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. [PMID: 38717066 DOI: 10.1089/vbz.2024.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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 P RIDE 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
|
Tang H, Sun M, Duan W, Ying Y, Liao Y, Xiao T, Li Y. Nucleophosmin 1a translocated from nucleus to cytoplasm and facilitate GCRV replication. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109153. [PMID: 37821004 DOI: 10.1016/j.fsi.2023.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
To decipher the functional characterization of Nucleophosmin 1a (NPM1a) from grass carp (Ctenopharyngodon idellus) (CiNPM1a), its cDNA was cloned and bioinformatic analysis were conducted. The full-length cDNA sequence of CiNPM1a is 1732 bp, which encodes 307 amino acids. CiNPM1a contains conserved domains of Nucleoplasmin domain, NPM1-C terminal domain, as well as nuclear localization signals, nuclear export signal (NES) and acid patches. There are 52 and 20 consensus amino acids exist in the Nucleoplasmin domain and the NPM1-C terminal domain of all blasted species. In addition, the immune function of CiNPM1a were analyzed. The Ciirf7, Ciifn1 and Ciifn2 transcription was inhibited, whereas the vp2 and vp7 expressions were enhanced in CiNPM1a overexpressing cells after GCRV infection (P < 0.05). Moreover, the Ciirf7, Ciifn1 and Ciifn2 mRNA levels were significantly up-regulated, but the vp2 and vp7 expressions were significantly down-regulated in CiNPM1a knockdown cells after infection. This indicated that CiNPM1a played negative roles in the induction of Type I IFN reaction and thus the GCRV replication. Finally, the NES domain that affect the nucleous-cytoplasm shuttle and the replication of GCRV were investigated. The deletion of NES1 and NES(1 + 2+3) absolutely limited the transloacation of CiNPM1a△NES1 protein and CiNPM1a △NES(1 + 2+3) protein to cytoplasm after infection, and the deletion of NES2 resulted in partially limitation of protein shuttle. In general, Ciirf3, Ciirf7, Ciifn1 and Ciifn2 expressions were enhanced in the CiNPM1a△NES1, CiNPM1a△NES2 and CiNPM1a△NES3 overexpression groups, and the deletion of functional domains in CiNPM1a led to significantly reduction of the vp2 and vp7 replication. The results indicated that CiNPM1a may be a target molecular for GCRV infection curation, and a candidate molecular for resistance strain breeding of grass carp.
Collapse
Affiliation(s)
- Hao Tang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Mingxue Sun
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Wei Duan
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Yanrong Ying
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Yijing Liao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China
| | - Yaoguo Li
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
3
|
Pradeep P, Sivakumar KC, Sreekumar E. Host Factor Nucleophosmin 1 (NPM1/B23) Exerts Antiviral Effects against Chikungunya Virus by Its Interaction with Viral Nonstructural Protein 3. Microbiol Spectr 2023; 11:e0537122. [PMID: 37409962 PMCID: PMC10433958 DOI: 10.1128/spectrum.05371-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 07/07/2023] Open
Abstract
Chikungunya virus (CHIKV) hijacks host cell machinery to support its replication. Nucleophosmin 1 (NPM1/B23), a nucleolar phosphoprotein, is one of the host proteins known to restrict CHIKV infection; however, the mechanistic details of the antiviral role of NPM1 are not elucidated. It was seen in our experiments that the level of NPM1 expression affected the expression levels of interferon-stimulated genes (ISGs) that play antiviral roles in CHIKV infection, such as IRF1, IRF7, OAS3, and IFIT1, indicating that one of the antiviral mechanisms could be through modulation of interferon-mediated pathways. Our experiments also identified that for CHIKV restriction, NPM1 must move from the nucleus to the cytoplasm. A deletion of the nuclear export signal (NES), which confines NPM1 within the nucleus, abolishes its anti-CHIKV action. We observed that NPM1 binds CHIKV nonstructural protein 3 (nsP3) strongly via its macrodomain, thereby exerting a direct interaction with viral proteins to limit infection. Based on site-directed mutagenesis and coimmunoprecipitation studies, it was also observed that amino acid residues N24 and Y114 of the CHIKV nsP3 macrodomain, known to be involved in virus virulence, bind ADP-ribosylated NPM1 to inhibit infection. Overall, the results show a key role of NPM1 in CHIKV restriction and indicate it as a promising host target for developing antiviral strategies against CHIKV. IMPORTANCE Chikungunya, a recently reemerged mosquito-borne infection caused by a positive-sense, single-stranded RNA virus, has caused explosive epidemics in tropical regions. Unlike the classical symptoms of acute fever and debilitating arthralgia, incidences of neurological complications and mortality were reported. Currently there are no antivirals or commercial vaccines available against chikungunya. Like all viruses, CHIKV uses host cellular machinery for establishment of infection and successful replication. To counter this, the host cell activates several restriction factors and innate immune response mediators. Understanding these host-virus interactions helps to develop host-targeted antivirals against the disease. Here, we report the antiviral role of the multifunctional host protein NPM1 against CHIKV. The significant inhibitory effect of this protein against CHIKV involves its increased expression and movement from its natural location within the nucleus to the cytoplasm. There, it interacts with functional domains of key viral proteins. Our results support ongoing efforts toward development of host-directed antivirals against CHIKV and other alphaviruses.
