1
|
Fakfum P, Chuljerm H, Parklak W, Roytrakul S, Phaonakrop N, Lerttrakarnnon P, Kulprachakarn K. Plasma Proteomics of Type 2 Diabetes, Hypertension, and Co-Existing Diabetes/Hypertension in Thai Adults. Life (Basel) 2024; 14:1269. [PMID: 39459569 PMCID: PMC11509282 DOI: 10.3390/life14101269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/31/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The study explored proteomics to better understand the relationship between type 2 diabetes (T2DM) and hypertension (HT) in Thai adults, using shotgun proteomics and bioinformatics analysis. Plasma samples were taken from 61 subjects: 14 healthy subjects (mean age = 40.85 ± 7.12), 13 with T2DM (mean age = 57.38 ± 6.03), 16 with HT (mean age = 66.87 ± 10.09), and 18 with coexisting T2DM/HT (mean age = 58.22 ± 10.65). Proteins were identified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Protein-protein interactions were analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) version 11.5. We identified six unique proteins in T2DM patients, including translationally controlled 1 (TPT1) and nibrin (NBN), which are associated with the DNA damage response. In HT patients, seven unique proteins were identified, among them long-chain fatty acid-CoA ligase (ASCL), which functions in the stimulation of triacylglycerol and cholesterol synthesis, and NADPH oxidase activator 1 (NOXA1), which is involved in high blood pressure via angiotensin II-induced reactive oxygen species (ROS)-generating systems. In coexisting T2DM/HT patients, six unique proteins were identified, of which two-microtubule-associated protein 1A (MAP1A)-might be involved in dementia via RhoB-p53 and diacylglycerol kinase beta (DGKB), associated with lipid metabolism. This study identified new candidate proteins that are possibly involved in the pathology of these diseases.
Collapse
Affiliation(s)
- Puriwat Fakfum
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Hataichanok Chuljerm
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Wason Parklak
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Narumon Phaonakrop
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Peerasak Lerttrakarnnon
- Aging and Aging Palliative Care Research Cluster, Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokwan Kulprachakarn
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| |
Collapse
|
2
|
Treffers EE, Tas A, Scholte FEM, de Ru AH, Snijder EJ, van Veelen PA, van Hemert MJ. The alphavirus nonstructural protein 2 NTPase induces a host translational shut-off through phosphorylation of eEF2 via cAMP-PKA-eEF2K signaling. PLoS Pathog 2023; 19:e1011179. [PMID: 36848386 PMCID: PMC9997916 DOI: 10.1371/journal.ppat.1011179] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/09/2023] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging alphavirus. Since 2005, it has infected millions of people during outbreaks in Africa, Asia, and South/Central America. CHIKV replication depends on host cell factors at many levels and is expected to have a profound effect on cellular physiology. To obtain more insight into host responses to infection, stable isotope labeling with amino acids in cell culture and liquid chromatography-tandem mass spectrometry were used to assess temporal changes in the cellular phosphoproteome during CHIKV infection. Among the ~3,000 unique phosphorylation sites analyzed, the largest change in phosphorylation status was measured on residue T56 of eukaryotic elongation factor 2 (eEF2), which showed a >50-fold increase at 8 and 12 h p.i. Infection with other alphaviruses (Semliki Forest, Sindbis and Venezuelan equine encephalitis virus (VEEV)) triggered a similarly strong eEF2 phosphorylation. Expression of a truncated form of CHIKV or VEEV nsP2, containing only the N-terminal and NTPase/helicase domains (nsP2-NTD-Hel), sufficed to induce eEF2 phosphorylation, which could be prevented by mutating key residues in the Walker A and B motifs of the NTPase domain. Alphavirus infection or expression of nsP2-NTD-Hel resulted in decreased cellular ATP levels and increased cAMP levels. This did not occur when catalytically inactive NTPase mutants were expressed. The wild-type nsP2-NTD-Hel inhibited cellular translation independent of the C-terminal nsP2 domain, which was previously implicated in directing the virus-induced host shut-off for Old World alphaviruses. We hypothesize that the alphavirus NTPase activates a cellular adenylyl cyclase resulting in increased cAMP levels, thus activating PKA and subsequently eukaryotic elongation factor 2 kinase. This in turn triggers eEF2 phosphorylation and translational inhibition. We conclude that the nsP2-driven increase of cAMP levels contributes to the alphavirus-induced shut-off of cellular protein synthesis that is shared between Old and New World alphaviruses. MS Data are available via ProteomeXchange with identifier PXD009381.
