1
|
Mahin A, Chikmagalur Ravindra S, Ramesh P, Naik P, Raju R, Keshava Prasad TS, Abhinand CS. Unveiling Actin Cytoskeleton Role in Mediating Chikungunya-Associated Arthritis: An Integrative Proteome-Metabolome Study. Vector Borne Zoonotic Dis 2024; 24:753-762. [PMID: 38717066 DOI: 10.1089/vbz.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Background: Chikungunya is a zoonotic disease caused by the Chikungunya virus (CHIKV), primarily transmitted to humans through infected Aedes mosquitoes. The infection is characterized by symptoms such as high fever, musculoskeletal pain, polyarthritis, and a rash, which can lead to severe complications such as encephalitis, meningitis, and even fatalities. While many disease manifestations resemble those of other viral infections, chronic arthritis caused by CHIKV is unique, and its molecular mechanisms remain ill-defined. Materials and Methods: Proteomics data from both cellular and patient levels of CHIKV infection were curated from PubMed and screened using inclusion and exclusion criteria. Patient serum proteomics data obtained from PRIDE underwent reanalysis using Proteome Discoverer 2.2. Enrichment and protein-protein interaction network analysis were conducted on differentially expressed proteins from both serum and cellular datasets. Metabolite data from CHIKV-infected patients were further retrieved, and their protein binding partners were identified using BindingDB. The protein-metabolite interaction pathway was further developed using MetaboAnalyst. Results: The proteomics data analysis revealed differential expression of proteins involved in critical host mechanisms, such as cholesterol metabolism and mRNA splicing, during CHIKV infection. Consistent upregulation of two actin cytoskeleton proteins, TAGLN2 and PFN1, was noted in both serum and cellular datasets, and their upregulations are associated with arthritis. Furthermore, alterations in purine metabolism were observed in the integrative proteome-metabolome analysis, correlating with cytoskeletal remodelling. Conclusion: Collectively, this integrative view sheds light on the involvement of actin cytoskeleton remodeling proteins and purine metabolic pathways in the development of arthritis during CHIKV infection.
Collapse
Affiliation(s)
- Althaf Mahin
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| | - Sourav Chikmagalur Ravindra
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
- Department of Biosciences, Mangalore University, Mangalore, India
| | - Poornima Ramesh
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| | - Prashantha Naik
- Department of Biosciences, Mangalore University, Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to Be University), Mangalore, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to Be University), Mangalore, India
| |
Collapse
|
2
|
Varikkodan MM, Kunnathodi F, Azmi S, Wu TY. An Overview of Indian Biomedical Research on the Chikungunya Virus with Particular Reference to Its Vaccine, an Unmet Medical Need. Vaccines (Basel) 2023; 11:1102. [PMID: 37376491 DOI: 10.3390/vaccines11061102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chikungunya virus (CHIKV) is an infectious agent spread by mosquitos, that has engendered endemic or epidemic outbreaks of Chikungunya fever (CHIKF) in Africa, South-East Asia, America, and a few European countries. Like most tropical infections, CHIKV is frequently misdiagnosed, underreported, and underestimated; it primarily affects areas with limited resources, like developing nations. Due to its high transmission rate and lack of a preventive vaccine or effective treatments, this virus poses a serious threat to humanity. After a 32-year hiatus, CHIKV reemerged as the most significant epidemic ever reported, in India in 2006. Since then, CHIKV-related research was begun in India, and up to now, more than 800 peer-reviewed research papers have been published by Indian researchers and medical practitioners. This review gives an overview of the outbreak history and CHIKV-related research in India, to favor novel high-quality research works intending to promote effective treatment and preventive strategies, including vaccine development, against CHIKV infection.
Collapse
Affiliation(s)
- Muhammed Muhsin Varikkodan
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| | - Faisal Kunnathodi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Sarfuddin Azmi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh 11159, Saudi Arabia
| | - Tzong-Yuan Wu
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- R&D Center of Membrane Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| |
Collapse
|
3
|
Resende F, de Araújo S, Tavares LP, Teixeira MM, Costa VV. The Multifaceted Role of Annexin A1 in Viral Infections. Cells 2023; 12:1131. [PMID: 37190040 PMCID: PMC10137178 DOI: 10.3390/cells12081131] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Dysregulated inflammatory responses are often correlated with disease severity during viral infections. Annexin A1 (AnxA1) is an endogenous pro-resolving protein that timely regulates inflammation by activating signaling pathways that culminate with the termination of response, clearance of pathogen and restoration of tissue homeostasis. Harnessing the pro-resolution actions of AnxA1 holds promise as a therapeutic strategy to control the severity of the clinical presentation of viral infections. In contrast, AnxA1 signaling might also be hijacked by viruses to promote pathogen survival and replication. Therefore, the role of AnxA1 during viral infections is complex and dynamic. In this review, we provide an in-depth view of the role of AnxA1 during viral infections, from pre-clinical to clinical studies. In addition, this review discusses the therapeutic potential for AnxA1 and AnxA1 mimetics in treating viral infections.