Collapse
Affiliation(s)
- Parvanendhu Pradeep
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Research Centre, University of Kerala, Thiruvananthapuram, India
| | | | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Thiruvananthapuram, India
| |
Collapse
|
4
|
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
|
5
|
Kumar R, Mehta D, Chaudhary S, Nayak D, Sunil S. Impact of CHIKV Replication on the Global Proteome of Aedes albopictus Cells. Proteomes 2022; 10:proteomes10040038. [PMID: 36412637 PMCID: PMC9680348 DOI: 10.3390/proteomes10040038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/12/2022] Open
Abstract
Arboviruses are some of the important causative agents of mosquito-mediated viral diseases. These viruses are transmitted between vector and host during the blood meal. Upon viral entry, host replication machinery is hijacked, supporting new virus particle production and thereby allowing viral survival in the host. In this process, host proteins interact with viral proteins to either facilitate viral replication, or they may provide antiviral defense mechanisms. In this study, we analyzed the impact of chikungunya virus (CHIKV) infection on the global proteome of Dicer active Aedes albopictus cells during the early and late time points of infection. We utilized a bottom-up approach of global proteomics analysis, and we used label-free quantitative mass spectrometry to identify the global protein signatures of Ae. albopictus at two different time points upon CHIKV infection. The mass spectrometry data analysis of the early time point revealed that proteins belonging to pathways such as translation, RNA processing, and cellular metabolic processes were less in abundance, whereas those belonging to pathways such as cellular catabolic process and organic substance transport were significantly abundant. At later time points, proteins belonging to pathways such as cellular metabolic processes, primary metabolic process, organonitrogen compound metabolic process, and organic substance metabolic process were found to be decreased in their presence, whereas those belonging to pathways such as RNA processing, gene expression, macromolecule metabolic processing, and nitrogen compound metabolic processing were found to be abundant during CHIKV infection, indicating that modulation in gene expression favoring cell survival occurs at a later time point, suggesting a survival strategy of Aedes cells to counter prolonged CHIKV infection.
Collapse
Affiliation(s)
- Ramesh Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore 453552, India
| | - Divya Mehta
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Sakshi Chaudhary
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Debasis Nayak
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore 453552, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
- Correspondence:
| |
Collapse
|
6
|
Monsalve-Escudero LM, Loaiza-Cano V, Pájaro-González Y, Oliveros-Díaz AF, Diaz-Castillo F, Quiñones W, Robledo S, Martinez-Gutierrez M. Indole alkaloids inhibit zika and chikungunya virus infection in different cell lines. BMC Complement Med Ther 2021; 21:216. [PMID: 34454481 PMCID: PMC8397866 DOI: 10.1186/s12906-021-03386-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/05/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In recent years, an increase in the occurrence of illnesses caused by two clinically- important arboviruses has been reported: Zika virus (ZIKV) and Chikungunya virus (CHIKV). There is no licensed antiviral treatment for either of the two abovementioned viruses. Bearing in mind that the antiviral effect of indole alkaloids has been reported for other arboviral models, the present study proposed to evaluate the antiviral in vitro and in silico effects of four indole alkaloids on infections by these two viruses in different cell lines. METHODS The antiviral effects of voacangine (VOAC), voacangine-7-hydroxyindolenine (VOAC-OH), rupicoline and 3-oxo voacangine (OXO-VOAC) were evaluated in Vero, U937 and A549 cells using different experimental strategies (Pre, Trans, Post and combined treatment). Viral infection was quantified by different methodologies, including infectious viral particles by plating, viral genome by RT-qPCR, and viral protein by cell ELISA. Moreover, molecular docking was used to evaluate the possible interactions between structural and nonstructural viral proteins and the compounds. The results obtained from the antiviral strategies for each experimental condition were compared in all cases with the untreated controls. Statistically significant differences were identified using a parametric Student's t-test. In all cases, p values below 0.05 (p < 0.05) were considered statistically significant. RESULTS In the pre-treatment strategy in Vero cells, VOAC and VOAC-OH inhibited both viral models and OXO-VOAC inhibited only ZIKV; in U937 cells infected with CHIKV/Col, only VOAC-OH inhibited infection, but none of the compounds had activity in A549 cells; in U937 cells and A549 cells infected with ZIKV/Col, the three compounds that were effective in Vero cells also had antiviral activity. In the trans-treatment strategy, only VOAC-OH was virucidal against ZIKV/Col. In the post-treatment strategy, only rupicoline was effective in the CHIKV/Col model in Vero and A549 cells, whereas VOAC and VOAC-OH inhibited ZIKV infection in all three cell lines. In the combined strategy, VOAC, VOAC-OH and rupicoline inhibited CHIKV/Col and ZIKV/Col, but only rupicoline improved the antiviral effect of ZIKV/Col-infected cultures with respect to the individual strategies. Molecular docking showed that all the compounds had favorable binding energies with the structural proteins E2 and NSP2 (CHIKV) and E and NS5 (ZIKV). CONCLUSIONS The present study demonstrates that indole alkaloids are promising antiviral drugs in the process of ZIKV and CHIKV infection; however, the mechanisms of action evaluated in this study would indicate that the effect is different in each viral model and, in turn, dependent on the cell line.
