR4) signaling. FMDV VP3 increased the expression of TLR4 by downregulating the expression of the lysozyme-related protein Rab7b. Additionally, Rab7b can interact with VP3 to promote the replication of FMDV. Our findings suggested that VP3 regulates the Rab7b-TLR4 axis to mediate the inflammatory response to FMDV. IMPORTANCE Foot-and-mouth disease virus (FMDV) infection causes a severe inflammatory response in cloven-hoofed animals, such as pigs, cattle, and sheep, with typical clinical manifestations of high fever, numerous blisters on the oral mucosa, hoof, and breast, as well as myocarditis (tigroid heart). However, the mechanism underlying the inflammatory response caused by FMDV is enigmatic. In this study, we identified the VP3 protein of FMDV as an important proinflammatory factor. Mechanistically, VP3 interacted with TLR4 to promote TLR4 expression by inhibiting the expression of the lysozyme-related protein Rab7b. Our findings suggest that FMDV VP3 is a major proinflammatory factor in FMDV-infected hosts.
Collapse
|
211
|
Sartorius R, Trovato M, Manco R, D'Apice L, De Berardinis P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines 2021; 6:127. [PMID: 34711839 PMCID: PMC8553822 DOI: 10.1038/s41541-021-00391-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are transmembrane proteins belonging to the family of pattern-recognition receptors. They function as sensors of invading pathogens through recognition of pathogen-associated molecular patterns. After their engagement by microbial ligands, TLRs trigger downstream signaling pathways that culminate into transcriptional upregulation of genes involved in immune defense. Here we provide an updated overview on members of the TLR family and we focus on their role in antiviral response. Understanding of innate sensing and signaling of viruses triggered by these receptors would provide useful knowledge to prompt the development of vaccines able to elicit effective and long-lasting immune responses. We describe the mechanisms developed by viral pathogens to escape from immune surveillance mediated by TLRs and finally discuss how TLR/virus interplay might be exploited to guide the design of innovative vaccine platforms.
Collapse
Affiliation(s)
- Rossella Sartorius
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | - Maria Trovato
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Roberta Manco
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy
| | - Luciana D'Apice
- Institute of Biochemistry and Cell Biology, C.N.R., Via Pietro Castellino 111, 80131, Naples, Italy.
| | | |
Collapse
|
212
|
Yang CH, Hwang CF, Chuang JH, Lian WS, Wang FS, Yang MY. Systemic toll-like receptor 9 agonist CpG oligodeoxynucleotides exacerbates aminoglycoside ototoxicity. Hear Res 2021; 411:108368. [PMID: 34678647 DOI: 10.1016/j.heares.2021.108368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 02/08/2023]
Abstract
The Toll-like receptor (TLR) signaling pathway is the key regulator of the innate immune system in response to systemic infection. Several studies have reported that the systemic TLR4 agonist lipopolysaccharide exacerbates aminoglycoside ototoxicity, but the influence of virus-associated TLR7 and TLR9 signaling cascades on the cochlea is unclear. The present study aimed to investigate the auditory effects of systemic TLR7 and TLR9 agonists during chronic kanamycin treatment. CBA/CaJ mice received the TLR7 agonist gardiquimod or TLR9 agonist CpG oligodeoxynucleotides (ODN) one day before kanamycin injection and on the 5th and 10th days during a 14-day course of kanamycin treatment. We observed that systemic gardiquimod or CpG ODN alone did not affect the baseline auditory brainstem response (ABR) threshold. Three weeks after kanamycin treatment, gardiquimod did not significantly change ABR threshold shifts, whereas CpG ODN significantly increased kanamycin-induced ABR threshold shifts. Furthermore, outer hair cell (OHC) evaluation revealed that CpG ODN reduced distortion product otoacoustic emission amplitudes and increased kanamycin-induced OHC loss. CpG ODN significantly elevated cochlear Irf-7, Tnf-α, Il-1, and Il-6 transcript levels. In addition, an increased number of Iba-1+ cells, which represented activated macrophages, was observed in the cochlea treated with CpG ODN. Our results indicated that systemic CpG ODN exacerbated kanamycin-induced ototoxicity and increased cochlear inflammation. This study implies that patients with underlying virus infection may experience more severe aminoglycoside-induced hearing loss if it occurs.
Collapse
Affiliation(s)
- Chao-Hui Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chung-Feng Hwang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jiin-Haur Chuang
- Division of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| | - Wei-Shiung Lian
- Core Laboratory for Phenomics & Diagnostics, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Feng-Sheng Wang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan; Core Laboratory for Phenomics & Diagnostics, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Ming-Yu Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan.
| |
Collapse
|
213
|
Guoying L, Li L, Siyue Y, Lei L, Guangliang C. Total Saponin of Dioscorea collettii Attenuates MSU Crystal-Induced Inflammation by Inhibiting the Activation of the TLR4/NF- κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:8728473. [PMID: 34721647 PMCID: PMC8550844 DOI: 10.1155/2021/8728473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Rhizomes from Dioscorea collettii are extensively used in traditional medicine for the treatment of arthritic diseases, particularly gouty arthritis (GA). This study aims to investigate whether the total saponin of Dioscorea collettii (TSD) can attenuate monosodium urate (MSU) crystal-induced inflammatory effects by suppressing the activation of the TLR4/NF-κB signaling pathway in vivo and in vitro. METHODS Seventy-two male Wistar rats and THP-1 cells were used in this study. Pathological examination was used to examine the ankle joints of rats. The expression levels of TLR4, NF-κB, MyD88, and IL-1β were detected by qRT-PCR, Western blotting, or immunofluorescence. RESULTS Compared with those in the normal group, the ankle joints of rats in the model group exhibited significant swelling, synovial tissue hyperplasia, inflammatory cell infiltration, and increased expression of IL-1β protein. The joint swelling degree of rats in the TSD high- and medium-dose groups and the colchicine group was significantly decreased, and the histopathology was obviously improved. TSD and colchicine reduced the levels of IL-1β and TNF-α in synovial fluid. They also decreased the mRNA expression of TLR4, NF-κB, and IL-1β in rat joint synovial tissue and the protein expression of TLR4, MyD88, and NF-κB. NF-κB protein expression in both the cytoplasm and nuclei of THP-1 cells showed the opposite trend. Furthermore, immunofluorescence showed that TSD reduced the nuclear translocation of NF-κBp65 in the model group. CONCLUSION TSD exhibits an anti-inflammatory effect in the MSU-induced inflammation model, and the mechanism may be to reduce the production of cytokines by inhibiting the activation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Li Guoying
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, Anhui, China
| | - Li Li
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, Anhui, China
| | - Yang Siyue
- College of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lv Lei
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, Anhui, China
| | - Chen Guangliang
- College of Integrative Medicine, Anhui University of Chinese Medicine, Hefei 230012, Anhui, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, Anhui, China
| |
Collapse
|
214
|
In Silico Modeling as a Perspective in Developing Potential Vaccine Candidates and Therapeutics for COVID-19. COATINGS 2021. [DOI: 10.3390/coatings11111273] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The potential of computational models to identify new therapeutics and repurpose existing drugs has gained significance in recent times. The current ‘COVID-19’ pandemic caused by the new SARS CoV2 virus has affected over 200 million people and caused over 4 million deaths. The enormity and the consequences of this viral infection have fueled the research community to identify drugs or vaccines through a relatively expeditious process. The availability of high-throughput datasets has cultivated new strategies for drug development and can provide the foundation towards effective therapy options. Molecular modeling methods using structure-based or computer-aided virtual screening can potentially be employed as research guides to identify novel antiviral agents. This review focuses on in-silico modeling of the potential therapeutic candidates against SARS CoVs, in addition to strategies for vaccine design. Here, we particularly focus on the recently published SARS CoV main protease (Mpro) active site, the RNA-dependent RNA polymerase (RdRp) of SARS CoV2, and the spike S-protein as potential targets for vaccine development. This review can offer future perspectives for further research and the development of COVID-19 therapies via the design of new drug candidates and multi-epitopic vaccines and through the repurposing of either approved drugs or drugs under clinical trial.
Collapse
|
215
|
How dendritic cells sense and respond to viral infections. Clin Sci (Lond) 2021; 135:2217-2242. [PMID: 34623425 DOI: 10.1042/cs20210577] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022]
Abstract
The ability of dendritic cells (DCs) to sense viral pathogens and orchestrate a proper immune response makes them one of the key players in antiviral immunity. Different DC subsets have complementing functions during viral infections, some specialize in antigen presentation and cross-presentation and others in the production of cytokines with antiviral activity, such as type I interferons. In this review, we summarize the latest updates concerning the role of DCs in viral infections, with particular focus on the complex interplay between DC subsets and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Despite being initiated by a vast array of immune receptors, DC-mediated antiviral responses often converge towards the same endpoint, that is the production of proinflammatory cytokines and the activation of an adaptive immune response. Nonetheless, the inherent migratory properties of DCs make them a double-edged sword and often viral recognition by DCs results in further viral dissemination. Here we illustrate these various aspects of the antiviral functions of DCs and also provide a brief overview of novel antiviral vaccination strategies based on DCs targeting.