Collapse
Affiliation(s)
- Filipe Resende
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Simone de Araújo
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Mauro Martins Teixeira
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
4
|
Lv C, Zhang Q, Zhao L, Yang J, Zou Z, Zhao Y, Li C, Sun X, Lin X, Jin M. African swine fever virus infection activates inflammatory responses through downregulation of the anti-inflammatory molecule C1QTNF3. Front Immunol 2022; 13:1002616. [PMID: 36311798 PMCID: PMC9598424 DOI: 10.3389/fimmu.2022.1002616] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
African swine fever (ASF) is the most dangerous pig disease, and causes enormous economic losses in the global pig industry. However, the mechanisms of ASF virus (ASFV) infection remains largely unclear. Hence, this study investigated the host response mechanisms to ASFV infection. We analyzed the differentially expressed proteins (DEPs) between serum samples from ASFV-infected and uninfected pigs using quantitative proteomics. Setting the p-value < 0.05 and |log2 (fold change)| > 1.5, we identified 173 DEPs, comprising 57 upregulated and 116 downregulated proteins, which belonged to various biological processes and pathways based on the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. The enriched pathways include immune responses, metabolism, and inflammation signaling pathways. Western blot analysis validated the DEPs identified using quantitative proteomics. Furthermore, our proteomics data showed that C1QTNF3 regulated the inflammatory signaling pathway. C1QTNF3 knockdown led to the upregulation of pro-inflammatory factors IL-1β, IL-8, and IL-6, thus inhibiting ASFV replication. These results indicated that C1QTNF3 was critical for ASFV infection. In conclusion, this study revealed the molecular mechanisms underlying the host-ASFV interaction, which may contribute to the development of novel antiviral strategies against ASFV infection in the future.
Collapse
Affiliation(s)
- Changjie Lv
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- New-onset department, Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
- Department of pig disease prevention and control, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Qiang Zhang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Li Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jingyu Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Zhong Zou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- New-onset department, Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Ya Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of pig disease prevention and control, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Chengfei Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of pig disease prevention and control, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xiaomei Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of pig disease prevention and control, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xian Lin
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Xian Lin, ; Meilin Jin,
| | - Meilin Jin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- New-onset department, Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
- Department of pig disease prevention and control, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- *Correspondence: Xian Lin, ; Meilin Jin,
| |
Collapse
|
5
|
de Araújo S, de Melo Costa VR, Santos FM, de Sousa CDF, Moreira TP, Gonçalves MR, Félix FB, Queiroz-Junior CM, Campolina-Silva GH, Nogueira ML, Sugimoto MA, Bonilha CS, Perretti M, Souza DG, Costa VV, Teixeira MM. Annexin A1-FPR2/ALX Signaling Axis Regulates Acute Inflammation during Chikungunya Virus Infection. Cells 2022; 11:cells11172717. [PMID: 36078125 PMCID: PMC9454528 DOI: 10.3390/cells11172717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
Chikungunya (CHIKV) is an arthritogenic alphavirus that causes a self-limiting disease usually accompanied by joint pain and/or polyarthralgia with disabling characteristics. Immune responses developed during the acute phase of CHIKV infection determine the rate of disease progression and resolution. Annexin A1 (AnxA1) is involved in both initiating inflammation and preventing over-response, being essential for a balanced end of inflammation. In this study, we investigated the role of the AnxA1-FPR2/ALX pathway during CHIKV infection. Genetic deletion of AnxA1 or its receptor enhanced inflammatory responses driven by CHIKV. These knockout mice showed increased neutrophil accumulation and augmented tissue damage at the site of infection compared with control mice. Conversely, treatment of wild-type animals with the AnxA1 mimetic peptide (Ac2–26) reduced neutrophil accumulation, decreased local concentration of inflammatory mediators and diminished mechanical hypernociception and paw edema induced by CHIKV-infection. Alterations in viral load were mild both in genetic deletion or with treatment. Combined, our data suggest that the AnxA1-FPR2/ALX pathway is a potential therapeutic strategy to control CHIKV-induced acute inflammation and polyarthralgia.
Collapse
Affiliation(s)
- Simone de Araújo
- Graduate Program in Biological Sciences Physiology and Pharmacology, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Victor R. de Melo Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciele M. Santos
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Carla D. Ferreira de Sousa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Thaiane P. Moreira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Matheus R. Gonçalves
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciel B. Félix
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Celso M. Queiroz-Junior
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Gabriel H. Campolina-Silva
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Obstetrics, Gynecology and Reproduction, CHU de Quebec Research Center (CHUL), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Maurício Lacerda Nogueira
- Department of Dermatological, Infections, and Parasitic Diseases, School of Medicine (FAMERP), São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Michelle A. Sugimoto
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Caio S. Bonilha
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research on Inflammatory Diseases, University of São Paulo, São Paulo 05508-000, Brazil
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mauro Perretti
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London E1 4NS, UK
| | - Danielle G. Souza
- Graduate Program in Microbiology, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian V. Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Graduate Program in Cell Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| | - Mauro M. Teixeira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| |
Collapse
|
6
|
Tavares LP, Melo EM, Sousa LP, Teixeira MM. Pro-resolving therapies as potential adjunct treatment for infectious diseases: Evidence from studies with annexin A1 and angiotensin-(1-7). Semin Immunol 2022; 59:101601. [PMID: 35219595 DOI: 10.1016/j.smim.2022.101601] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/06/2022] [Accepted: 02/17/2022] [Indexed: 01/15/2023]
Abstract
Infectious diseases, once believed to be an eradicable public health threat, still represent a leading cause of death worldwide. Environmental and social changes continuously favor the emergence of new pathogens and rapid dissemination around the world. The limited availability of anti-viral therapies and increased antibiotic resistance has made the therapeutic management of infectious disease a major challenge. Inflammation is a primordial defense to protect the host against invading microorganisms. However, dysfunctional inflammatory responses contribute to disease severity and mortality during infections. In recent years, a few studies have examined the relevance of resolution of inflammation in the context of infections. Inflammation resolution is an active integrated process transduced by several pro-resolving mediators, including Annexin A1 and Angiotensin-(1-7). Here, we examine some of the cellular and molecular circuits triggered by pro-resolving molecules and that may be beneficial in the context of infectious diseases.