Collapse
Affiliation(s)
- Laura Milena Monsalve-Escudero
- Grupo de Investigación en Ciencias Animales-GRICA. Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Vanessa Loaiza-Cano
- Grupo de Investigación en Ciencias Animales-GRICA. Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Yina Pájaro-González
- Laboratorio de Investigaciones Fitoquímicas y Farmacológicas de la Universidad de Cartagena - LIFFUC, Universidad de Cartagena, Cartagena, Colombia.,Grupo de Investigación en Farmacia Asistencial y Farmacología, Universidad del Atlántico, Barranquilla, Colombia
| | - Andrés Felipe Oliveros-Díaz
- Laboratorio de Investigaciones Fitoquímicas y Farmacológicas de la Universidad de Cartagena - LIFFUC, Universidad de Cartagena, Cartagena, Colombia
| | - Fredyc Diaz-Castillo
- Laboratorio de Investigaciones Fitoquímicas y Farmacológicas de la Universidad de Cartagena - LIFFUC, Universidad de Cartagena, Cartagena, Colombia
| | - Wiston Quiñones
- Grupo de Química Orgánica de Productos Naturales. Universidad de Antioquia, Medellín, Colombia
| | - Sara Robledo
- Programa de Estudio y Control de Enfermedades Tropicales-PECET, Universidad de Antioquia, Medellín, Colombia
| | - Marlen Martinez-Gutierrez
- Grupo de Investigación en Ciencias Animales-GRICA. Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia.
| |
Collapse
|
7
|
Mudaliar P, Pradeep P, Abraham R, Sreekumar E. Targeting cap-dependent translation to inhibit Chikungunya virus replication: selectivity of p38 MAPK inhibitors to virus-infected cells due to autophagy-mediated down regulation of phospho-ERK. J Gen Virol 2021; 102. [PMID: 34328830 DOI: 10.1099/jgv.0.001629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The 5' capped, message-sense RNA genome of Chikungunya virus (CHIKV) utilizes the host cell machinery for translation. Translation is regulated by eIF2 alpha at the initiation phase and by eIF4F at cap recognition. Translational suppression by eIF2 alpha phosphorylation occurs as an early event in many alphavirus infections. We observe that in CHIKV-infected HEK293 cells, this occurs as a late event, by which time the viral replication has reached an exponential phase, implying its minimal role in virus restriction. The regulation by eIF4F is mediated through the PI3K-Akt-mTOR, p38 MAPK and RAS-RAF-MEK-ERK pathways. A kinetic analysis revealed that CHIKV infection did not modulate AKT phosphorylation, but caused a significant reduction in p38 MAPK phosphorylation. It caused degradation of phospho-ERK 1/2 by increased autophagy, leaving the PI3K-Akt-mTOR and p38 MAPK pathways for pharmacological targeting. mTOR inhibition resulted in moderate reduction in viral titre, but had no effect on CHIKV E2 protein expression, indicating a minimal role of the mTOR complex in virus replication. Inhibition of p38 MAPK using SB202190 caused a significant reduction in viral titre and CHIKV E2 and nsP3 protein expression. Furthermore, inhibiting the two pathways together did not offer any synergism, indicating that inhibiting the p38 MAPK pathway alone is sufficient to cause restriction of CHIKV replication. Meanwhile, in uninfected cells the fully functional RAS-RAF-MEK-ERK pathway can circumvent the effect of p38 MAPK inhibition on cap-dependent translation. Thus, our results show that host-directed antiviral strategies targeting cellular p38 MAPK are worth exploring against Chikungunya as they could be selective against CHIKV-infected cells with minimal effects on uninfected host cells.
Collapse
Affiliation(s)
- Prashant Mudaliar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India.,Research Centre, University of Kerala, Thiruvananthapuram 695034, Kerala, India
| | - Parvanendhu Pradeep
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India.,Research Centre, University of Kerala, Thiruvananthapuram 695034, Kerala, India
| | - Rachy Abraham
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| |
Collapse
|
8
|
Application of multiple omics and network projection analyses to drug repositioning for pathogenic mosquito-borne viruses. Sci Rep 2021; 11:10136. [PMID: 33980888 PMCID: PMC8115341 DOI: 10.1038/s41598-021-89171-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Pathogenic mosquito-borne viruses are a serious public health issue in tropical and subtropical regions and are increasingly becoming a problem in other climate zones. Drug repositioning is a rapid, pharmaco-economic approach that can be used to identify compounds that target these neglected tropical diseases. We have applied a computational drug repositioning method to five mosquito-borne viral infections: dengue virus (DENV), zika virus (ZIKV), West Nile virus (WNV), Japanese encephalitis virus (JEV) and Chikungunya virus (CHIV). We identified signature molecules and pathways for each virus infection based on omics analyses, and determined 77 drug candidates and 146 proteins for those diseases by using a filtering method. Based on the omics analyses, we analyzed the relationship among drugs, target proteins and the five viruses by projecting the signature molecules onto a human protein-protein interaction network. We have classified the drug candidates according to the degree of target proteins in the protein-protein interaction network for the five infectious diseases.