Collapse
|
216
|
Olech M, Ropka-Molik K, Szmatoła T, Piórkowska K, Kuźmak J. Transcriptome Analysis for Genes Associated with Small Ruminant Lentiviruses Infection in Goats of Carpathian Breed. Viruses 2021; 13:v13102054. [PMID: 34696484 DOI: 10.3390/v13102054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Small ruminant lentiviruses (SRLV) are economically important viral pathogens of sheep and goats. SRLV infection may interfere in the innate and adaptive immunity of the host, and genes associated with resistance or susceptibility to infection with SRLV have not been fully recognized. The presence of animals with relatively high and low proviral load suggests that some host factors are involved in the control of virus replication. To better understand the role of the genes involved in the host response to SRLV infection, RNA sequencing (RNA-seq) method was used to compare whole gene expression profiles in goats carrying both a high (HPL) and low (LPL) proviral load of SRLV and uninfected animals. Data enabled the identification of 1130 significant differentially expressed genes (DEGs) between control and LPL groups: 411 between control and HPL groups and 1434 DEGs between HPL and LPL groups. DEGs detected between the control group and groups with a proviral load were found to be significantly enriched in several gene ontology (GO) terms, including an integral component of membrane, extracellular region, response to growth factor, inflammatory and innate immune response, transmembrane signaling receptor activity, myeloid differentiation primary response gene 88 (MyD88)-dependent toll-like receptor signaling pathway as well as regulation of cytokine secretion. Our results also demonstrated significant deregulation of selected pathways in response to viral infection. The presence of SRLV proviral load in blood resulted in the modification of gene expression belonging to the toll-like receptor signaling pathway, the tumor necrosis factor (TNF) signaling pathway, the cytokine-cytokine receptor interaction, the phagosome, the Ras signaling pathway, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) (PI3K-Akt) signaling pathway and rheumatoid arthritis. It is worth mentioning that the most predominant in all pathways were genes represented by toll-like receptors, tubulins, growth factors as well as interferon gamma receptors. DEGs detected between LPL and HPL groups were found to have significantly enriched regulation of signaling receptor activity, the response to toxic substances, nicotinamide adenine dinucleotide (NADH) dehydrogenase complex assembly, cytokine production, vesicle, and vacuole organization. In turn, the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway tool classified DEGs that enrich molecular processes such as B and T-cell receptor signaling pathways, natural killer cell-mediated cytotoxicity, Fc gamma R-mediated phagocytosis, toll-like receptor signaling pathways, TNF, mammalian target of rapamycin (mTOR) signaling and forkhead box O (Foxo) signaling pathways, etc. Our data indicate that changes in SRLV proviral load induced altered expression of genes related to different biological processes such as immune response, inflammation, cell locomotion, and cytokine production. These findings provide significant insights into defense mechanisms against SRLV infection. Furthermore, these data can be useful to develop strategies against SRLV infection by selection of animals with reduced SRLV proviral concentration that may lead to a reduction in the spread of the virus.
Collapse
Affiliation(s)
- Monika Olech
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Kraków, Poland
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Kraków, Poland
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Rędzina 1c, 30-248 Kraków, Poland
| | - Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Kraków, Poland
| | - Jacek Kuźmak
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland
| |
Collapse
|
217
|
Abdelmoaty MM, Yeapuri P, Machhi J, Olson KE, Shahjin F, Kumar V, Zhou Y, Liang J, Pandey K, Acharya A, Byrareddy SN, Mosley RL, Gendelman HE. Defining the Innate Immune Responses for SARS-CoV-2-Human Macrophage Interactions. Front Immunol 2021; 12:741502. [PMID: 34671355 PMCID: PMC8521106 DOI: 10.3389/fimmu.2021.741502] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Host innate immune response follows severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and it is the driver of the acute respiratory distress syndrome (ARDS) amongst other inflammatory end-organ morbidities. Such life-threatening coronavirus disease 2019 (COVID-19) is heralded by virus-induced activation of mononuclear phagocytes (MPs; monocytes, macrophages, and dendritic cells). MPs play substantial roles in aberrant immune secretory activities affecting profound systemic inflammation and end-organ malfunctions. All follow the presence of persistent viral components and virions without evidence of viral replication. To elucidate SARS-CoV-2-MP interactions we investigated transcriptomic and proteomic profiles of human monocyte-derived macrophages. While expression of the SARS-CoV-2 receptor, the angiotensin-converting enzyme 2, paralleled monocyte-macrophage differentiation, it failed to affect productive viral infection. In contrast, simple macrophage viral exposure led to robust pro-inflammatory cytokine and chemokine expression but attenuated type I interferon (IFN) activity. Both paralleled dysregulation of innate immune signaling pathways, specifically those linked to IFN. We conclude that the SARS-CoV-2-infected host mounts a robust innate immune response characterized by a pro-inflammatory storm heralding end-organ tissue damage.
Collapse
Affiliation(s)
- Mai M. Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Pravin Yeapuri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core, University of Nebraska Medical Center, Omaha, NE, United States
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jingjing Liang
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
218
|
Farooq M, Batool M, Kim MS, Choi S. Toll-Like Receptors as a Therapeutic Target in the Era of Immunotherapies. Front Cell Dev Biol 2021; 9:756315. [PMID: 34671606 PMCID: PMC8522911 DOI: 10.3389/fcell.2021.756315] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) are the pattern recognition receptors, which are activated by foreign and host molecules in order to initiate the immune response. They play a crucial role in the regulation of innate immunity, and several studies have shown their importance in bacterial, viral, and fungal infections, autoimmune diseases, and cancers. The consensus view from an immunological perspective is that TLR agonists can serve either as a possible therapeutic agent or as a vaccine adjuvant toward cancers or infectious diseases and that TLR inhibitors may be a promising approach to the treatment of autoimmune diseases, some cancers, bacterial, and viral infections. These notions are based on the fact that TLR agonists stimulate the secretion of proinflammatory cytokines and in general, the development of proinflammatory responses. Some of the TLR-based inhibitory agents have shown to be efficacious in preclinical models and have now entered clinical trials. Therefore, TLRs seem to hold the potential to serve as a perfect target in the era of immunotherapies. We offer a perspective on TLR-based therapeutics that sheds light on their usefulness and on combination therapies. We also highlight various therapeutics that are in the discovery phase or in clinical trials.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Suwon, South Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Suwon, South Korea
| |
Collapse
|
219
|
Li K, Lu X, Liu S, Wu X, Xie Y, Zheng X. Boron-incorporated micro/nano-topographical calcium silicate coating dictates osteo/angio-genesis and inflammatory response toward enhanced osseointegration. Biol Trace Elem Res 2021; 199:3801-3816. [PMID: 33405083 DOI: 10.1007/s12011-020-02517-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Orthopedic implant coatings with optimal surface features to achieve favorable osteo/angio-genesis and inflammatory response would be of great importance. However, to date, few coatings are capable of fully satisfying these requirements. In this work, to take advantage of the structural complexity of micro/nano-topography and benefits of biological trace elements, two types of boron-containing nanostructures (nanoflakes and nanolamellars) were introduced onto plasma-sprayed calcium silicate (F-BCS and L-BCS) coatings via hydrothermal treatment. The C-CS coating using deionized water as hydrothermal medium served as control. Boron-incorporated CS coating stimulated osteoblastic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Specifically, the combination of β1 integrin-vinculin-mediated cell spreading and activation of bone morphogenetic protein signaling pathway acted synergistically to cause significant upregulation of runt-related transcription factor 2 (RUNX2) protein and Runx2 gene expression in BMSCs on the F-BCS coating surface, which induced the transcription of downstream osteogenic differentiation marker genes. F-BCS coating allowed specific boron ion release, which favored angiogenesis as evidenced by the enhanced migration and tube formation of human umbilical vein endothelial cells in the coating extract. Boron-incorporated coatings significantly suppressed the expression of toll-like receptor adaptor genes in RAW264.7 macrophages and subsequently the degradation of nuclear factor-κB inhibitor α, accompanied by the inactivation of the downstream pro-inflammatory genes. In vivo experiments confirmed that F-BCS-coated Ti implant possessed enhanced osseointegration compared with L-BCS- and C-CS-coated implants. These data highlighted the synergistic effect of specific nanotopography and boron release from orthopedic implant coating on improvement of osseointegration.
Collapse
Affiliation(s)
- Kai Li
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xiang Lu
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Shiwei Liu
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xiaodong Wu
- Department of Orthopedic, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China.
| | - Youtao Xie
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xuebin Zheng
- Key Laboratory of Inorganic Coating Materials CAS, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, People's Republic of China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, People's Republic of China.
| |
Collapse
|
220
|
Cantwell AM, Singh H, Platt M, Yu Y, Lin YH, Ikeno Y, Hubbard G, Xiang Y, Gonzalez-Juarbe N, Dube PH. Kinetic Multi-omic Analysis of Responses to SARS-CoV-2 Infection in a Model of Severe COVID-19. J Virol 2021; 95:e0101021. [PMID: 34319784 PMCID: PMC8475517 DOI: 10.1128/jvi.01010-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
The host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is poorly understood due to a lack of an animal model that recapitulates severe human disease. Here, we report a Syrian hamster model that develops progressive lethal pulmonary disease that closely mimics severe coronavirus disease 2019 (COVID-19). We evaluated host responses using a multi-omic, multiorgan approach to define proteome, phosphoproteome, and transcriptome changes. These data revealed both type I and type II interferon-stimulated gene and protein expression along with a progressive increase in chemokines, monocytes, and neutrophil-associated molecules throughout the course of infection that peaked in the later time points correlating with a rapidly developing diffuse alveolar destruction and pneumonia that persisted in the absence of active viral infection. Extrapulmonary proteome and phosphoproteome remodeling was detected in the heart and kidneys following viral infection. Together, our results provide a kinetic overview of multiorgan host responses to severe SARS-CoV-2 infection in vivo. IMPORTANCE The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has created an urgent need to understand the pathogenesis of this infection. These efforts have been impaired by the lack of animal models that recapitulate severe coronavirus disease 2019 (COVID-19). Here, we report a hamster model that develops severe COVID-19-like disease following infection with human isolates of SARS-CoV-2. To better understand pathogenesis, we evaluated changes in gene transcription and protein expression over the course of infection to provide an integrated multiorgan kinetic analysis of the host response to infection. These data reveal a dynamic innate immune response to infection and corresponding immune pathologies consistent with severe human disease. Altogether, this model will be useful for understanding the pathogenesis of severe COVID-19 and for testing interventions.