Collapse
Affiliation(s)
- Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Eliza Mathias Melo
- Immunopharmacology Laboratory, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lirlândia Pires Sousa
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Mauro Martins Teixeira
- Immunopharmacology Laboratory, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
7
|
Kochan K, Bedolla DE, Perez-Guaita D, Adegoke JA, Chakkumpulakkal Puthan Veettil T, Martin M, Roy S, Pebotuwa S, Heraud P, Wood BR. Infrared Spectroscopy of Blood. APPLIED SPECTROSCOPY 2021; 75:611-646. [PMID: 33331179 DOI: 10.1177/0003702820985856] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The magnitude of infectious diseases in the twenty-first century created an urgent need for point-of-care diagnostics. Critical shortages in reagents and testing kits have had a large impact on the ability to test patients with a suspected parasitic, bacteria, fungal, and viral infections. New point-of-care tests need to be highly sensitive, specific, and easy to use and provide results in rapid time. Infrared spectroscopy, coupled to multivariate and machine learning algorithms, has the potential to meet this unmet demand requiring minimal sample preparation to detect both pathogenic infectious agents and chronic disease markers in blood. This focal point article will highlight the application of Fourier transform infrared spectroscopy to detect disease markers in blood focusing principally on parasites, bacteria, viruses, cancer markers, and important analytes indicative of disease. Methodologies and state-of-the-art approaches will be reported and potential confounding variables in blood analysis identified. The article provides an up to date review of the literature on blood diagnosis using infrared spectroscopy highlighting the recent advances in this burgeoning field.
Collapse
Affiliation(s)
- Kamila Kochan
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - Diana E Bedolla
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - David Perez-Guaita
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - John A Adegoke
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | | | - Miguela Martin
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - Supti Roy
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - Savithri Pebotuwa
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - Philip Heraud
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| | - Bayden R Wood
- 2541Monash University - Centre for Biospectroscopy, Clayton, Victoria, Australia
| |
Collapse
|
8
|
Aggarwal S, Acharjee A, Mukherjee A, Baker MS, Srivastava S. Role of Multiomics Data to Understand Host-Pathogen Interactions in COVID-19 Pathogenesis. J Proteome Res 2021; 20:1107-1132. [PMID: 33426872 PMCID: PMC7805606 DOI: 10.1021/acs.jproteome.0c00771] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Indexed: 12/15/2022]
Abstract
Human infectious diseases are contributed equally by the host immune system's efficiency and any pathogens' infectivity. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the coronavirus strain causing the respiratory pandemic coronavirus disease 2019 (COVID-19). To understand the pathobiology of SARS-CoV-2, one needs to unravel the intricacies of host immune response to the virus, the viral pathogen's mode of transmission, and alterations in specific biological pathways in the host allowing viral survival. This review critically analyzes recent research using high-throughput "omics" technologies (including proteomics and metabolomics) on various biospecimens that allow an increased understanding of the pathobiology of SARS-CoV-2 in humans. The altered biomolecule profile facilitates an understanding of altered biological pathways. Further, we have performed a meta-analysis of significantly altered biomolecular profiles in COVID-19 patients using bioinformatics tools. Our analysis deciphered alterations in the immune response, fatty acid, and amino acid metabolism and other pathways that cumulatively result in COVID-19 disease, including symptoms such as hyperglycemic and hypoxic sequelae.