Collapse
Affiliation(s)
- Emmely E. Treffers
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ali Tas
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Florine E. M. Scholte
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric J. Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. van Veelen
- Center for Proteomics & Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J. van Hemert
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
3
|
Singh A, Ansari VA, Mahmood T, Ahsan F, Wasim R. Neurodegeneration: Microglia: Nf-Kappab Signaling Pathways. Drug Res (Stuttg) 2022; 72:496-499. [PMID: 36055286 DOI: 10.1055/a-1915-4861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Microglia is cells of mesodermal/mesenchymal origin that migrate into the central nervous system (CNS) to form resident macrophages inside the special brain microenvironment. Intact with both neuronal and non-neuronal cells, microglia is highly active cells. Continuous process extension and retraction allows microglia to scan the brain parenchyma for threats. They are also able to change their morphology from ramified to amoeboid, which is a sign of cell activity. In response to pleiotropic stimuli such as neurotransmitters, cytokines, and plasma proteins, microglia express a diverse range of receptors. As controllers of synaptic activities and phagocytosis of developing neurons, they serve a critical role in the healthy brain and have significant effects on synaptic plasticity and adult neurogenesis. A frequent cause of hypoparathyroidism is a mutation in the gene glial cells missing-2 (GCM2). Neonatal hypoparathyroidism has an amorphic recessive GCM2 mutation, while autosomal dominant hypoparathyroidism has a dominant-negative GCM2 mutation. Curiously, familial isolated hyperparathyroidism has been associated with activating GCM2 mutation. In addition to seizures, neurocognitive impairment, carpopedal spasm, tingling and numbness are common clinical manifestations of hypoparathyroidism. Biogenic amines are a group of four neurotransmitters that belong to that category and these include serotonin, dopamine, norepinephrine, and epinephrine. Numerous antidepressants prevent the reuptake from occurring the brain-gut axis is hardwired through the CNS, enteric nervous system (ENS), neuroendocrine linkages and highly innervated nerve plexuses.
Collapse
Affiliation(s)
- Aditya Singh
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow (India)
| | - Vaseem Ahamad Ansari
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow (India)
| | - Tarique Mahmood
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow (India)
| | - Farogh Ahsan
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow (India)
| | - Rufaida Wasim
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow (India)
| |
Collapse
|
4
|
Traverse EM, Millsapps EM, Underwood EC, Hopkins HK, Young M, Barr KL. Chikungunya Immunopathology as It Presents in Different Organ Systems. Viruses 2022; 14:v14081786. [PMID: 36016408 PMCID: PMC9414582 DOI: 10.3390/v14081786] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 12/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is currently an urgent public health problem as high morbidity from the virus leaves populations with negative physical, social, and economic impacts. CHIKV has the potential to affect every organ of an individual, leaving patients with lifelong impairments which negatively affect their quality of life. In this review, we show the importance of CHIKV in research and public health by demonstrating the immunopathology of CHIKV as it presents in different organ systems. Papers used in this review were found on PubMed, using “chikungunya and [relevant organ system]”. There is a significant inflammatory response during CHIKV infection which affects several organ systems, such as the brain, heart, lungs, kidneys, skin, and joints, and the immune response to CHIKV in each organ system is unique. Whilst there is clinical evidence to suggest that serious complications can occur, there is ultimately a lack of understanding of how CHIKV can affect different organ systems. It is important for clinicians to understand the risks to their patients.
Collapse
|
5
|
Qadri SW, Kumar N, Santhoshkumar R, Desai A, Ravi V, Venkataswamy MM. Infection of human microglial cell line CHME-3 to study neuropathogenesis of chikungunya virus. J Neurovirol 2022; 28:374-382. [DOI: 10.1007/s13365-022-01070-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/02/2022] [Accepted: 03/03/2022] [Indexed: 02/02/2023]
|
6
|
Mamidi P, Nayak TK, Kumar A, Kumar S, Chatterjee S, De S, Datey A, Ghosh S, Keshry SS, Singh S, Laha E, Ray A, Chattopadhyay S, Chattopadhyay S. MK2a inhibitor CMPD1 abrogates chikungunya virus infection by modulating actin remodeling pathway. PLoS Pathog 2021; 17:e1009667. [PMID: 34780576 PMCID: PMC8592423 DOI: 10.1371/journal.ppat.1009667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) epidemics around the world have created public health concern with the unavailability of effective drugs and vaccines. This emphasizes the need for molecular understanding of host-virus interactions for developing effective targeted antivirals. Microarray analysis was carried out using CHIKV strain (Prototype and Indian) infected Vero cells and two host isozymes, MAPK activated protein kinase 2 (MK2) and MAPK activated protein kinase 3 (MK3) were selected for further analysis. The substrate spectrum of both enzymes is indistinguishable and covers proteins involved in cytokines production, endocytosis, reorganization of the cytoskeleton, cell migration, cell cycle control, chromatin remodeling and transcriptional regulation. Gene silencing and drug treatment were performed in vitro and in vivo to unravel the role of MK2/MK3 in CHIKV infection. Gene silencing of MK2 and MK3 abrogated around 58% CHIKV progeny release from the host cell and a MK2 activation inhibitor (CMPD1) treatment demonstrated 68% inhibition of viral infection suggesting a major role of MAPKAPKs during late CHIKV infection in vitro. Further, it was observed that the inhibition in viral infection is primarily due to the abrogation of lamellipodium formation through modulation of factors involved in the actin cytoskeleton remodeling pathway. Moreover, CHIKV-infected C57BL/6 mice demonstrated reduction in the viral copy number, lessened disease score and better survivability after CMPD1 treatment. In addition, reduction in expression of key pro-inflammatory mediators such as CXCL13, RAGE, FGF, MMP9 and increase in HGF (a CHIKV infection recovery marker) was observed indicating the effectiveness of the drug against CHIKV. Taken together it can be proposed that MK2 and MK3 are crucial host factors for CHIKV infection and can be considered as important target for developing effective anti-CHIKV strategies. Chikungunya virus has been a dreaded disease from the first time it occurred in 1952 Tanzania. Since then it has been affecting the different parts of the world at different time periods in large scale. It is typically transmitted to humans by bites of Aedes aegypti and Aedes albopictus mosquitoes. Although, studies have been undertaken to combat its prevalence still there are no effective strategies like vaccines or antivirals against it. Therefore it is essential to understand the virus and host interaction to overcome this hurdle. In this study two host factors MK2 and MK3 have been taken into consideration to see how they affect the multiplication of the virus. The in vitro and in vivo experiments conducted demonstrated that inhibition of MK2 and MK3 not only restricted viral release but also decreased the disease score and allowed better survivability. Therefore, MK2 and MK3 could be considered as the key targets in the anti CHIKV approach.