Collapse
Affiliation(s)
- Angelene M. Cantwell
- Department of Microbiology, Immunology, and Molecular Genetics, UT-Health San Antonio, San Antonio, Texas, USA
- Select Agent Research Core, UT-Health San Antonio, San Antonio, Texas, USA
| | - Harinder Singh
- Infectious Disease and Genomic Medicine Group, J. Craig Venter Institute, Rockville, Maryland, USA
| | - Maryann Platt
- Infectious Disease and Genomic Medicine Group, J. Craig Venter Institute, Rockville, Maryland, USA
| | - Yanbao Yu
- Infectious Disease and Genomic Medicine Group, J. Craig Venter Institute, Rockville, Maryland, USA
| | - Yi-Han Lin
- Infectious Disease and Genomic Medicine Group, J. Craig Venter Institute, Rockville, Maryland, USA
| | - Yuji Ikeno
- Barshop Institute for Longevity and Aging Studies, UT-Health San Antonio, San Antonio, Texas, USA
- Department of Pathology, UT-Health San Antonio, San Antonio, Texas, USA
| | - Gene Hubbard
- Barshop Institute for Longevity and Aging Studies, UT-Health San Antonio, San Antonio, Texas, USA
| | - Yan Xiang
- Department of Microbiology, Immunology, and Molecular Genetics, UT-Health San Antonio, San Antonio, Texas, USA
- Select Agent Research Core, UT-Health San Antonio, San Antonio, Texas, USA
| | - Norberto Gonzalez-Juarbe
- Infectious Disease and Genomic Medicine Group, J. Craig Venter Institute, Rockville, Maryland, USA
| | - Peter H. Dube
- Department of Microbiology, Immunology, and Molecular Genetics, UT-Health San Antonio, San Antonio, Texas, USA
- Select Agent Research Core, UT-Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
221
|
Tripathi U, Nchioua R, Prata LGPL, Zhu Y, Gerdes EOW, Giorgadze N, Pirtskhalava T, Parker E, Xue A, Espindola-Netto JM, Stenger S, Robbins PD, Niedernhofer LJ, Dickinson SL, Allison DB, Kirchhoff F, Sparrer KMJ, Tchkonia T, Kirkland JL. SARS-CoV-2 causes senescence in human cells and exacerbates the senescence-associated secretory phenotype through TLR-3. Aging (Albany NY) 2021; 13:21838-21854. [PMID: 34531331 PMCID: PMC8507266 DOI: 10.18632/aging.203560] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Senescent cells, which arise due to damage-associated signals, are apoptosis-resistant and can express a pro-inflammatory, tissue-destructive senescence-associated secretory phenotype (SASP). We recently reported that a component of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) surface protein, S1, can amplify the SASP of senescent cultured human cells and that a related mouse β-coronavirus, mouse hepatitis virus (MHV), increases SASP factors and senescent cell burden in infected mice. Here, we show that SARS-CoV-2 induces senescence in human non-senescent cells and exacerbates the SASP in human senescent cells through Toll-like receptor-3 (TLR-3). TLR-3, which senses viral RNA, was increased in human senescent compared to non-senescent cells. Notably, genetically or pharmacologically inhibiting TLR-3 prevented senescence induction and SASP amplification by SARS-CoV-2 or Spike pseudotyped virus. While an artificial TLR-3 agonist alone was not sufficient to induce senescence, it amplified the SASP in senescent human cells. Consistent with these findings, lung p16INK4a+ senescent cell burden was higher in patients who died from acute SARS-CoV-2 infection than other causes. Our results suggest that induction of cellular senescence and SASP amplification through TLR-3 contribute to SARS-CoV-2 morbidity, indicating that clinical trials of senolytics and/or SASP/TLR-3 inhibitors for alleviating acute and long-term SARS-CoV-2 sequelae are warranted.
Collapse
Affiliation(s)
- Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Rayhane Nchioua
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | | | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Bioengineering, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Erik Parker
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University-Bloomington, Bloomington, IN 47405, USA
| | - Ailing Xue
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, Ulm University Medical Center, Ulm 89081, Germany
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Stephanie L. Dickinson
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University-Bloomington, Bloomington, IN 47405, USA
| | - David B. Allison
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University-Bloomington, Bloomington, IN 47405, USA
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | | | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Bioengineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Bioengineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
222
|
Innate Immune Evasion of Porcine Epidemic Diarrhea Virus through Degradation of F-box and WD repeat domain-containing 7 protein via Ubiquitin-proteasome Pathway. J Virol 2021; 96:e0088921. [PMID: 34495699 DOI: 10.1128/jvi.00889-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes a porcine disease associated with swine epidemic diarrhea. Different antagonistic strategies have been identified, and the mechanism by which PEDV infection impairs the production of interferon (IFN) and delays the activation of the IFN response to escape host innate immunity has been determined, but the pathogenic mechanisms of PEDV infection remain enigmatic. Our preliminary results revealed that endogenous F-box and WD repeat domain-containing 7 (FBXW7), the substrate recognition component of the SCF-type E3 ubiquitin ligase, is downregulated in PEDV-infected Vero E6 cells, according to the results from an isobaric tags for relative and absolute quantification (iTRAQ) analysis. Overexpression of FBXW7 in target cells makes them more resistant to PEDV infection, whereas ablation of FBXW7 expression by small interfering RNA (siRNA) significantly promotes PEDV infection. In addition, FBXW7 was verified as an innate antiviral factor capable of enhancing the expression of RIG-I and TBK1, and it was found to induce interferon-stimulated genes (ISGs), which led to an elevated antiviral state of the host cells. Moreover, we revealed that PEDV nonstructural protein 2 (nsp2) interacts with FBXW7 and targets FBXW7 for degradation through the K48-linked ubiquitin-proteasome pathway. Consistent with the results proven in vitro, FBXW7 reduction was also confirmed in different intestinal tissues from PEDV-infected specific-pathogen-free (SPF) pigs. Taken together, the data indicated that PEDV has evolved with a distinct antagonistic strategy to circumvent the host antiviral response by targeting the ubiquitin-proteasome-mediated degradation of FBXW7. Our findings provide novel insights into PEDV infection and pathogenesis. IMPORTANCE To counteract the host antiviral defenses, most viruses, including coronaviruses, have evolved with diverse strategies to dampen host IFN-mediated antiviral response, wither by interfering with or evading specific host regulators at multiple steps of this response. In this study, a novel antagonistic strategy was revealed showing that PEDV infection could circumvent the host innate response by targeted degradation of endogenous FBXW7 in target cells, a process that was verified to be a positive modulator for the host innate immune system. Degradation of FBXW7 hampers host innate antiviral activation and facilitates PEDV replication. Our findings reveal a new mechanism exploited by PEDV to suppress the host antiviral response.
Collapse
|
223
|
Estebanez EB, Alconero LL, Fernández BJ, Marguello MG, Caro JCL, Vallejo JD, Sampedro MF, Cacho PM, Espiga CR, Saiz MMG. The effectiveness of early colchicine administration in patients over 60 years old with high risk of developing severe pulmonary complications associated with coronavirus pneumonia SARS-CoV-2 (COVID-19): study protocol for an investigator-driven randomized controlled clinical trial in primary health care-COLCHICOVID study. Trials 2021; 22:590. [PMID: 34488841 PMCID: PMC8419390 DOI: 10.1186/s13063-021-05544-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/13/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND There is no strong evidence that any drug is beneficial either for the treatment of SARS-CoV-2 disease or for post-exposure prophylaxis. Therefore, clinical research is crucial to generate results and evaluate strategies against COVID-19. Primary care (PC) centers, the first level of care in the health system, are in a favorable position to carry out clinical trials (CD), as they work with a large volume of patients with varied profiles (from acute to chronic pathologies). During the COVID-19 pandemic, the need for hospital admission and mortality is higher in people > 60 years. Therefore, this is a target population to try to reduce the serious complications and lethality of COVID pneumonia and to avoid overloading the hospital system. Given the pharmacological properties of colchicine (anti-inflammatory and anti-fibrotic, possible inhibition of viral replication, and inhibitory effect on coagulation activation), early treatment with colchicine may reduce the rate of death and serious pulmonary complications from COVID-19 in vulnerable patients. METHODS The COLCHICOVID study is a randomized, multicenter, controlled, open-label parallel group (2:1 ratio), phase III clinical trial to investigate the efficacy of early administration of colchicine in reducing the development of severe pulmonary complications associated with COVID-19 infection in patients over 60 years of age with at-risk comorbidities. DISCUSSION This is a pragmatic clinical trial, adapted to usual clinical practice. The demonstration that early administration of colchicine has clinical effectiveness in reducing the complications of SARS-CoV-2 infection in a population highly susceptible may mitigate the health crisis and prevent the collapse of the health system in the successive waves of the coronavirus pandemic. In addition, colchicine is a well-known medicine, simple to use in the primary care setting and with a low cost for the health system. TRIAL REGISTRATION ClinicalTrials.gov NCT04416334 . Registered on 4 June 2020. Protocol version: v 3.0, dated 22 September 2020.