Collapse
|
9
|
Kronemberger GS, Carneiro FA, Rezende DF, Baptista LS. Spheroids and organoids as humanized 3D scaffold-free engineered tissues for SARS-CoV-2 viral infection and drug screening. Artif Organs 2021; 45:548-558. [PMID: 33264436 PMCID: PMC7753831 DOI: 10.1111/aor.13880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022]
Abstract
The new coronavirus (2019‐nCoV) or the severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) was officially declared by the World Health Organization (WHO) as a pandemic in March 2020. To date, there are no specific antiviral drugs proven to be effective in treating SARS‐CoV‐2, requiring joint efforts from different research fronts to discover the best route of treatment. The first decisions in drug discovery are based on 2D cell culture using high‐throughput screening. In this context, spheroids and organoids emerge as a reliable alternative. Both are scaffold‐free 3D engineered constructs that recapitulate key cellular and molecular events of tissue physiology. Different studies have already shown their advantages as a model for different infectious diseases, including SARS‐CoV‐2 and for drug screening. The use of these 3D engineered tissues as an in vitro model can fill the gap between 2D cell culture and in vivo preclinical assays (animal models) as they could recapitulate the entire viral life cycle. The main objective of this review is to understand spheroid and organoid biology, highlighting their advantages and disadvantages, and how these scaffold‐free engineered tissues can contribute to a better comprehension of viral infection by SARS‐CoV‐2 and to the development of in vitro high‐throughput models for drug screening.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil.,Postgraduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, Brazil
| | - Fabiana A Carneiro
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil
| | | | - Leandra S Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Rio de Janeiro, Brazil.,Postgraduation Program of Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, Brazil
| |
Collapse
|
10
|
Singh PK, Bohr SSR, Hatzakis NS. Direct Observation of Sophorolipid Micelle Docking in Model Membranes and Cells by Single Particle Studies Reveals Optimal Fusion Conditions. Biomolecules 2020; 10:E1291. [PMID: 32906821 PMCID: PMC7564020 DOI: 10.3390/biom10091291] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/03/2020] [Accepted: 09/05/2020] [Indexed: 11/16/2022] Open
Abstract
Sophorolipids (SLs) are naturally produced glycolipids that acts as drug delivery for a spectrum of biomedical applications, including as an antibacterial antifungal and anticancer agent, where they induce apoptosis selectively in cancerous cells. Despite their utility, the mechanisms underlying their membrane interactions, and consequently cell entry, remains unknown. Here, we combined a single liposome assay to observe directly and quantify the kinetics of interaction of SL micelles with model membrane systems, and single particle studies on live cells to record their interaction with cell membranes and their cytotoxicity. Our single particle readouts revealed several repetitive docking events on individual liposomes and quantified how pH and membrane charges, which are known to vary in cancer cells, affect the docking of SL micelles on model membranes. Docking of sophorolipids micelles was found to be optimal at pH 6.5 and for membranes with -5% negatively charge lipids. Single particle studies on mammalian cells reveled a two-fold increased interaction on Hela cells as compared to HEK-293 cells. This is in line with our cell viability readouts recording an approximate two-fold increased cytotoxicity by SLs interactions for Hela cells as compared to HEK-293 cells. The combined in vitro and cell assays thus support the increased cytotoxicity of SLs on cancer cells to originate from optimal charge and pH interactions between membranes and SL assemblies. We anticipate studies combining quantitative single particle studies on model membranes and live cell may reveal hitherto unknown molecular insights on the interactions of sophorolipid and additional nanocarriers mechanism.
Collapse
Affiliation(s)
- Pradeep Kumar Singh
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Thorvaldsensvej 40, C 1871 Frederiksberg, Denmark
- Department of Chemistry, University of Akron, Akron, OH 44325, USA
| | - Søren S-R Bohr
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Thorvaldsensvej 40, C 1871 Frederiksberg, Denmark
- Novo Nordisk Center for Protein Research (CPR), University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Nikos S Hatzakis
- Department of Chemistry & Nanoscience Center, University of Copenhagen, Thorvaldsensvej 40, C 1871 Frederiksberg, Denmark
- Novo Nordisk Center for Protein Research (CPR), University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
11
|
Gupta S, Mishra KP, Kumar B, Singh SB, Ganju L. Andrographolide Mitigates Unfolded Protein Response Pathway and Apoptosis Involved in Chikungunya Virus Infection. Comb Chem High Throughput Screen 2020; 24:849-859. [PMID: 32819227 DOI: 10.2174/1386207323999200818165029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/20/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chikungunya virus (CHIKV) is an arthropod-borne RNA virus which induces host Endoplasmic Reticulum (ER) stress by accumulating unfolded or misfolded proteins. ER stress activates the unfolded protein response (UPR) pathway to enable proper protein folding and maintain cellular homeostasis. There is no approved drug or vaccine available for CHIKV treatment, therefore, a pharmacological countermeasure is warranted for preventing CHIKV infection. OBJECTIVE With a view to find a treatment modality for chikungunya infection, "andrographolide", a plant-derived diterpenoid with reported antiviral, anti-inflammatory and immunomodulatory effects, was used to investigate its role in chikungunya induced unfolded protein stress and apoptosis. METHODS Cells and supernatant collected on andrographolide and VER-155008, a GRP78 inhibitor, treatment in CHIKV infected and mock-infected THP-1 cells were tested for differential expression of UPR pathway proteins including GRP78, PERK, EIF-2α, IRE-1α, XBP-1 and ATF6. Furthermore, the inflammasome and apoptosis pathway proteins, i.e., caspase-1, caspase-3 and PARP, were tested by immunoblotting, and cytokines, i.e., IL-1β, IL-6 and IFN-γ were tested by ELISA. RESULTS Andrographolide treatment in CHIKV infected THP-1 cells significantly reduced IRE1α and downstream spliced XBP1 protein expression. Furthermore, CHIKV induced apoptosis and viral protein expression were also reduced on andrographolide treatment. A comparative analysis of andrographolide versus VER-155008, confirmed that andrographolide surpasses the effects of VER-155008 in suppressing the CHIKV induced ER stress. CONCLUSION The study, therefore, confirms that andrographolide is a potential remedy for chikungunya infection and suppresses CHIKV induced ER stress and apoptosis.