Collapse
Affiliation(s)
- Shalini Aggarwal
- Department of Biosciences and
Bioengineering, Indian Institute of Technology
Bombay, Mumbai 400076,
India
| | - Arup Acharjee
- Department of Biosciences and
Bioengineering, Indian Institute of Technology
Bombay, Mumbai 400076,
India
| | - Amrita Mukherjee
- Department of Biosciences and
Bioengineering, Indian Institute of Technology
Bombay, Mumbai 400076,
India
| | - Mark S. Baker
- Department of Biomedical Science,
Faculty of Medicine, Health and Human Sciences, Macquarie
University, Sydney 2109,
Australia
| | - Sanjeeva Srivastava
- Department of Biosciences and
Bioengineering, Indian Institute of Technology
Bombay, Mumbai 400076,
India
| |
Collapse
|
9
|
Vago JP, Tavares LP, Riccardi C, Teixeira MM, Sousa LP. Exploiting the pro-resolving actions of glucocorticoid-induced proteins Annexin A1 and GILZ in infectious diseases. Biomed Pharmacother 2020; 133:111033. [PMID: 33378946 DOI: 10.1016/j.biopha.2020.111033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 02/08/2023] Open
Abstract
For decades, glucocorticoids (GC) have been used to treat several inflammatory conditions, including chronic and autoimmune diseases, due to their potent anti-inflammatory properties. In the context of infectious diseases, the use of GCs may be effective as adjuvant to antibiotic therapy by controlling excessive inflammatory responses resulting in better outcome in some cases. However, the use of GCs has been associated with a vast number of side effects, including increased probability of immunosuppression and consequent risk of opportunistic infection. Glucocorticoid-induced leucine zipper (GILZ) and Annexin A1 (AnxA1) are GC-induced proteins intrinsically involved with the anti-inflammatory functions of GCs without the associated adverse metabolic effects. Recent studies have shown that these GC-proteins exhibit pro-resolving effects. An essential characteristic of pro-resolving molecules is their ability to coordinate the resolution of inflammation and promote host defense in most experimental models of infection. Although the role of GILZ and AnxA1 in the context of infectious diseases remain to be better explored, herein we provide an overview of the emerging functions of these GC-proteins obtained from pre-clinical models of infectious diseases.
Collapse
Affiliation(s)
- Juliana P Vago
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Luciana P Tavares
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carlo Riccardi
- Departament of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
10
|
Mahmud I, Garrett TJ. Mass Spectrometry Techniques in Emerging Pathogens Studies: COVID-19 Perspectives. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2013-2024. [PMID: 32880453 PMCID: PMC7496948 DOI: 10.1021/jasms.0c00238] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 05/04/2023]
Abstract
As corona virus disease 2019 (COVID-19) is a rapidly growing public health crisis across the world, our knowledge of meaningful diagnostic tests and treatment for severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) is still evolving. This novel coronavirus disease COVID-19 can be diagnosed using RT-PCR, but inadequate access to reagents, equipment, and a nonspecific target has slowed disease detection and management. Precision medicine, individualized patient care, requires suitable diagnostics approaches to tackle the challenging aspects of viral outbreaks where many tests are needed in a rapid and deployable approach. Mass spectrometry (MS)-based technologies such as proteomics, glycomics, lipidomics, and metabolomics have been applied in disease outbreaks for identification of infectious disease agents such as virus and bacteria and the molecular phenomena associated with pathogenesis. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF/MS) is widely used in clinical diagnostics in the United States and Europe for bacterial pathogen identification. Paper spray ionization mass spectrometry (PSI-MS), a rapid ambient MS technique, has recently open a new opportunity for future clinical investigation to diagnose pathogens. Ultra-high-pressure liquid chromatography coupled high-resolution mass spectrometry (UHPLC-HRMS)-based metabolomics and lipidomics have been employed in large-scale biomedical research to discriminate infectious pathogens and uncover biomarkers associated with pathogenesis. PCR-MS has emerged as a new technology with the capability to directly identify known pathogens from the clinical specimens and the potential to identify genetic evidence of undiscovered pathogens. Moreover, miniaturized MS offers possible applications with relatively fast, highly sensitive, and potentially portable ways to analyze for viral compounds. However, beneficial aspects of these rapidly growing MS technologies in pandemics like COVID-19 outbreaks has been limited. Hence, this perspective gives a brief of the existing knowledge, current challenges, and opportunities for MS-based techniques as a promising avenue in studying emerging pathogen outbreaks such as COVID-19.
Collapse
Affiliation(s)
- Iqbal Mahmud
- Department of Pathology, Immunology,
and Laboratory Medicine, University of
Florida, College of Medicine, Gainesville, Florida
32610, United States
- Southeast Center for Integrated
Metabolomics (SECIM), Clinical and Translational Science Institute,
University of Florida, Gainesville,
Florida 32610, United States
- University of Florida Health,
University of Florida, Gainesville,
Florida 32610, United States
| | - Timothy J. Garrett
- Department of Pathology, Immunology,
and Laboratory Medicine, University of
Florida, College of Medicine, Gainesville, Florida
32610, United States
- Southeast Center for Integrated
Metabolomics (SECIM), Clinical and Translational Science Institute,
University of Florida, Gainesville,
Florida 32610, United States
- University of Florida Health,
University of Florida, Gainesville,
Florida 32610, United States
| |
Collapse
|
11
|
Sukkaew A, Suksatu A, Roytrakul S, Smith DR, Ubol S. Proteomic analysis of CHIKV-infected human fibroblast-like synoviocytes: Identification of host factors potentially associated with CHIKV replication and cellular pathogenesis. Microbiol Immunol 2020; 64:445-457. [PMID: 32246487 DOI: 10.1111/1348-0421.12793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 01/02/2023]
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne virus that causes arthralgic fever. Fibroblast-like synoviocytes play a key role in joint damage in inflammatory arthritides and can additionally serve as target cells for CHIKV infection. To gain a better understanding of CHIKV-induced arthralgia, the interaction between CHIKV and synoviocytes was investigated at the protein level. A gel-enhanced liquid chromatography-mass spectrometry (GeLC-MS/MS) approach was used to examine protein expression from primary human fibroblast-like synoviocytes (HFLS) infected with clinical isolates of CHIKV at 12 and 24 hr post infection. Our analysis identified 259 and 241 proteins of known function that were differentially expressed (>1.5 or <-1.5 fold change) following CHIKV infection at 12 and 24 hpi, respectively. These proteins are involved in cellular homeostasis, including cellular trafficking, cytoskeletal organization, immune response, metabolic process, and protein modification. Some of these proteins have previously been reported to participate in arthralgia/arthritis and the death of infected cells. Our results provide information on the CHIKV-induced modulation of cellular proteins of HFLS at an early stage of infection, as well as highlighting biological processes associated with CHIKV infection in the main target cells of the joint.