Collapse
Affiliation(s)
- Elena Bustamante Estebanez
- Management of primary health care centers, Area I, Area II, Area III and Area IV, Servicio Cantabro de Salud, C. Vargas 57, 39010 Santander, Cantabria Spain
| | - Lucía Lavín Alconero
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Clinical Trials Agency Valdecilla-IDIVAL, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
| | - Beatriz Josa Fernández
- Management of primary health care centers, Area I, Area II, Area III and Area IV, Servicio Cantabro de Salud, C. Vargas 57, 39010 Santander, Cantabria Spain
| | - Monica Gozalo Marguello
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Department of Microbiology, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
| | - Juan Carlos López Caro
- Management of primary health care centers, Area I, Area II, Area III and Area IV, Servicio Cantabro de Salud, C. Vargas 57, 39010 Santander, Cantabria Spain
| | - Jonathan Diez Vallejo
- Management of primary health care centers, Area I, Area II, Area III and Area IV, Servicio Cantabro de Salud, C. Vargas 57, 39010 Santander, Cantabria Spain
| | - Marta Fernandez Sampedro
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Department of Infectious Diseases, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
| | - Pedro Muñoz Cacho
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Department of Community Health, Servicio Cantabro de Salud, C. Luis Vicente de Velasco 1, 39011 Santander, Cantabria Spain
| | - Carlos Richard Espiga
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Emeritus Doctor, Department of Hematology, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
| | - María Mar García Saiz
- Marqués de Valdecilla Research Institute (IDIVAL), s/n, Calle Cardenal Herrera Oria, 39012 Santander, Cantabria Spain
- Clinical Trials Agency Valdecilla-IDIVAL, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
- Department of Clinical Pharmacology, Marqués de Valdecilla University Hospital, Av. Valdecilla, 25, 39008 Santander, Cantabria Spain
| |
Collapse
|
224
|
Vaghari-Tabari M, Mohammadzadeh I, Qujeq D, Majidinia M, Alemi F, Younesi S, Mahmoodpoor A, Maleki M, Yousefi B, Asemi Z. Vitamin D in respiratory viral infections: a key immune modulator? Crit Rev Food Sci Nutr 2021; 63:2231-2246. [PMID: 34470511 DOI: 10.1080/10408398.2021.1972407] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Respiratory viral infections are common respiratory diseases. Influenza viruses, RSV and SARS-COV2 have the potential to cause severe respiratory infections. Numerous studies have shown that unregulated immune response to these viruses can cause excessive inflammation and tissue damage. Therefore, regulating the antiviral immune response in the respiratory tract is of importance. In this regard, recent years studies have emphasized the importance of vitamin D in respiratory viral infections. Although, the most well-known role of vitamin D is to regulate the metabolism of phosphorus and calcium, it has been shown that this vitamin has other important functions. One of these functions is immune regulation. Vitamin D can regulate the antiviral immune response in the respiratory tract in order to provide an effective defense against respiratory viral infections and prevention from excessive inflammatory response and tissue damage. In addition, this vitamin has preventive effects against respiratory viral infections. Some studies during the COVID-19 pandemic have shown that vitamin D deficiency may be associated with a higher risk of mortality and sever disease in patients with COVID-19. Since, more attention has recently been focused on vitamin D. In this article, after a brief overview of the antiviral immune response in the respiratory system, we will review the role of vitamin D in regulating the antiviral immune response comprehensively. Then we will discuss the importance of this vitamin in influenza, RSV, and COVID-19.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Iraj Mohammadzadeh
- Non-Communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran.,Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Forough Alemi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Younesi
- Schoole of Health and Biomedical Sciences, RMIT University, Melborne, VIC, Australia
| | - Ata Mahmoodpoor
- Department of Anesthesiology and Intensive Care, School of Medicine, Tabriz University of Medical Science and Health Services, Tabriz, Iran
| | - Masomeh Maleki
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
225
|
Dyavar SR, Singh R, Emani R, Pawar GP, Chaudhari VD, Podany AT, Avedissian SN, Fletcher CV, Salunke DB. Role of toll-like receptor 7/8 pathways in regulation of interferon response and inflammatory mediators during SARS-CoV2 infection and potential therapeutic options. Biomed Pharmacother 2021; 141:111794. [PMID: 34153851 PMCID: PMC8189763 DOI: 10.1016/j.biopha.2021.111794] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/17/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) is the causative agent of Corona Virus Disease 2019 (COVID-19). Lower production of type I and III interferons and higher levels of inflammatory mediators upon SARS-CoV2 infection contribute to COVID-19 pathogenesis. Optimal interferon production and controlled inflammation are essential to limit COVID-19 pathogenesis. However, the aggravated inflammatory response observed in COVID-19 patients causes severe damage to the host and frequently advances to acute respiratory distress syndrome (ARDS). Toll-like receptor 7 and 8 (TLR7/8) signaling pathways play a central role in regulating induction of interferons (IFNs) and inflammatory mediators in dendritic cells. Controlled inflammation is possible through regulation of TLR mediated response without influencing interferon production to reduce COVID-19 pathogenesis. This review focuses on inflammatory mediators that contribute to pathogenic effects and the role of TLR pathways in the induction of interferon and inflammatory mediators and their contribution to COVID-19 pathogenesis. We conclude that potential TLR7/8 agonists inducing antiviral interferon response and controlling inflammation are important therapeutic options to effectively eliminate SARS-CoV2 induced pathogenesis. Ongoing and future studies may provide additional evidence on their safety and efficacy to treat COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Shetty Ravi Dyavar
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA.
| | - Rahul Singh
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Rohini Emani
- Buck Institute for Research on Ageing, Novato, CA, USA
| | - Ganesh P Pawar
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India
| | - Vinod D Chaudhari
- Division of Medicinal Chemistry, CSIR-Institute of Microbiology Technology Chandigarh, Sector-39A, Chandigarh,160036, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anthony T Podany
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Sean N Avedissian
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Courtney V Fletcher
- University of Nebraska Medical Center (UNMC) Center for Drug Discovery, UNMC, Omaha, NE 68198, USA
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India; National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
226
|
Viral and Bacterial Co-Infections in the Lungs: Dangerous Liaisons. Viruses 2021; 13:v13091725. [PMID: 34578306 PMCID: PMC8472850 DOI: 10.3390/v13091725] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
Respiratory tract infections constitute a significant public health problem, with a therapeutic arsenal that remains relatively limited and that is threatened by the emergence of antiviral and/or antibiotic resistance. Viral–bacterial co-infections are very often associated with the severity of these respiratory infections and have been explored mainly in the context of bacterial superinfections following primary influenza infection. This review summarizes our current knowledge of the mechanisms underlying these co-infections between respiratory viruses (influenza viruses, RSV, and SARS-CoV-2) and bacteria, at both the physiological and immunological levels. This review also explores the importance of the microbiome and the pathological context in the evolution of these respiratory tract co-infections and presents the different in vitro and in vivo experimental models available. A better understanding of the complex functional interactions between viruses/bacteria and host cells will allow the development of new, specific, and more effective diagnostic and therapeutic approaches.
Collapse
|
227
|
Guerrero-Arguero I, Tellez-Freitas CM, Weber KS, Berges BK, Robison RA, Pickett BE. Alphaviruses: Host pathogenesis, immune response, and vaccine & treatment updates. J Gen Virol 2021; 102. [PMID: 34435944 DOI: 10.1099/jgv.0.001644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human pathogens belonging to the Alphavirus genus, in the Togaviridae family, are transmitted primarily by mosquitoes. The signs and symptoms associated with these viruses include fever and polyarthralgia, defined as joint pain and inflammation, as well as encephalitis. In the last decade, our understanding of the interactions between members of the alphavirus genus and the human host has increased due to the re-appearance of the chikungunya virus (CHIKV) in Asia and Europe, as well as its emergence in the Americas. Alphaviruses affect host immunity through cytokines and the interferon response. Understanding alphavirus interactions with both the innate immune system as well as the various cells in the adaptive immune systems is critical to developing effective therapeutics. In this review, we summarize the latest research on alphavirus-host cell interactions, underlying infection mechanisms, and possible treatments.
Collapse
Affiliation(s)
- Israel Guerrero-Arguero
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Bradford K Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Brett E Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
228
|
Hanna J, Tipparaju P, Mulherkar T, Lin E, Mischley V, Kulkarni R, Bolton A, Byrareddy SN, Jain P. Risk Factors Associated with the Clinical Outcomes of COVID-19 and Its Variants in the Context of Cytokine Storm and Therapeutics/Vaccine Development Challenges. Vaccines (Basel) 2021; 9:938. [PMID: 34452063 PMCID: PMC8402745 DOI: 10.3390/vaccines9080938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/08/2021] [Accepted: 08/17/2021] [Indexed: 12/17/2022] Open
Abstract
The recent appearance of SARS-CoV-2 is responsible for the ongoing coronavirus disease 2019 (COVID-19) pandemic and has brought to light the importance of understanding this highly pathogenic agent to prevent future pandemics. This virus is from the same single-stranded positive-sense RNA family, Coronaviridae, as two other epidemic-causing viruses, SARS-CoV-1 and MERS-CoV. During this pandemic, one crucial focus highlighted by WHO has been to understand the risk factors that may contribute to disease severity and predict COVID-19 outcomes. In doing so, it is imperative to understand the virology of SARS-CoV-2 and the immunological response eliciting the clinical manifestation and progression of COVID-19. In this review, we provide clinical data-based analyses of how multiple risk factors (such as sex, race, HLA genotypes, blood groups, vitamin D deficiency, obesity, smoking, and asthma) contribute to the inflammatory overactivation and cytokine storm (frequently seen in COVID-19 patients) with a focus on the IL-6 pathway. We also draw comparisons to the virulence and pathophysiology of SARS and MERS to establish parallels in immune response and discuss the potential for therapeutic approaches that may limit disease progression in patients with higher risk profiles than others. Moreover, we cover the latest information on approved or upcoming COVID-19 vaccines. This paper also provides perspective on emerging variants and associated opportunistic infections such as black molds and fungus that have added to mortality in some parts of the world, such as India. This compilation of existing COVID-19 studies and data will provide an excellent referencing tool for the research, clinical, and public health communities.
Collapse
Affiliation(s)
- John Hanna
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Padmavathi Tipparaju
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Tania Mulherkar
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Edward Lin
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Victoria Mischley
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Ratuja Kulkarni
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Aliyah Bolton
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University College of Medicine Philadelphia, 2900 Queen Lane, Philadelphia, PA 19129, USA; (J.H.); (P.T.); (T.M.); (E.L.); (V.M.); (R.K.); (A.B.)
| |
Collapse
|
229
|
Deep survey for designing a vaccine against SARS-CoV-2 and its new mutations. Biologia (Bratisl) 2021; 76:3465-3476. [PMID: 34421121 PMCID: PMC8369332 DOI: 10.1007/s11756-021-00866-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023]
Abstract
The ongoing global pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has prompted worldwide vaccine development. Several vaccines have been authorized by WHO, FDA, or MOH of different countries. However, issues such as need for cold chain, price, and most importantly access problems have limited vaccine usage in some nations especially developing countries. Moreover, the vast global demand justifies further attempts for vaccine development. Multi-epitope polypeptide vaccines enjoy several key features including safety and lower production and transfer costs and could be designed by in silico tools. Spike protein (S), membrane protein (M), and nucleocapsid protein (N), the three major structural proteins of SARS-CoV-2, are ideal candidates for epitope selection. ORF3a (open reading frame3a), a transmembrane protein with pro-apoptotic functions, could be another proper target. Thus, a novel multi-epitope vaccine against SARS-CoV-2 was designed using these four proteins and LL37, a TLR3 agonist adjuvant, through different immunoinformatics and bioinformatics tools. The proposed multi-epitope vaccine is expected to induce robust humoral and cellular immune responses against SARS-CoV-2 with a population coverage of 76.92 % due to containing different immunodominant epitopes and LL37 adjuvant. Selecting epitopes derived from one functional and three structural proteins suggests the protective ability of the vaccine irrespective of probable virus mutations. The computationally observed proper interaction of LL37 with TLR3 implies its ability to induce immune responses effectively. Besides, it showed acceptable structural and physicochemical properties. The in-silico cloning results predicted its high efficiency production in Escherichia coli. Future experimental studies could further confirm its immunological efficacy.