Collapse
Affiliation(s)
- Swati Gupta
- Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi-110054, India
| | - Kamla Prasad Mishra
- Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi-110054, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi-110054, India
| | - Shashi Bala Singh
- Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi-110054, India
| | - Lilly Ganju
- Defence Institute of Physiology & Allied Sciences (DIPAS), Lucknow Road, Timarpur, Delhi-110054, India
| |
Collapse
|
12
|
Kaur P, Lello LS, Utt A, Dutta SK, Merits A, Chu JJH. Bortezomib inhibits chikungunya virus replication by interfering with viral protein synthesis. PLoS Negl Trop Dis 2020; 14:e0008336. [PMID: 32469886 PMCID: PMC7286522 DOI: 10.1371/journal.pntd.0008336] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 06/10/2020] [Accepted: 04/29/2020] [Indexed: 12/31/2022] Open
Abstract
Chikungunya virus (CHIKV) is an alphavirus that causes a febrile illness accompanied by myalgia and arthralgia. Despite having re-emerged as a significant public health threat, there are no approved therapeutics or prophylactics for CHIKV infection. In this study, we explored the anti-CHIKV effects of proteasome inhibitors and their potential mechanism of antiviral action. A panel of proteasome inhibitors with different functional groups reduced CHIKV infectious titers in a dose-dependent manner. Bortezomib, which has been FDA-approved for multiple myeloma and mantle cell lymphoma, was further investigated in downstream studies. The inhibitory activities of bortezomib were confirmed using different cellular models and CHIKV strains. Time-of-addition and time-of-removal studies suggested that bortezomib inhibited CHIKV at an early, post-entry stage of replication. In western blot analysis, bortezomib treatment resulted in a prominent decrease in structural protein levels as early as 6 hpi. Contrastingly, nsP4 levels showed strong elevations across all time-points. NsP2 and nsP3 levels showed a fluctuating trend, with some elevations between 12 to 20 hpi. Finally, qRT-PCR data revealed increased levels of both positive- and negative-sense CHIKV RNA at late stages of infection. It is likely that the reductions in structural protein levels is a major factor in the observed reductions in virus titer, with the alterations in non-structural protein ratios potentially being a contributing factor. Proteasome inhibitors like bortezomib likely disrupt CHIKV replication through a variety of complex mechanisms and may display a potential for use as therapeutics against CHIKV infection. They also represent valuable tools for studies of CHIKV molecular biology and virus-host interactions.
Collapse
Affiliation(s)
- Parveen Kaur
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Age Utt
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Sujit Krishna Dutta
- School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, Singapore
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Collaborative and Translational Unit for HFMD, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- * E-mail:
| |
Collapse
|
13
|
Ceccin ADF, Souza APD, Hilário GT, Muller DM, Romão PRT, Rodrigues Junior LC. HspBP1 and anti-HspBP1 levels in the serum of HIV-infected individuals are associated to the disease progression. J Appl Microbiol 2019; 127:576-585. [PMID: 30786116 DOI: 10.1111/jam.14230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/16/2019] [Accepted: 02/06/2019] [Indexed: 12/24/2022]
Abstract
AIMS The objective of this research was to quantify the levels of circulating HspBP1 and anti-HspBP1 IgG in HIV-infected individuals and to correlate them with CD4 T cell counts and viral load, as well as to determine the kinetics of those proteins during acute phase. METHODS AND RESULTS Sixty serum samples from HIV-positive outpatients, thirty with high viral load and thirty with low viral load were analysed. The HspBP1 and anti-HspBP1 were quantified by ELISA. To investigate the kinetic of HspBP1 and anti-HspBp1 during the acute phase, these proteins and antibodies were quantified in samples of a commercial seroconverting HIV panel. All dosages were compared with the CD4 and CD8 T cell counts and HIV viral load. The results indicated that HIV positive outpatients presented significant increase in HspBP1 and anti-HspBP1 serum levels, compared with uninfected healthy. HspBP1 and anti-HspBP1 were negatively correlated with CD4 counts and CD4:CD8 ratio. In the acute phase, HspBP1 became significantly elevated 15 days after HIV infection. CONCLUSIONS These results indicate that the quantification of HspBP1 can be associated to others well-established parameters of the HIV progression. SIGNIFICANCE AND IMPACT OF THE STUDY The discovery that HspBp1 and anti-HspBp1 are associated with progression of HIV infection is new and corroborates to validate the quantification of these proteins as an additional strategy in the management of the HIV infection.