Collapse
Affiliation(s)
- Apamas Sukkaew
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ampa Suksatu
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, Genome Institute, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Duncan R Smith
- Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Thailand.,Institute of Molecular Bioscience, Mahidol University Salaya Campus, Nakorn Pathom, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Thailand
| |
Collapse
|
12
|
Srivastava P, Kumar A, Hasan A, Mehta D, Kumar R, Sharma C, Sunil S. Disease Resolution in Chikungunya-What Decides the Outcome? Front Immunol 2020; 11:695. [PMID: 32411133 PMCID: PMC7198842 DOI: 10.3389/fimmu.2020.00695] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Chikungunya disease (CHIKD) is a viral infection caused by an alphavirus, chikungunya virus (CHIKV), and triggers large outbreaks leading to epidemics. Despite the low mortality rate, it is a major public health concern owing to high morbidity in affected individuals. The complete spectrum of this disease can be divided into four phases based on its clinical presentation and immunopathology. When a susceptible individual is bitten by an infected mosquito, the bite triggers inflammatory responses attracting neutrophils and initiating a cascade of events, resulting in the entry of the virus into permissive cells. This phase is termed the pre-acute or the intrinsic incubation phase. The virus utilizes the cellular components of the innate immune system to enter into circulation and reach primary sites of infection such as the lymph nodes, spleen, and liver. Also, at this point, antigen-presenting cells (APCs) present the viral antigens to the T cells thereby activating and initiating adaptive immune responses. This phase is marked by the exhibition of clinical symptoms such as fever, rashes, arthralgia, and myalgia and is termed the acute phase of the disease. Viremia reaches its peak during this phase, thereby enhancing the antigen-specific host immune response. Simultaneously, T cell-mediated activation of B cells leads to the formation of CHIKV specific antibodies. Increase in titres of neutralizing IgG/IgM antibodies results in the clearance of virus from the bloodstream and marks the initiation of the post-acute phase. Immune responses mounted during this phase of the infection determine the degree of disease progression or its resolution. Some patients may progress to a chronic arthritic phase of the disease that may last from a few months to several years, owing to a compromised disease resolution. The present review discusses the immunopathology of CHIKD and the factors that dictate disease progression and its resolution.
Collapse
Affiliation(s)
- Priyanshu Srivastava
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ankit Kumar
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Abdul Hasan
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Divya Mehta
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Ramesh Kumar
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Chetan Sharma
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sujatha Sunil
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
13
|
Plasma/serum proteomics: depletion strategies for reducing high-abundance proteins for biomarker discovery. Bioanalysis 2019; 11:1799-1812. [PMID: 31617391 DOI: 10.4155/bio-2019-0145] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Plasma and serum are widely used for proteomics-based biomarker discovery. However, analysis of these biofluids is highly challenging due to the complexity and wide dynamic range of their proteomes. Notably, highly abundant proteins tend to obscure the detection of potential biomarkers that are usually of lower concentrations. Among the strategies to resolve this problem are: depletion of high-abundance proteins, enrichment of low abundant proteins of interest and prefractionation. In this review, we focus on current and emerging depletion techniques used to enhance the detection and identification of the less abundant proteins in plasma and serum. We discuss the applications and contributions of these methods to proteomics analysis of plasma and serum alongside their limitations and future perspectives.
Collapse
|
14
|
Vasconcellos AF, Silva JMF, de Oliveira AS, Prado PS, Nagata T, Resende RO. Genome sequences of chikungunya virus isolates circulating in midwestern Brazil. Arch Virol 2019; 164:1205-1208. [PMID: 30729309 DOI: 10.1007/s00705-019-04174-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/16/2019] [Indexed: 12/17/2022]
Abstract
Chikungunya virus (CHIKV) is a reemerging arbovirus of the family Togaviridae that causes CHIKV fever, a disease that can extend from weeks to years depending on whether clinical signs of arthralgia persist. CHIKV is mainly transmitted by Aedes aegypti mosquitoes and possibly reached the Americas in 2013, causing an outbreak in Brazil in 2015. So far, two evolutionary lineages of CHIKV have been reported in Brazil: the Asian and the East-Central-South African (ECSA) lineages. In this study, six CHIKV isolates circulating in midwestern Brazil (Mato Grosso state) were isolated from patient sera, and their complete genomes were sequenced using a high-throughput sequencing platform. All of these isolates shared high nucleotide sequence similarity with CHIKV isolates from northeastern Brazil and were found to belong to the ECSA lineage. These CHIKV isolates did not contain the A226V or L210Q mutations that are associated with increased transmissibility by A. albopictus, suggesting that the CHIKV isolates circulating in midwestern Brazil are predominantly transmitted by A. aegypti.