Collapse
|
230
|
Bridges JP, Vladar EK, Huang H, Mason RJ. Respiratory epithelial cell responses to SARS-CoV-2 in COVID-19. Thorax 2021; 77:203-209. [PMID: 34404754 DOI: 10.1136/thoraxjnl-2021-217561] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
COVID-19 has different clinical stages, and effective therapy depends on the location and extent of the infection. The purpose of this review is to provide a background for understanding the progression of the disease throughout the pulmonary epithelium and discuss therapeutic options. The prime sites for infection that will be contrasted in this review are the conducting airways and the gas exchange portions of the lung. These two sites are characterised by distinct cellular composition and innate immune responses, which suggests the use of distinct therapeutic agents. In the nose, ciliated cells are the primary target cells for SARS-CoV-2 viral infection, replication and release. Infected cells shed their cilia, which disables mucociliary clearance. Evidence further points to a suppressed or incompletely activated innate immune response to SARS-CoV-2 infection in the upper airways. Asymptomatic individuals can still have a productive viral infection and infect others. In the gas exchange portion of the lung, the alveolar type II epithelial cell is the main target cell type. Cell death and marked innate immune response during infection likely contribute to alveolar damage and resultant acute respiratory distress syndrome. Alveolar infection can precipitate a hyperinflammatory state, which is the target of many therapies in severe COVID-19. Disease resolution in the lung is variable and may include scaring and long-term sequalae because the alveolar type II cells are also progenitor cells for the alveolar epithelium.
Collapse
Affiliation(s)
- James P Bridges
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA .,Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Eszter K Vladar
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine and Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Hua Huang
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Robert J Mason
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
231
|
Uribe-Diaz S, Nazeer N, Jaime J, Vargas-Bermúdez DS, Yitbarek A, Ahmed M, Rodríguez-Lecompte JC. Folic acid enhances proinflammatory and antiviral molecular pathways in chicken B-lymphocytes infected with a mild infectious bursal disease virus. Br Poult Sci 2021; 63:1-13. [PMID: 34287101 DOI: 10.1080/00071668.2021.1958298] [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: 12/14/2022]
Abstract
1. This study evaluated the effect of folic acid (FA) supplementation on the proinflammatory and antiviral molecular pathways of B-lymphocytes infected with a modified live IBDV (ST-12) mild vaccine strain during a timed post-infection analysis.2. A chicken B-lymphocytes (DT-40) cell line was cultured in triplicate at a concentration of 5 × 105 cells per well in 24-well plates; and was divided into three groups: 1: No virus, FA; 2: Virus, no FA; 3: Virus + FA at a concentration of 3.96 mM. The experiment was repeated three times.3. Cells in groups 2 and 3 were infected with a modified live IBDV (ST-12) mild vaccine strain at one multiplicity of infection (MOI: 1). After 1 hour of virus adsorption, samples were collected at 0, 3, 6, 12, 24 and 36 hours post-infection (hpi).4. The modified live IBDV (ST-12) mild vaccine strain triggered a B-lymphocyte specific immune response associated with the upregulation of genes involved in virus recognition (Igß), virus sensing (TLR-2, TLR-3, TLR-4 and MDA5), signal transduction and regulation (TRIF, MyD88 and IRF7), and the antiviral effector molecules (IFN-α, OAS, PKR, and viperin).5. FA supplementation modulated IBDV replication and regulated the proinflammatory and antiviral downstream molecular pathways.6. In conclusion, the low virulent pathotype serotype I modified live IBDV (ST-12) mild vaccine strain was able to trigger and mount an immune response in chicken B-lymphocytes without affecting B-cell viability. FA supplementation modulated B lymphocytes response and improved their innate immune proinflammatory and antiviral response molecular pathways.
Collapse
Affiliation(s)
- S Uribe-Diaz
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada.,Department of Chemistry, University of Prince Edward Island, Charlottetown, Canada
| | - N Nazeer
- Department of Chemistry, University of Prince Edward Island, Charlottetown, Canada
| | - J Jaime
- Bogotá. Faculty of Veterinary Medicine and Zootechnic. Animal Health Department; Infectiology and Immunology Research Centre (CI3V), National University of Colombia, Bogotá, Colombia
| | - D S Vargas-Bermúdez
- Bogotá. Faculty of Veterinary Medicine and Zootechnic. Animal Health Department; Infectiology and Immunology Research Centre (CI3V), National University of Colombia, Bogotá, Colombia
| | - A Yitbarek
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - M Ahmed
- Department of Chemistry, University of Prince Edward Island, Charlottetown, Canada
| | - J C Rodríguez-Lecompte
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, Canada
| |
Collapse
|
232
|
Lim KS, Cheng J, Putz A, Dong Q, Bai X, Beiki H, Tuggle CK, Dyck MK, Canada PG, Fortin F, Harding JCS, Plastow GS, Dekkers JCM. Quantitative analysis of the blood transcriptome of young healthy pigs and its relationship with subsequent disease resilience. BMC Genomics 2021; 22:614. [PMID: 34384354 PMCID: PMC8361860 DOI: 10.1186/s12864-021-07912-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background Disease resilience, which is the ability of an animal to maintain performance under disease, is important for pigs in commercial herds, where they are exposed to various pathogens. Our objective was to investigate population-level gene expression profiles in the blood of 912 healthy F1 barrows at ~ 27 days of age for associations with performance and health before and after their exposure to a natural polymicrobial disease challenge at ~ 43 days of age. Results Most significant (q < 0.20) associations of the level of expression of individual genes in blood of young healthy pigs were identified for concurrent growth rate and subjective health scores prior to the challenge, and for mortality, a combined mortality-treatment trait, and feed conversion rate after the challenge. Gene set enrichment analyses revealed three groups of gene ontology biological process terms that were related to disease resilience: 1) immune and stress response-related terms were enriched among genes whose increased expression was unfavorably associated with both pre- and post-challenge traits, 2) heme-related terms were enriched among genes that had favorable associations with both pre- and post-challenge traits, and 3) terms related to protein localization and viral gene expression were enriched among genes that were associated with reduced performance and health traits after but not before the challenge. Conclusions Gene expression profiles in blood from young healthy piglets provide insight into their performance when exposed to disease and other stressors. The expression of genes involved in stress response, heme metabolism, and baseline expression of host genes related to virus propagation were found to be associated with host response to disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07912-8.
Collapse
Affiliation(s)
- Kyu-Sang Lim
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | - Jian Cheng
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | - Austin Putz
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA.,Swine Business Unit, Hendrix Genetics, Boxmeer, 5831, CK, The Netherlands
| | - Qian Dong
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA.,Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xuechun Bai
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Hamid Beiki
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA
| | | | - Michael K Dyck
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Pig Gen Canada
- PigGen Canada Research Consortium, Guelph, Ontario, N1H4G8, Canada
| | - Frederic Fortin
- Centre de Développement du Porc du Québec Inc. (CDPQ), Québec City, QC, G1V 4M6, Canada
| | - John C S Harding
- Department of Large Animal Clinical Sciences, University of Saskatchewan, Saskatoon, SK, S7N 5A2, Canada
| | - Graham S Plastow
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
233
|
Hong Y, Lee J, Vu TH, Lee S, Lillehoj HS, Hong YH. Immunomodulatory effects of poly(I:C)-stimulated exosomes derived from chicken macrophages. Poult Sci 2021; 100:101247. [PMID: 34174563 PMCID: PMC8242060 DOI: 10.1016/j.psj.2021.101247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 05/02/2021] [Indexed: 11/13/2022] Open
Abstract
Exosomes are small membrane vesicles that contain proteins and nucleic acids derived from secretory cells and mediate intracellular communication. Immune cell-derived exosomes regulate immune responses and gene expression of recipient cells. Macrophages recognize viral dsRNA via Toll-like receptor 3, thereby inducing the activation of transcription factors such as interferon regulatory factor 3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In this study, we aimed to identify the immunomodulatory functions of exosomes derived from chicken macrophages (HD11) stimulated with polyinosinic-polycytidylic acid (poly[I:C]); exosomes were then delivered into HD11 cells and CU91 chicken T cells. Exosomes purified from poly(I:C)-activated macrophages stimulated the expression of type I interferons, proinflammatory cytokines, anti-inflammatory cytokines, and chemokines in HD11 and CU91 cells. Moreover, poly(I:C)-stimulated exosomes induced the NF-κB signaling pathway by phosphorylating TAK1 and NF-κB1. Therefore, we suggest that after the activation of Toll-like receptor 3 ligands following infection with dsRNA virus, chicken macrophages regulate the immune response of naive macrophages and T cells through the NF-κB signaling pathway. Furthermore, poly(I:C)-activated exosomes can be potentially utilized as immunostimulators.
Collapse
Affiliation(s)
- Yeojin Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Jiae Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sooyeon Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea.
| |
Collapse
|
234
|
Mahmud S, Rafi MO, Paul GK, Promi MM, Shimu MSS, Biswas S, Emran TB, Dhama K, Alyami SA, Moni MA, Saleh MA. Designing a multi-epitope vaccine candidate to combat MERS-CoV by employing an immunoinformatics approach. Sci Rep 2021; 11:15431. [PMID: 34326355 PMCID: PMC8322212 DOI: 10.1038/s41598-021-92176-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/21/2021] [Indexed: 01/26/2023] Open
Abstract
Currently, no approved vaccine is available against the Middle East respiratory syndrome coronavirus (MERS-CoV), which causes severe respiratory disease. The spike glycoprotein is typically considered a suitable target for MERS-CoV vaccine candidates. A computational strategy can be used to design an antigenic vaccine against a pathogen. Therefore, we used immunoinformatics and computational approaches to design a multi-epitope vaccine that targets the spike glycoprotein of MERS-CoV. After using numerous immunoinformatics tools and applying several immune filters, a poly-epitope vaccine was constructed comprising cytotoxic T-cell lymphocyte (CTL)-, helper T-cell lymphocyte (HTL)-, and interferon-gamma (IFN-γ)-inducing epitopes. In addition, various physicochemical, allergenic, and antigenic profiles were evaluated to confirm the immunogenicity and safety of the vaccine. Molecular interactions, binding affinities, and the thermodynamic stability of the vaccine were examined through molecular docking and dynamic simulation approaches, during which we identified a stable and strong interaction with Toll-like receptors (TLRs). In silico immune simulations were performed to assess the immune-response triggering capabilities of the vaccine. This computational analysis suggested that the proposed vaccine candidate would be structurally stable and capable of generating an effective immune response to combat viral infections; however, experimental evaluations remain necessary to verify the exact safety and immunogenicity profile of this vaccine.