Collapse
Affiliation(s)
- A D F Ceccin
- Hemocenter of Santa Maria - Alameda Santiago de Chile Avenue, Santa Maria, RS, Brazil
| | - A P D Souza
- Laboratório de Imunologia Clínica e Experimental, Escola de Ciências. Pontifícia Universidade Católica do Rio Grande do Sul. Av. Ipiranga, Porto Alegre, RS, Brazil
| | - G T Hilário
- Laboratório de Imunologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA). Av. Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - D M Muller
- Laboratório de Imunologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA). Av. Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - P R T Romão
- Laboratório de Imunologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA). Av. Sarmento Leite, 245, Porto Alegre, RS, Brazil
| | - L C Rodrigues Junior
- Laboratório de Imunologia, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA). Av. Sarmento Leite, 245, Porto Alegre, RS, Brazil
| |
Collapse
|
14
|
Lobaina Y, Perera Y. Implication of B23/NPM1 in Viral Infections, Potential Uses of B23/NPM1 Inhibitors as Antiviral Therapy. Infect Disord Drug Targets 2019; 19:2-16. [PMID: 29589547 DOI: 10.2174/1871526518666180327124412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/08/2018] [Accepted: 02/12/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND B23/nucleophosmin (B23/NPM1) is an abundant multifunctional protein mainly located in the nucleolus but constantly shuttling between the nucleus and cytosol. As a consequence of its constitutive expression, intracellular dynamics and binding capacities, B23/NPM1 interacts with multiple cellular factors in different cellular compartments, but also with viral proteins from both DNA and RNA viruses. B23/NPM1 influences overall viral replication of viruses like HIV, HBV, HCV, HDV and HPV by playing functional roles in different stages of viral replication including nuclear import, viral genome transcription and assembly, as well as final particle formation. Of note, some virus modify the subcellular localization, stability and/or increases B23/NPM1 expression levels on target cells, probably to foster B23/NPM1 functions in their own replicative cycle. RESULTS This review summarizes current knowledge concerning the interaction of B23/NPM1 with several viral proteins during relevant human infections. The opportunities and challenges of targeting this well-conserved host protein as a potentially new broad antiviral treatment are discussed in detail. Importantly, although initially conceived to treat cancer, a handful of B23/NPM1 inhibitors are currently available to test on viral infection models. CONCLUSION As B23/NPM1 partakes in key steps of viral replication and some viral infections remain as unsolved medical needs, an appealing idea may be the expedite evaluation of B23/NPM1 inhibitors in viral infections. Furthermore, worth to be addressed is if the up-regulation of B23/NPM1 protein levels that follows persistent viral infections may be instrumental to the malignant transformation induced by virus like HBV and HCV.
Collapse
Affiliation(s)
- Yadira Lobaina
- Therapeutic Hepatitis B Vaccine Group, Vaccine Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| | - Yasser Perera
- Molecular Oncology Group, Pharmaceuticals Division, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Havana, CP 10600, Cuba
| |
Collapse
|
15
|
de Almeida Barreto FK, Montenegro RM, Fernandes VO, Oliveira R, de Araújo Batista LA, Hussain A, de Góes Cavalcanti LP. Chikungunya and diabetes, what do we know? Diabetol Metab Syndr 2018; 10:32. [PMID: 29686737 PMCID: PMC5899414 DOI: 10.1186/s13098-018-0329-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/25/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Chikungunya (CHIK) is a viral disease transmitted by mosquitoes. The first cases in Brazil were confirmed in 2014. Between 2016 and 2017, over 300,000 cases were identified during this period, with nearly 300 deaths. The clinical manifestations, pathogenesis and risk factors for occurrence of severe cases are not yet well understood, although it is known that the severity of the cases is associated with the presence of comorbidities, especially diabetes mellitus (DM). OBJECTIVE To review the medical literature for the associations between DM and CHIK and to understand the potential impact on metabolic state and its complications. METHODS Literature review was carried out to search for articles (English, Portuguese and Spanish) in Medline and Virtual Health Library databases for the period between 1952 and 2017, with the following keywords: "Chikungunya fever", "Chikungunya virus", "diabetes mellitus", "diabetes", "diabetes complications "and "multi-morbidities (MeSH) "with interposition of the Boolean operator "AND". RESULTS After removal of duplicities and following exclusion criteria, 11 articles were selected. Our results showed that the patients of CHIK with DM had more severe and prolonged symptoms of CHIK and more frequently required hospitalization. No study investigated the biological process to explain how hyperglycemic state worsened the clinical manifestations of Chikungunya in diabetic patients. CONCLUSION An important association between DM and the severity of CHIK is observed. Prospective and more rigorous controlled studies are required to generate evidence that might y elucidate the causes of this relationship. Given the fast expanding viral infection of Chikungunya in Central and South America, Asia and Africa in recent years in the context of exponential increase in diabetes globally, the issue deserves global attention.