Collapse
Affiliation(s)
- A F Vasconcellos
- Departamento de Biologia Celular, Bloco K Térreo, Instituto de Biologia, Universidade de Brasília Campus Darcy Ribeiro, Brasília, DF, CEP 70910-970, Brazil
| | - J M F Silva
- Departamento de Biologia Celular, Bloco K Térreo, Instituto de Biologia, Universidade de Brasília Campus Darcy Ribeiro, Brasília, DF, CEP 70910-970, Brazil
| | - A S de Oliveira
- Departamento de Biologia Celular, Bloco K Térreo, Instituto de Biologia, Universidade de Brasília Campus Darcy Ribeiro, Brasília, DF, CEP 70910-970, Brazil
| | - P S Prado
- Laboratório Central de Saúde Pública do Distrito Federal (LACEN-DF), Setor de Grandes Áreas Norte, Quadras 601, Blocos Oe P., Brasília, DF, CEP 70830-010, Brazil
| | - T Nagata
- Departamento de Biologia Celular, Bloco K Térreo, Instituto de Biologia, Universidade de Brasília Campus Darcy Ribeiro, Brasília, DF, CEP 70910-970, Brazil
| | - R O Resende
- Departamento de Biologia Celular, Bloco K Térreo, Instituto de Biologia, Universidade de Brasília Campus Darcy Ribeiro, Brasília, DF, CEP 70910-970, Brazil.
| |
Collapse
|
15
|
Advances in Clinical Diagnosis and Management of Chikungunya Virus Infection. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2018. [DOI: 10.1007/s40506-018-0172-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Abraham R, Singh S, Nair SR, Hulyalkar NV, Surendran A, Jaleel A, Sreekumar E. Nucleophosmin (NPM1)/B23 in the Proteome of Human Astrocytic Cells Restricts Chikungunya Virus Replication. J Proteome Res 2017; 16:4144-4155. [PMID: 28959884 DOI: 10.1021/acs.jproteome.7b00513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chikungunya virus (CHIKV), a positive-stranded RNA virus, can cause neurological complications by infecting the major parenchymal cells of the brain such as neurons and astrocytes. A proteomic analysis of CHIKV-infected human astrocytic cell line U-87 MG revealed tight functional associations among the modulated proteins. The predominant cellular pathways involved were of transcription-translation machinery, cytoskeletol reorganization, apoptosis, ubiquitination, and metabolism. In the proteome, we could also identify a few proteins that are reported to be involved in host-virus interactions. One such protein, Nucleophosmin (NPM1)/B23, a nucleolar protein, showed enhanced cytoplasmic aggregation in CHIKV-infected cells. NPM1 aggregation was predominantly localized in areas wherein CHIKV antigen could be detected. Furthermore, we observed that inhibition of this aggregation using a specific NPM1 oligomerization inhibitor, NSC348884, caused a significant dose-dependent enhancement in virus replication. There was a marked increase in the amount of intracellular viral RNA, and ∼105-fold increase in progeny virions in infected cells. Our proteomic analysis provides a comprehensive spectrum of host proteins modulated in response to CHIKV infection in astrocytic cells. Our results also show that NPM1/B23, a multifunctional chaperone, plays a critical role in restricting CHIKV replication and is a possible target for antiviral strategies.