Collapse
MESH Headings
- Computational Biology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Epitopes/immunology
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Humans
- Immunogenicity, Vaccine/immunology
- Middle East Respiratory Syndrome Coronavirus/genetics
- Middle East Respiratory Syndrome Coronavirus/immunology
- Middle East Respiratory Syndrome Coronavirus/pathogenicity
- Models, Molecular
- Molecular Docking Simulation
- Phylogeny
- Protein Binding
- Spike Glycoprotein, Coronavirus/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Vaccines/immunology
- Vaccines/pharmacology
- Vaccines, DNA
- Vaccines, Subunit/immunology
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Shafi Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh
| | - Md Oliullah Rafi
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Gobindo Kumar Paul
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh
| | - Maria Meha Promi
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh
| | - Mst Sharmin Sultana Shimu
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh
| | - Suvro Biswas
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, Uttar Pradesh, India
| | - Salem A Alyami
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11432, Saudi Arabia
| | - Mohammad Ali Moni
- Faculty of Medicine, WHO Collaborating Centre on eHealth, UNSW Digital Health, School of Public Health and Community Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| | - Md Abu Saleh
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, 6505, Bangladesh.
| |
Collapse
|
235
|
Bhattacharya S, Agarwal S, Shrimali NM, Guchhait P. Interplay between hypoxia and inflammation contributes to the progression and severity of respiratory viral diseases. Mol Aspects Med 2021; 81:101000. [PMID: 34294412 PMCID: PMC8287505 DOI: 10.1016/j.mam.2021.101000] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/07/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
History of pandemics is dominated by viral infections and specifically respiratory viral diseases like influenza and COVID-19. Lower respiratory tract infection is the fourth leading cause of death worldwide. Crosstalk between resultant inflammation and hypoxic microenvironment may impair ventilatory response of lungs. This reduces arterial partial pressure of oxygen, termed as hypoxemia, which is observed in a section of patients with respiratory virus infections including SARS-CoV-2 (COVID-19). In this review, we describe the interplay between inflammation and hypoxic microenvironment in respiratory viral infection and its contribution to disease pathogenesis.
Collapse
Affiliation(s)
- Sulagna Bhattacharya
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; School of Biotechnology, Kalinga Institute of Industrial Technology, Orissa, India
| | - Sakshi Agarwal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Nishith M Shrimali
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
236
|
Zhang Q, Yu J, Chen Q, Yan H, Du H, Luo W. Regulation of pathophysiological and tissue regenerative functions of MSCs mediated via the WNT signaling pathway (Review). Mol Med Rep 2021; 24:648. [PMID: 34278470 PMCID: PMC8299209 DOI: 10.3892/mmr.2021.12287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 06/22/2021] [Indexed: 12/18/2022] Open
Abstract
Tissues have remarkable natural capabilities to regenerate for the purpose of physiological turnover and repair of damage. Adult mesenchymal stem cells (MSCs) are well known for their unique self-renewal ability, pluripotency, homing potential, paracrine effects and immunomodulation. Advanced research of the unique properties of MSCs have opened up new horizons for tissue regenerative therapies. However, certain drawbacks of the application of MSCs, such as the low survival rate of transplanted MSCs, unsatisfactory efficiency and even failure to regenerate under an unbalanced microenvironment, are concerning with regards to their wider therapeutic applications. The activity of stem cells is mainly regulated by the anatomical niche; where they are placed during their clinical and therapeutic applications. Crosstalk between various niche signals maintains MSCs in homeostasis, in which the WNT signaling pathway plays vital roles. Several external or internal stimuli have been reported to interrupt the normal bioactivity of stem cells. The irreversible tissue loss that occurs during infection at the site of tissue grafting suggests an inhibitory effect mediated by microbial infections within MSC niches. In addition, MSC-seeded tissue engineering success is difficult in various tissues, when sites of injury are under the effects of a severe infection despite the immunomodulatory properties of MSCs. In the present review, the current understanding of the way in which WNT signaling regulates MSC activity modification under physiological and pathological conditions was summarized. An effort was also made to illustrate parts of the underlying mechanism, including the inflammatory factors and their interactions with the regulatory WNT signaling pathway, aiming to promote the clinical translation of MSC-based therapy.
Collapse
Affiliation(s)
- Qingtao Zhang
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Jian Yu
- Department of Stomatology, Zhejiang Hospital, Hangzhou, Zhejiang 310030, P.R. China
| | - Qiuqiu Chen
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Honghai Yan
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Hongjiang Du
- Department of Stomatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310085, P.R. China
| | - Wenjing Luo
- Department of General Dentistry, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA 02118, USA
| |
Collapse
|
237
|
In silico designing of vaccine candidate against Clostridium difficile. Sci Rep 2021; 11:14215. [PMID: 34244557 PMCID: PMC8271013 DOI: 10.1038/s41598-021-93305-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Clostridium difficile is a spore-forming gram-positive bacterium, recognized as the primary cause of antibiotic-associated nosocomial diarrhoea. Clostridium difficile infection (CDI) has emerged as a major health-associated infection with increased incidence and hospitalization over the years with high mortality rates. Contamination and infection occur after ingestion of vegetative spores, which germinate in the gastro-intestinal tract. The surface layer protein and flagellar proteins are responsible for the bacterial colonization while the spore coat protein, is associated with spore colonization. Both these factors are the main concern of the recurrence of CDI in hospitalized patients. In this study, the CotE, SlpA and FliC proteins are chosen to form a multivalent, multi-epitopic, chimeric vaccine candidate using the immunoinformatics approach. The overall reliability of the candidate vaccine was validated in silico and the molecular dynamics simulation verified the stability of the vaccine designed. Docking studies showed stable vaccine interactions with Toll‐Like Receptors of innate immune cells and MHC receptors. In silico codon optimization of the vaccine and its insertion in the cloning vector indicates a competent expression of the modelled vaccine in E. coli expression system. An in silico immune simulation system evaluated the effectiveness of the candidate vaccine to trigger a protective immune response.
Collapse
|
238
|
Xu X, Nie Y, Wang W, Ma N, Lendlein A. Periodic thermomechanical modulation of toll-like receptor expression and distribution in mesenchymal stromal cells. MRS COMMUNICATIONS 2021; 11:425-431. [PMID: 34258101 PMCID: PMC8265727 DOI: 10.1557/s43579-021-00049-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/25/2021] [Indexed: 06/13/2023]
Abstract
ABSTRACT Toll-like receptor (TLR) can trigger an immune response against virus including SARS-CoV-2. TLR expression/distribution is varying in mesenchymal stromal cells (MSCs) depending on their culture environments. Here, to explore the effect of periodic thermomechanical cues on TLRs, thermally controlled shape-memory polymer sheets with programmable actuation capacity were created. The proportion of MSCs expressing SARS-CoV-2-associated TLRs was increased upon stimulation. The TLR4/7 colocalization was promoted and retained in the endoplasmic reticula. The TLR redistribution was driven by myosin-mediated F-actin assembly. These results highlight the potential of boosting the immunity for combating COVID-19 via thermomechanical preconditioning of MSCs. GRAPHIC ABSTRACT Periodic thermal and synchronous mechanical stimuli provided by polymer sheet actuators selectively promoted the expression of SARS-CoV-2-associated TLRs 4 and 7 in adipose-derived MSCs and recruited TLR4 to Endoplasmic reticulum region where TLR7 was located via controlling myosin-mediated F-actin cytoskeleton assembly.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
| | - Yan Nie
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Weiwei Wang
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
| | - Nan Ma
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Hereon, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
239
|
de Oliveira Mann CC, Hornung V. Molecular mechanisms of nonself nucleic acid recognition by the innate immune system. Eur J Immunol 2021; 51:1897-1910. [PMID: 34138462 DOI: 10.1002/eji.202049116] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/13/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022]
Abstract
Nucleic acids (NAs) represent one of the most important classes of molecules recognized by the innate immune system. However, NAs are not limited to pathogens, but are also present within the host. As such, the immune system has evolved an elaborate set of pathogen recognition receptors (PRRs) that employ various strategies to recognize distinct types of NAs, while reliably distinguishing between self and nonself. The here-employed strategies encompass the positioning of NA-sensing PRRs in certain subcellular compartments that potentially come in contact with pathogens but not host NAs, the existence of counterregulatory measures that keep endogenous NAs below a certain threshold, and also the specific identification of certain nonself patterns. Here, we review recent advances in the molecular mechanisms of NA recognition by TLRs, RLRs, and the cGAS-STING axis. We highlight the differences in NA-PRR interfaces that confer specificity and selectivity toward an NA ligand, as well as the NA-dependent induced conformational changes required for signal transduction.