Collapse
Affiliation(s)
| | - Renan Magalhães Montenegro
- Federal University of Ceará (UFC), Rua Professor Costa Mendes, 1608, Rodolfo Teófilo, Fortaleza, CE 60416-200 Brazil
| | - Virginia Oliveira Fernandes
- Federal University of Ceará (UFC), Rua Professor Costa Mendes, 1608, Rodolfo Teófilo, Fortaleza, CE 60416-200 Brazil
| | - Rhaquel Oliveira
- Federal University of Ceará (UFC), Rua Professor Costa Mendes, 1608, Rodolfo Teófilo, Fortaleza, CE 60416-200 Brazil
| | - Lívia Aline de Araújo Batista
- Federal University of Ceará (UFC), Rua Professor Costa Mendes, 1608, Rodolfo Teófilo, Fortaleza, CE 60416-200 Brazil
| | - Akhtar Hussain
- Federal University of Ceará (UFC), Rua Professor Costa Mendes, 1608, Rodolfo Teófilo, Fortaleza, CE 60416-200 Brazil
- Department of International Health, Institute of Health and Society, Faculty of Medicine, University of Oslo, P.O. Box 1130, Blindern, N-0317 Oslo, Norway
| | | |
Collapse
|
16
|
Sengodan SK, Rajan A, Hemalatha SK, Nadhan R, Jaleel A, Srinivas P. Proteomic Profiling of β-hCG-Induced Spheres in BRCA1 Defective Triple Negative Breast Cancer Cells. J Proteome Res 2017; 17:276-289. [DOI: 10.1021/acs.jproteome.7b00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Satheesh Kumar Sengodan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Arathi Rajan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Sreelatha Krishnakumar Hemalatha
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Revathy Nadhan
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Abdul Jaleel
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| | - Priya Srinivas
- Cancer Research Program and ‡Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala 695 014, India
| |
Collapse
|
17
|
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
|
18
|
Vidotto A, Morais ATS, Ribeiro MR, Pacca CC, Terzian ACB, Gil LHVG, Mohana-Borges R, Gallay P, Nogueira ML. Systems Biology Reveals NS4B-Cyclophilin A Interaction: A New Target to Inhibit YFV Replication. J Proteome Res 2017; 16:1542-1555. [PMID: 28317380 DOI: 10.1021/acs.jproteome.6b00933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Yellow fever virus (YFV) replication is highly dependent on host cell factors. YFV NS4B is reported to be involved in viral replication and immune evasion. Here interactions between NS4B and human proteins were determined using a GST pull-down assay and analyzed using 1-DE and LC-MS/MS. We present a total of 207 proteins confirmed using Scaffold 3 Software. Cyclophilin A (CypA), a protein that has been shown to be necessary for the positive regulation of flavivirus replication, was identified as a possible NS4B partner. 59 proteins were found to be significantly increased when compared with a negative control, and CypA exhibited the greatest difference, with a 22-fold change. Fisher's exact test was significant for 58 proteins, and the p value of CypA was the most significant (0.000000019). The Ingenuity Systems software identified 16 pathways, and this analysis indicated sirolimus, an mTOR pathway inhibitor, as a potential inhibitor of CypA. Immunofluorescence and viral plaque assays showed a significant reduction in YFV replication using sirolimus and cyclosporine A (CsA) as inhibitors. Furthermore, YFV replication was strongly inhibited in cells treated with both inhibitors using reporter BHK-21-rep-YFV17D-LucNeoIres cells. Taken together, these data suggest that CypA-NS4B interaction regulates YFV replication. Finally, we present the first evidence that YFV inhibition may depend on NS4B-CypA interaction.
Collapse
Affiliation(s)
- Alessandra Vidotto
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Ana T S Morais
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Milene R Ribeiro
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Carolina C Pacca
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Ana C B Terzian
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Laura H V G Gil
- Departamento de Virologia, Centro de Pesquisa Aggeu Magalhães , Fundação Oswaldo Cruz (FIOCRUZ) - Recife, Pernambuco 50740-465, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro - UFRJ , Rio de Janeiro RJ 21941-902, Brazil
| | - Philippe Gallay
- Department of Immunology & Microbial Science, The Scripps Research Institute - La Jolla , San Diego, California 92037, United States
| | - Mauricio L Nogueira
- Laboratório de Virologia, Faculdade de Medicina de José do Rio Preto , São José do Rio Preto, São Paulo 15090-000, Brazil
| |
Collapse
|
19
|
Tambo E, Khayeka-Wandabwa C, Olalubi OA, Adedeji AA, Ngogang JY, Khater EI. Addressing knowledge gaps in molecular, sero-surveillance and monitoring approaches on Zika epidemics and other arbovirus co-infections: A structured review. Parasite Epidemiol Control 2017; 2:50-60. [PMID: 29774281 PMCID: PMC5952677 DOI: 10.1016/j.parepi.2017.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 01/29/2017] [Accepted: 01/29/2017] [Indexed: 01/02/2023] Open
Abstract
Globalization, with consequent increased travel and trade, rapid urbanization and growing weather variation events due to climate change has contributed to the recent unprecedented Zika virus (ZIKV) pandemic. This has emphasized the pressing need for local, national, regional and global community collaborative proactiveness, leadership and financial investment resilience in research and development. This paper addresses the potential knowledge gaps and impact of early detection and monitoring approaches on ZIKV epidemics and related arboviral infections steered towards effective prevention and smart response strategies. We advocate for the development and validation of robust field and point of care diagnostic tools that are more sensitive, specific and cost effective for use in ZIKV epidemics and routine pathophysiology surveillance and monitoring systems as an imperative avenue in understanding Zika-related and other arbovirus trends and apply genomic and proteomic characterisation approaches in guiding annotation efforts in order to design and implement public health burden mitigation and adaptation strategies.