Collapse
Affiliation(s)
- Rachy Abraham
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Sneha Singh
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Sreeja R Nair
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Neha Vijay Hulyalkar
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Arun Surendran
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Abdul Jaleel
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory and ‡Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology (RGCB) , Thiruvananthapram 695014, Kerala, India
| |
Collapse
|
17
|
Oxford KL, Wendler JP, McDermott JE, White III RA, Powell JD, Jacobs JM, Adkins JN, Waters KM. The landscape of viral proteomics and its potential to impact human health. Expert Rev Proteomics 2016; 13:579-91. [DOI: 10.1080/14789450.2016.1184091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
18
|
Treffers EE, Tas A, Scholte FE, Van MN, Heemskerk MT, de Ru AH, Snijder EJ, van Hemert MJ, van Veelen PA. Temporal SILAC-based quantitative proteomics identifies host factors involved in chikungunya virus replication. Proteomics 2015; 15:2267-80. [DOI: 10.1002/pmic.201400581] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/12/2015] [Accepted: 03/06/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Emmely E. Treffers
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
- Department of Immunohematology and Blood transfusion; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Ali Tas
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Florine E.M. Scholte
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Myrthe N. Van
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Matthias T. Heemskerk
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Arnoud H. de Ru
- Department of Immunohematology and Blood transfusion; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Eric J. Snijder
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Martijn J. van Hemert
- Molecular Virology Laboratory; Department of Medical Microbiology; Leiden University Medical Center; ZA Leiden The Netherlands
| | - Peter A. van Veelen
- Department of Immunohematology and Blood transfusion; Leiden University Medical Center; ZA Leiden The Netherlands
| |
Collapse
|
19
|
High throughput proteomic analysis and a comparative review identify the nuclear chaperone, Nucleophosmin among the common set of proteins modulated in Chikungunya virus infection. J Proteomics 2015; 120:126-41. [PMID: 25782748 PMCID: PMC7102674 DOI: 10.1016/j.jprot.2015.03.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 02/14/2015] [Accepted: 03/04/2015] [Indexed: 01/14/2023]
Abstract
Global re-emergence of Chikungunya virus (CHIKV) has renewed the interest in its cellular pathogenesis. We subjected CHIKV-infected Human Embryo Kidney cells (HEK293), a widely used cell-based system for CHIKV infection studies, to a high throughput expression proteomics analysis by Liquid Chromatography–tandem mass spectrometry. A total of 1047 differentially expressed proteins were identified in infected cells, consistently in three biological replicates. Proteins involved in transcription, translation, apoptosis and stress response were the major ones among the 209 proteins that had significant up-regulation. In the set of 45 down-regulated proteins, those involved in carbohydrate and lipid metabolism predominated. A STRING network analysis revealed tight interaction of proteins within the apoptosis, stress response and protein synthesis pathways. We short-listed a common set of 30 proteins that can be implicated in cellular pathology of CHIKV infection by comparing our results and results of earlier CHIKV proteomics studies. Modulation of eight proteins selected from this set was re-confirmed at transcript level. One among them, Nucleophosmin, a nuclear chaperone, showed temporal modulation and cytoplasmic aggregation upon CHIKV infection in double immunofluorescence staining and confocal microscopy. The short-listed cellular proteins will be potential candidates for targeted study of the molecular interactions of CHIKV with host cells. Biological significance Chikungunya remained as a neglected tropical disease till its re-emergence in 2005 in the La RéUnion islands and subsequently, in India and many parts of South East Asia. These and the epidemics that followed in subsequent years ran an explosive course leading to extreme morbidity and attributed mortality to this originally benign virus infection. Apart from classical symptoms of acute fever and debilitating polyarthralgia lasting for several weeks, a number of complications were documented. These included aphthous-like ulcers and vesiculo-bullous eruptions on the skin, hepatic involvement, central nervous system complications such as encephalopathy and encephalitis, and transplacental transmission. The disease has recently spread to the Americas with its initial documentation in the Caribbean islands. The Asian genotype of this positive-stranded RNA virus of the Alphavirus genus has been attributed in these outbreaks. However, the disease ran a similar course as the one caused by the East, Central and South African (ECSA) genotype in the other parts of the world. Studies have documented a number of mutations in the re-emerging strains of the virus that enhances mosquito adaptability and modulates virus infectivity. This might support the occurrence of fiery outbreaks in the absence of herd immunity in affected population. Several research groups work to understand the pathogenesis of chikungunya and the mechanisms of complications using cellular and animal models. A few proteomics approaches have been employed earlier to understand the protein level changes in the infected cells. Our present study, which couples a high throughput proteomic analysis and a comparative review of these earlier studies, identifies a few critical molecules as hypothetical candidates that might be important in this infection and for future study. High throughput expression proteomics analysis in HEK293 cells Identified four major cellular pathways affected in Chikungunya virus infection Short-listed 30 key proteins modulated by a comparative review Confirmed modulation of Nucleophosmin and other selected proteins upon infection
Collapse
|
20
|
Long KM, Heise MT. Protective and Pathogenic Responses to Chikungunya Virus Infection. CURRENT TROPICAL MEDICINE REPORTS 2015; 2:13-21. [PMID: 26366337 DOI: 10.1007/s40475-015-0037-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chikungunya virus (CHIKV) is an arbovirus responsible for causing epidemic outbreaks of human disease characterized by painful and often debilitating arthralgia. Recently CHIKV has moved into the Caribbean and the Americas resulting in massive outbreaks in naïve human populations. Given the importance of CHIKV as an emerging disease, a significant amount of effort has gone into interpreting the virus-host interactions that contribute to protection or virus-induced pathology following CHIKV infection, with the long term goal of using this information to develop new therapies or safe and effective anti-CHIKV vaccines. This work has made it clear that numerous distinct host responses are involved in the response to CHIKV infection, where some aspects of the host innate and adaptive immune response protect from or limit virus-induced disease, while other pathways actually exacerbate the virus-induced disease process. This review will discuss mechanisms that have been identified as playing a role in the host response to CHIKV infection and illustrate the importance of carefully evaluating these responses to determine whether they play a protective or pathologic role during CHIKV infection.