Collapse
Affiliation(s)
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
240
|
Xia P, Wu Y, Lian S, Yan L, Meng X, Duan Q, Zhu G. Research progress on Toll-like receptor signal transduction and its roles in antimicrobial immune responses. Appl Microbiol Biotechnol 2021; 105:5341-5355. [PMID: 34180006 PMCID: PMC8236385 DOI: 10.1007/s00253-021-11406-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
When microorganisms invade a host, the innate immune system first recognizes the pathogen-associated molecular patterns of these microorganisms through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are known transmembrane PRRs existing in both invertebrates and vertebrates. Upon ligand recognition, TLRs initiate a cascade of signaling events; promote the pro-inflammatory cytokine, type I interferon, and chemokine expression; and play an essential role in the modulation of the host's innate and adaptive immunity. Therefore, it is of great significance to improve our understanding of antimicrobial immune responses by studying the role of TLRs and their signal molecules in the host's defense against invading microbes. This paper aims to summarize the specificity of TLRs in recognition of conserved microbial components, such as lipoprotein, lipopolysaccharide, flagella, endosomal nucleic acids, and other bioactive metabolites derived from microbes. This set of interactions helps to elucidate the immunomodulatory effect of TLRs and the signal transduction changes involved in the infectious process and provide a novel therapeutic strategy to combat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Xia Meng
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Qiangde Duan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| |
Collapse
|
241
|
Ouyang W, Cen M, Yang L, Zhang W, Xia J, Xu F. NMI Facilitates Influenza A Virus Infection by Promoting Degradation of IRF7 through TRIM21. Am J Respir Cell Mol Biol 2021; 65:30-40. [PMID: 33761305 DOI: 10.1165/rcmb.2020-0391oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Acute respiratory infections caused by influenza A virus (IAV) spread widely and lead to substantial morbidity and mortality. Host cell induction of type I interferon (IFN-I) plays a fundamental role in eliminating the virus during the innate antiviral response. The potential role of N-myc and STAT interactor (NMI) and its underlying mechanisms of action during IAV infection, however, remain elusive. In this study, we found that the expression of NMI increased after IAV infection. Nmi-knockout mice infected with IAV displayed increased survival rate, decreased weight loss, lower viral replication, and attenuated lung inflammation when compared with wild-type mice. Deficiency of NMI promoted the production of IFN-I and IFN-stimulated genes in vivo and in vitro. Reduced levels of NMI also resulted in an increase of the expression of IFN regulator factor (IRF) 7. Further studies have revealed that NMI could interact with IRF7 after IAV infection, and this interaction involved its NID1 and NID2 domain. In addition, NMI facilitated ubiquitination and proteasome-dependent degradation of IRF7 through recruitment of the E3 ubiquitin ligase TRIM21 (tripartite motif-containing 21) to limit the IAV-triggered innate immunity. Our findings reveal a clearer understanding of the role of NMI in regulating the host innate antiviral response and provide a potential therapeutic target for controlling IAV infection.
Collapse
Affiliation(s)
| | | | | | | | - Jingyan Xia
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Xu
- Department of Infectious Diseases and
| |
Collapse
|
242
|
Steinmaurer A, Wimmer I, Berger T, Rommer PS, Sellner J. Bruton's tyrosine kinase inhibition in the treatment of preclinical models and multiple sclerosis. Curr Pharm Des 2021; 28:437-444. [PMID: 34218776 DOI: 10.2174/1381612827666210701152934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/10/2021] [Indexed: 11/22/2022]
Abstract
Significant progress has been made in understanding the immunopathogenesis of multiple sclerosis (MS) over recent years. Successful clinical trials with CD20-depleting monoclonal antibodies have corroborated the fundamental role of B cells in the pathogenesis of MS and reinforced the notion that cells of the B cell lineage are an attractive treatment target. Therapeutic inhibition of Bruton's tyrosine kinase (BTK), an enzyme involved in B cell and myeloid cell activation and function, is regarded as a next-generation approach that aims to attenuate both errant innate and adaptive immune functions. Moreover, brain-penetrant BTK inhibitors may impact compartmentalized inflammation and neurodegeneration within the central nervous system by targeting brain-resident B cells and microglia, respectively. Preclinical studies in animal models of MS corroborated an impact of BTK inhibition on meningeal inflammation and cortical demyelination. Notably, BTK inhibition attenuated the antigen-presenting capacity of B cells and the generation of encephalitogenic T cells. Evobrutinib, a selective oral BTK inhibitor, has been tested recently in a phase 2 study of patients with relapsing-remitting MS. The study met the primary endpoint of a significantly reduced cumulative number of Gadolinium-enhancing lesions under treatment with evobrutinib compared to placebo treatment. Thus, the results of ongoing phase 2 and 3 studies with evobrutinib, fenobrutinib, and tolebrutinib in relapsing-remitting and progressive MS are eagerly awaited. This review article introduces the physiological role of BTK, summarizes the pre-clinical and trial evidence, and addresses the potential beneficial effects of BTK inhibition in MS.
Collapse
Affiliation(s)
- Anja Steinmaurer
- Department of Neurology, Medical University of Vienna, Vienna. Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna. Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna. Austria
| | | | - Johann Sellner
- Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, Mistelbach. Austria
| |
Collapse
|
243
|
Conte C. Possible Link between SARS-CoV-2 Infection and Parkinson's Disease: The Role of Toll-Like Receptor 4. Int J Mol Sci 2021; 22:7135. [PMID: 34281186 PMCID: PMC8269350 DOI: 10.3390/ijms22137135] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disorder characterized by selective degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) of the midbrain, depletion of dopamine (DA), and impaired nigrostriatal pathway. The pathological hallmark of PD includes the aggregation and accumulation α-synuclein (α-SYN). Although the precise mechanisms underlying the pathogenesis of PD are still unknown, the activation of toll-like receptors (TLRs), mainly TLR4 and subsequent neuroinflammatory immune response, seem to play a significant role. Mounting evidence suggests that viral infection can concur with the precipitation of PD or parkinsonism. The recently identified coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of ongoing pandemic coronavirus disease 2019 (COVID-19), responsible for 160 million cases that led to the death of more than three million individuals worldwide. Studies have reported that many patients with COVID-19 display several neurological manifestations, including acute cerebrovascular diseases, conscious disturbance, and typical motor and non-motor symptoms accompanying PD. In this review, the neurotropic potential of SARS-CoV-2 and its possible involvement in the pathogenesis of PD are discussed. Specifically, the involvement of the TLR4 signaling pathway in mediating the virus entry, as well as the massive immune and inflammatory response in COVID-19 patients is explored. The binding of SARS-CoV-2 spike (S) protein to TLR4 and the possible interaction between SARS-CoV-2 and α-SYN as contributing factors to neuronal death are also considered.
Collapse
Affiliation(s)
- Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, via Fabretti, 06123 Perugia, Italy
| |
Collapse
|
244
|
Olech M, Ropka-Molik K, Szmatoła T, Piórkowska K, Kuźmak J. Single Nucleotide Polymorphisms in Genes Encoding Toll-Like Receptors 7 and 8 and Their Association with Proviral Load of SRLVs in Goats of Polish Carpathian Breed. Animals (Basel) 2021; 11:ani11071908. [PMID: 34206971 PMCID: PMC8300119 DOI: 10.3390/ani11071908] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022] Open
Abstract
Toll-like receptors (TLRs) 7 and 8 are important in single-stranded viral RNA recognition, so genetic variation of these genes may play a role in SRLVs infection and disease progression. Present study aimed to identify SNPs in genes encoding TLR7 and TLR8 in goats of Carpathian breed and analyze their association with the SRLVs provirus concentration as index of disease progression. A total of 14 SNPs were detected, 6 SNPs in the TLR7 gene locus and 8 SNPs in the TLR8 gene. Nine of the 14 identified polymorphisms, 4 in the TLR7 gene and 5 in TLR8 gene, were significantly associated with the SRLVs proviral concentration. These SNPs were located in 3'UTR, 5'UTR and intron sequences as well as in the coding sequences, but they led to silent changes. Homozygous genotypes of three TLR7 SNPs (synonymous variant 1:50703293, 3'UTR variant 1:50701297 and 5'UTR variant 1:50718645) were observed in goats with lower provirus copy number as well as in seronegative animals. The results obtained in this study suggest that SNPs of TLR7/TLR8 genes may induce differential innate immune response towards SRLVs affecting proviral concentration and thereby disease pathogenesis and progression. These findings support a role for genetic variations of TLR7 and TLR8 in SRLVs infection and warrants further studies on the effect of TLR7/TLR8 polymorphisms on SRLVs infection in different populations.
Collapse
Affiliation(s)
- Monika Olech
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland;
- Correspondence: ; Tel.: +48-8188-9300; Fax: +48-818-862-595
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland; (K.R.-M.); (T.S.); (K.P.)
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland; (K.R.-M.); (T.S.); (K.P.)
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Rędzina 1c, 30-248 Krakow, Poland
| | - Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1, 32-083 Balice, Poland; (K.R.-M.); (T.S.); (K.P.)
| | - Jacek Kuźmak
- Department of Biochemistry, National Veterinary Research Institute, 24-100 Pulawy, Poland;
| |
Collapse
|
245
|
Chen CY, Hung YF, Tsai CY, Shih YC, Chou TF, Lai MZ, Wang TF, Hsueh YP. Transcriptomic Analysis and C-Terminal Epitope Tagging Reveal Differential Processing and Signaling of Endogenous TLR3 and TLR7. Front Immunol 2021; 12:686060. [PMID: 34211474 PMCID: PMC8240634 DOI: 10.3389/fimmu.2021.686060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023] Open
Abstract
Toll-like receptor (TLR) signaling is critical for defense against pathogenic infection, as well as for modulating tissue development. Activation of different TLRs triggers common inflammatory responses such as cytokine induction. Here, we reveal differential impacts of TLR3 and TLR7 signaling on transcriptomic profiles in bone marrow-derived macrophages (BMDMs). Apart from self-regulation, TLR3, but not TLR7, induced expression of other TLRs, suggesting that TLR3 activation globally enhances innate immunity. Moreover, we observed diverse influences of TLR3 and TLR7 signaling on genes involved in methylation, caspase and autophagy pathways. We compared endogenous TLR3 and TLR7 by using CRISPR/Cas9 technology to knock in a dual Myc-HA tag at the 3’ ends of mouse Tlr3 and Tlr7. Using anti-HA antibodies to detect endogenous tagged TLR3 and TLR7, we found that both TLRs display differential tissue expression and posttranslational modifications. C-terminal tagging did not impair TLR3 activity. However, it disrupted the interaction between TLR7 and myeloid differentiation primary response 88 (MYD88), the Tir domain-containing adaptor of TLR7, which blocked its downstream signaling necessary to trigger cytokine and chemokine expression. Our study demonstrates different properties for TLR3 and TLR7, and also provides useful mouse models for further investigation of these two RNA-sensing TLRs.