Collapse
Affiliation(s)
- Ernest Tambo
- Department Biochemistry, Higher Institute of Health Sciences, Universite des Montagnes, Bangangté, Cameroon.,Africa Disease Intelligence and Surveillance, Communication and Response (Africa DISCoR) Foundation, Yaoundé, Cameroon
| | - Christopher Khayeka-Wandabwa
- African Population and Health Research Center (APHRC), Nairobi, Kenya.,Health Sciences Platform, School of Pharmaceutical Science and Technology (SPST), Tianjin University, China
| | - Oluwasogo A Olalubi
- Department of Public Health, Kwara State University (KWASU), Malete, Nigeria
| | - Ahmed A Adedeji
- Department of Pharmacology, Faculty of Health Sciences, Habib Medical School, Islamic University, Kibuuli, Kampala, Uganda
| | - Jeanne Y Ngogang
- Department Biochemistry, Higher Institute of Health Sciences, Universite des Montagnes, Bangangté, Cameroon.,Service de Biochimie, Centre Hospitalier Universitaire (CHU), Yaoundé, Cameroon
| | - Emad Im Khater
- Medical Entomology Department, Faculty of Science, Ain Shams University, Cairo, Egypt.,Public Health Pests Laboratory of Jeddah Governorate, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Jean-Baptiste E, von Oettingen J, Larco P, Raphaël F, Larco NC, Cauvin MM, Charles R. Chikungunya Virus Infection and Diabetes Mellitus: A Double Negative Impact. Am J Trop Med Hyg 2016; 95:1345-1350. [PMID: 27729569 PMCID: PMC5154448 DOI: 10.4269/ajtmh.16-0320] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 09/06/2016] [Indexed: 12/16/2022] Open
Abstract
The impact of chikungunya virus (CHIKV) infection on diabetic patients (DPs) has not been described. We aimed to compare clinical features of CHIKV infection in DPs and nondiabetic patients (NDPs), and to evaluate its effects on glycemic control among DPs. We recorded clinical information and, in DPs, glycemic control. Forty-six DPs and 53 NDPs aged ≥ 20 years living in Haiti, with acute CHIKV infection, were studied. Diabetes duration was 7.1 ± 6.1 years. The most common acute CHIKV clinical manifestations were arthralgia (100.0% DPs and 98.1% NDPs, P = 1.000) and fever (86.9% DPs and 90.5% NDPs, P = 0.750). In DPs as compared with NDPs, arthralgia was more intense (mean pain score of 6.0/10 ± 2.2 versus 5.1/10 ± 2.0, P = 0.04) and took longer to improve (8.2 ± 3.0 versus 3.5 ± 2.5 days, P < 0.0001). Severe arthralgia was more prevalent (58.7% versus 20.8%, P = 0.0002), as was myalgia (80.4% versus 50.9%, P = 0.003), and fever lasted longer (5.1 ± 1.8 versus 3.7 ± 1.7 days, P = 0.0002). Among DPs, median fasting capillary glucose before versus after disease onset was 132.5 and 167.5 mg/dL (P < 0.001), corresponding to a median increase of 26.8% (interquartile range: 14.4-50.1%). Antidiabetic medication was titrated up in 41.3%. In summary, among DPs, CHIKV infection has a significant negative impact on glycemic control and, compared with NDPs, results in greater morbidity. Close clinical and glycemic observation is recommended in DPs with CHIKV infection.
Collapse
Affiliation(s)
- Eddy Jean-Baptiste
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| | - Julia von Oettingen
- Division of Pediatric Endocrinology, Montreal Children's Hospital, Montreal, Canada
| | - Philippe Larco
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| | - Frédérica Raphaël
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| | - Nancy Charles Larco
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| | - Marie Marcelle Cauvin
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| | - René Charles
- FHADIMAC (Haitian Foundation of Diabetes and Cardiovascular Diseases), Port-au-Prince, Haiti
| |
Collapse
|
21
|
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]
|
22
|
Biophysical Characterization of Nucleophosmin Interactions with Human Immunodeficiency Virus Rev and Herpes Simplex Virus US11. PLoS One 2015; 10:e0143634. [PMID: 26624888 PMCID: PMC4704560 DOI: 10.1371/journal.pone.0143634] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/06/2015] [Indexed: 02/07/2023] Open
Abstract
Nucleophosmin (NPM1, also known as B23, numatrin or NO38) is a pentameric RNA-binding protein with RNA and protein chaperon functions. NPM1 has increasingly emerged as a potential cellular factor that directly associates with viral proteins; however, the significance of these interactions in each case is still not clear. In this study, we have investigated the physical interaction of NPM1 with both human immunodeficiency virus type 1 (HIV-1) Rev and Herpes Simplex virus type 1 (HSV-1) US11, two functionally homologous proteins. Both viral proteins show, in mechanistically different modes, high affinity for a binding site on the N-terminal oligomerization domain of NPM1. Rev, additionally, exhibits low-affinity for the central histone-binding domain of NPM1. We also showed that the proapoptotic cyclic peptide CIGB-300 specifically binds to NPM1 oligomerization domain and blocks its association with Rev and US11. Moreover, HIV-1 virus production was significantly reduced in the cells treated with CIGB-300. Results of this study suggest that targeting NPM1 may represent a useful approach for antiviral intervention.
Collapse
|