Collapse
Affiliation(s)
- Kristin M Long
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, CB 7292, Chapel Hill, NC 27599
| | - Mark T Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, CB 7292, Chapel Hill, NC 27599 ; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, CB 7292, Chapel Hill, NC 27599
| |
Collapse
|
21
|
A comprehensive proteomic analysis of totarol induced alterations in Bacillus subtilis by multipronged quantitative proteomics. J Proteomics 2015; 114:247-62. [DOI: 10.1016/j.jprot.2014.10.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/28/2014] [Accepted: 10/20/2014] [Indexed: 12/25/2022]
|
22
|
Sahu A, Kumar S, Sreenivasamurthy SK, Selvan LDN, Madugundu AK, Yelamanchi SD, Puttamallesh VN, Dey G, Anil AK, Srinivasan A, Mukherjee KK, Gowda H, Satishchandra P, Mahadevan A, Pandey A, Prasad TSK, Shankar SK. Host response profile of human brain proteome in toxoplasma encephalitis co-infected with HIV. Clin Proteomics 2014; 11:39. [PMID: 25404878 PMCID: PMC4232683 DOI: 10.1186/1559-0275-11-39] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 09/02/2014] [Indexed: 01/27/2023] Open
Abstract
Background Toxoplasma encephalitis is caused by the opportunistic protozoan parasite Toxoplasma gondii. Primary infection with T. gondii in immunocompetent individuals remains largely asymptomatic. In contrast, in immunocompromised individuals, reactivation of the parasite results in severe complications and mortality. Molecular changes at the protein level in the host central nervous system and proteins associated with pathogenesis of toxoplasma encephalitis are largely unexplored. We used a global quantitative proteomic strategy to identify differentially regulated proteins and affected molecular networks in the human host during T. gondii infection with HIV co-infection. Results We identified 3,496 proteins out of which 607 proteins were differentially expressed (≥1.5-fold) when frontal lobe of the brain from patients diagnosed with toxoplasma encephalitis was compared to control brain tissues. We validated differential expression of 3 proteins through immunohistochemistry, which was confirmed to be consistent with mass spectrometry analysis. Pathway analysis of differentially expressed proteins indicated deregulation of several pathways involved in antigen processing, immune response, neuronal growth, neurotransmitter transport and energy metabolism. Conclusions Global quantitative proteomic approach adopted in this study generated a comparative proteome profile of brain tissues from toxoplasma encephalitis patients co-infected with HIV. Differentially expressed proteins include previously reported and several new proteins in the context of T. gondii and HIV infection, which can be further investigated. Molecular pathways identified to be associated with the disease should enhance our understanding of pathogenesis in toxoplasma encephalitis. Electronic supplementary material The online version of this article (doi:10.1186/1559-0275-11-39) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Apeksha Sahu
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India
| | - Satwant Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Sreelakshmi K Sreenivasamurthy
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Manipal University, Madhav Nagar, Manipal, 576104 India
| | - Lakshmi Dhevi N Selvan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Amrita School of Biotechnology, Amrita University, Kollam, 690525 India
| | - Anil K Madugundu
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India
| | - Soujanya D Yelamanchi
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; School of Biotechnology, KIIT University, Bhubaneswar, 751024 India
| | | | - Gourav Dey
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Manipal University, Madhav Nagar, Manipal, 576104 India
| | | | - Anand Srinivasan
- Department of Pharmacology, Postgraduate Institute of Medical Education & Research, Chandigarh, 160012 India
| | - Kanchan K Mukherjee
- Department of Neurosurgery, Postgraduate Institute of Medical Education & Research, Chandigarh, 160012 India
| | - Harsha Gowda
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | | | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India ; Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ; Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 1205 USA ; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA ; The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Thottethodi Subrahmanya Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India ; Bioinformatics Centre, School of Life Sciences, Pondicherry University, Puducherry, 605014 India ; Manipal University, Madhav Nagar, Manipal, 576104 India ; Amrita School of Biotechnology, Amrita University, Kollam, 690525 India ; NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India ; Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences, Bangalore, 560029 India
| |
Collapse
|
23
|
Smith DR. Global protein profiling studies of chikungunya virus infection identify different proteins but common biological processes. Rev Med Virol 2014; 25:3-18. [PMID: 25066270 DOI: 10.1002/rmv.1802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/26/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022]
Abstract
Chikungunya fever (CHIKF) caused by the mosquito-transmitted chikungunya virus (CHIKV) swept into international prominence from late 2005 as an epidemic of CHIKF spread around countries surrounding the Indian Ocean. Although significant advances have been made in understanding the pathobiology of CHIKF, numerous questions still remain. In the absence of commercially available specific drugs to treat the disease, or a vaccine to prevent the diseases, the questions have particular significance. A number of studies have used global proteome analysis to increase our understanding of the process of CHIKV infection using a number of different experimental techniques and experimental systems. In all, over 700 proteins have been identified in nine different analyses by five different groups as being differentially regulated. Remarkably, only a single protein, eukaryotic elongation factor 2, has been identified by more than two different groups as being differentially regulated during CHIKV infection. This review provides a critical overview of the studies that have used global protein profiling to understand CHIKV infection and shows that while a broad consensus is emerging on which biological processes are altered during CHIKV infection, this consensus is poorly supported in terms of consistent identification of any key proteins mediating those biological processes.
Collapse
Affiliation(s)
- Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand; Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Thailand
| |
Collapse
|