Collapse
Affiliation(s)
- Chiung-Ya Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yun-Fen Hung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ching-Yen Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Shih
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ting-Fang Chou
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ting-Fang Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
246
|
Abstract
Type I interferons (IFN-Is) are a very important group of cytokines that are produced by innate immune cells but also act on adaptive immune cells. IFN-Is possess antiviral, antitumor, and anti-proliferative effects, as well are associated with the initiation and maintenance of autoimmune disorders. Studies have shown that aberrantly expressed IFN-Is and/or type I IFN-inducible gene signatures in the serum or tissues of patients with autoimmune disorders are linked to their pathogenesis, clinical manifestations, and disease activity. Type I interferonopathies with mutations in genes impacting the type I IFN signaling pathway have shown symptoms and characteristics similar to those of systemic lupus erythematosus (SLE). Furthermore, both interventions in animal models and clinical trials of therapies targeting the type I IFN signaling pathway have shown efficacy in the treatment of autoimmune diseases. Our review aims to summarize the functions and targeted therapies (as well as clinical trials) of IFN-Is in both adult and pediatric autoimmune diseases, such as SLE, pediatric SLE (pSLE), rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), juvenile dermatomyositis (JDM), Sjögren syndrome (SjS), and systemic sclerosis (SSc), discussing the potential abnormal regulation of transcription factors and epigenetic modifications and providing a potential mechanism for pathogenesis and therapeutic strategies for future clinical use.
Collapse
|
247
|
Tavakol S, Alavijeh MS, Seifalian AM. COVID-19 Vaccines in Clinical Trials and their Mode of Action for Immunity against the Virus. Curr Pharm Des 2021; 27:1553-1563. [PMID: 33100195 DOI: 10.2174/1381612826666201023143956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/10/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
For nearly two decades, coronaviruses have caused many health and economic problems, while no effective commercial vaccine has yet been developed. It is worth mentioning that despite some mutations and recombination in SARS-CoV-2, its genotype is very close to the original strain from Wuhan, China. Therefore, the development of an effective vaccine would be promising. It might be hypothesized that BCG vaccination is performed in high-risk populations before the commercialization of an effective SARS-CoV-2 vaccine. However, the development of an effective vaccine without considering the adverse immune reactions derived from antibody-dependent or cell-based immune enhancement may threaten vaccinated people's lives and long-term side effects must be considered. To this end, targeting of the receptor-binding domain (RBD) in spike and not whole spike, glycolization of FC receptors, PD-1 blockers, CPPs, etc., are promising. Therefore, the subunit vaccines or RNA vaccines that encode the RBP segment of the spike are of interest. To enhance the vaccine efficacy, its co-delivery with an adjuvant has been recommended. Nanoparticles modulate immune response with higher efficiency than the soluble form of antigens and can be functionalized with the positively charged moieties and ligands of targeted cells, such as dendritic cells, to increase cellular uptake of the antigens and their presentation on the surface of immune cells. This research aimed to discuss the COVID-19 vaccines entering the clinical trial and their mode of action effective immunity against the virus and discusses their advantages compared to each other.
Collapse
Affiliation(s)
- Shima Tavakol
- Pharmidex Pharmaceutical Services Ltd., London, United Kingdom
| | - Mo S Alavijeh
- Pharmidex Pharmaceutical Services Ltd., London, United Kingdom
| | - Alexander M Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
248
|
Sánchez-Luquez K, Schadock IC, Gonçalves CV, Tornatore M, Finger-Jardim F, Avila EC, Soares MA, de Martínez AMB, Ellwanger JH, Chies JAB, da Hora VP. Impact of TLR7 and TLR9 polymorphisms on susceptibility to placental infections and pregnancy complications. J Reprod Immunol 2021; 146:103342. [PMID: 34102513 DOI: 10.1016/j.jri.2021.103342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
This study evaluated the impact of the TLR7 Gln11Leu (rs179008) and TLR9 -1237 T/C (rs5743836) single nucleotide polymorphisms (SNPs) on susceptibility to placental infections and pregnancy complications in 455 Brazilian women. Demographic, socioeconomic, gynecological, and clinical characteristics of the women were collected. Placental tissues were sampled from pregnant women and human and viral DNA was extracted. Human alphaherpesvirus 1 (Herpes simplex virus type 1, HSV-1), Human alphaherpesvirus 2 (Herpes simplex virus type 2, HSV-2) and Human betaherpesvirus 5 (Human cytomegalovirus, HCMV) were detected by nested PCR. TLR9 and TLR7 SNPs were genotyped by PCR amplification of bi-directional specific alleles (Bi-PASA) and restriction fragment length polymorphism (RFLP), respectively. Infections at the time of birth were detected in 45.71 % of women. The presence of the TT genotype (recessive model) of the TLR7 SNP was associated with increased susceptibility to HSV-1 infection (O.R. = 2.23, p = 0.05). The presence of the C allele of the TLR9 SNP, in heterozygosis or homozygosis (dominant model), decreased the infection risk by HCMV (O.R. = 0.31, p-mod<0.05). The TT genotype (recessive model) of the TLR7 SNP was significantly associated (p < 0.05) with increased occurrence of pre-treated hypertension. The codominant model of the TLR9 SNP was significantly associated (p < 0.05) with reduced risk of hospitalization during pregnancy. In combination, the AA/CT (TLR7-TLR9) genotypes significantly decreased the risk of placental infection by HSV-1 and/or HSV-2 (O.R. = 0.47, p = 0.02), the susceptibility to all infectious agents considered in combination (O.R. = 0.4, p = 0.00), and the need of hospitalization (O.R. = 0.48, p = 0.02). In conclusion, TLR7 and TLR9 SNPs are potential modulating factors for the risk of placental infections and pregnancy complications.
Collapse
Affiliation(s)
- Karen Sánchez-Luquez
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil.
| | - Ines Claudia Schadock
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Carla Vitola Gonçalves
- Center for Obstetrics and Gynecology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Michele Tornatore
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Fabiana Finger-Jardim
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Emiliana Claro Avila
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Marcelo Alves Soares
- Oncovirology Program, National Cancer Institute (INCA), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Barral de Martínez
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Vanusa Pousada da Hora
- Laboratory of Molecular Biology, School of Medicine, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| |
Collapse
|
249
|
Ghafouri-Fard S, Abak A, Shoorei H, Talebi SF, Mohaqiq M, Sarabi P, Taheri M, Mokhtari M. Interaction between non-coding RNAs and Toll-like receptors. Biomed Pharmacother 2021; 140:111784. [PMID: 34087695 DOI: 10.1016/j.biopha.2021.111784] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptors (TLRs) are a large group of pattern recognition receptors which are involved in the regulation of innate immune responses. Based on the interplay between TLRs and adapter molecules, two distinctive signaling cascades, namely the MyD88-dependent and TRIF-dependent pathways have been recognized. TLRs are involved in the development of a wide variety of diseases including cancer and autoimmune disorders. A large body of evidence has shown interaction between two classes of non-coding RNAs, namely microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). These interactions have prominent roles in the pathogenesis of several disorders including infectious disorders, autoimmune conditions and neoplastic disorders. This review aims at description of the interaction between these non-coding RNAs and TLRs.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Mahdi Mohaqiq
- School of Advancement, Centennial College, Ashtonbee Campus, Toronto, ON, Canada
| | - Parisa Sarabi
- Deputy for Research & Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Mokhtari
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
250
|
Cerqueira JXM, Saavalainen P, Kurppa K, Laurikka P, Huhtala H, Nykter M, L E Koskinen L, Yohannes DA, Kilpeläinen E, Shcherban A, Palotie A, Kaukinen K, Lindfors K. Independent and cumulative coeliac disease-susceptibility loci are associated with distinct disease phenotypes. J Hum Genet 2021; 66:613-623. [PMID: 33446885 PMCID: PMC8144013 DOI: 10.1038/s10038-020-00888-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
The phenotype of coeliac disease varies considerably for incompletely understood reasons. We investigated whether established coeliac disease susceptibility variants (SNPs) are individually or cumulatively associated with distinct phenotypes. We also tested whether a polygenic risk score (PRS) based on genome-wide associated (GWA) data could explain the phenotypic variation. The phenotypic association of 39 non-HLA coeliac disease SNPs was tested in 625 thoroughly phenotyped coeliac disease patients and 1817 controls. To assess their cumulative effects a weighted genetic risk score (wGRS39) was built, and stratified by tertiles. In our PRS model in cases, we took the summary statistics from the largest GWA study in coeliac disease and tested their association at eight P value thresholds (PT) with phenotypes. Altogether ten SNPs were associated with distinct phenotypes after correction for multiple testing (PEMP2 ≤ 0.05). The TLR7/TLR8 locus was associated with disease onset before and the SH2B3/ATXN2, ITGA4/UBE2E3 and IL2/IL21 loci after 7 years of age. The latter three loci were associated with a more severe small bowel mucosal damage and SH2B3/ATXN2 with type 1 diabetes. Patients at the highest wGRS39 tertiles had OR > 1.62 for having coeliac disease-related symptoms during childhood, a more severe small bowel mucosal damage, malabsorption and anaemia. PRS was associated only with dermatitis herpetiformis (PT = 0.2, PEMP2 = 0.02). Independent coeliac disease-susceptibility loci are associated with distinct phenotypes, suggesting that genetic factors play a role in determining the disease presentation. Moreover, the increased number of coeliac disease susceptibility SNPs might predispose to a more severe disease course.
Collapse
Affiliation(s)
- Juliana X M Cerqueira
- Coeliac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Päivi Saavalainen
- Research Programs Unit, Immunobiology, and the Haartman Institute, Department of Molecular Genetics, University of Helsinki, Helsinki, Finland
| | - Kalle Kurppa
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, and the University Consortium of Seinäjoki, Seinäjoki, Finland
| | - Pilvi Laurikka
- Coeliac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Heini Huhtala
- Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Matti Nykter
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lotta L E Koskinen
- Research Programs Unit, Immunobiology, and the Haartman Institute, Department of Molecular Genetics, University of Helsinki, Helsinki, Finland
| | - Dawit A Yohannes
- Research Programs Unit, Immunobiology, and the Haartman Institute, Department of Molecular Genetics, University of Helsinki, Helsinki, Finland
| | - Elina Kilpeläinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Anastasia Shcherban
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Analytic and Translational Genetics Unit, Department of Medicine, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Katri Kaukinen
- Coeliac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Katri Lindfors
- Coeliac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|