Collapse
Affiliation(s)
| | - Tapas Kumar Nayak
- National Institute of Science Education and Research, Bhubaneswar, India
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Abhishek Kumar
- Institute of Life Sciences, Bhubaneswar, India
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, United States of America
| | - Sameer Kumar
- Institute of Life Sciences, Bhubaneswar, India
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Sanchari Chatterjee
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Saikat De
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Ankita Datey
- Institute of Life Sciences, Bhubaneswar, India
- KIIT school of Biotechnology, Bhubaneswar, India
| | - Soumyajit Ghosh
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Supriya Suman Keshry
- Institute of Life Sciences, Bhubaneswar, India
- KIIT school of Biotechnology, Bhubaneswar, India
| | - Sharad Singh
- Institute of Life Sciences, Bhubaneswar, India
- KIIT school of Biotechnology, Bhubaneswar, India
| | - Eshna Laha
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Amrita Ray
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | | | | |
Collapse
|
7
|
Constant LEC, Rajsfus BF, Carneiro PH, Sisnande T, Mohana-Borges R, Allonso D. Overview on Chikungunya Virus Infection: From Epidemiology to State-of-the-Art Experimental Models. Front Microbiol 2021; 12:744164. [PMID: 34675908 PMCID: PMC8524093 DOI: 10.3389/fmicb.2021.744164] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/27/2022] Open
Abstract
Chikungunya virus (CHIKV) is currently one of the most relevant arboviruses to public health. It is a member of the Togaviridae family and alphavirus genus and causes an arthritogenic disease known as chikungunya fever (CHIKF). It is characterized by a multifaceted disease, which is distinguished from other arbovirus infections by the intense and debilitating arthralgia that can last for months or years in some individuals. Despite the great social and economic burden caused by CHIKV infection, there is no vaccine or specific antiviral drugs currently available. Recent outbreaks have shown a change in the severity profile of the disease in which atypical and severe manifestation lead to hundreds of deaths, reinforcing the necessity to understand the replication and pathogenesis processes. CHIKF is a complex disease resultant from the infection of a plethora of cell types. Although there are several in vivo models for studying CHIKV infection, none of them reproduces integrally the disease signature observed in humans, which is a challenge for vaccine and drug development. Therefore, understanding the potentials and limitations of the state-of-the-art experimental models is imperative to advance in the field. In this context, the present review outlines the present knowledge on CHIKV epidemiology, replication, pathogenesis, and immunity and also brings a critical perspective on the current in vitro and in vivo state-of-the-art experimental models of CHIKF.
Collapse
Affiliation(s)
- Larissa E. C. Constant
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bia F. Rajsfus
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro H. Carneiro
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Kril V, Aïqui-Reboul-Paviet O, Briant L, Amara A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu Rev Virol 2021; 8:327-347. [PMID: 34255544 DOI: 10.1146/annurev-virology-091919-102021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus responsible for major outbreaks of disease since 2004 in the Indian Ocean islands, South east Asia, and the Americas. CHIKV causes debilitating musculoskeletal disorders in humans that are characterized by fever, rash, polyarthralgia, and myalgia. The disease is often self-limiting and nonlethal; however, some patients experience atypical or severe clinical manifestations, as well as a chronic rheumatic syndrome. Unfortunately, no efficient antivirals against CHIKV infection are available so far, highlighting the importance of deepening our knowledge of CHIKV host cell interactions and viral replication strategies. In this review, we discuss recent breakthroughs in the molecular mechanisms that regulate CHIKV infection and lay down the foundations to understand viral pathogenesis. We describe the role of the recently identified host factors co-opted by the virus for infection and pathogenesis, and emphasize the importance of CHIKV nonstructural proteins in both replication complex assembly and host immune response evasion. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Vasiliya Kril
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| | - Olivier Aïqui-Reboul-Paviet
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Laurence Briant
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Ali Amara
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| |
Collapse
|
9
|
Shukla A, Bandyopadhyay T, Vallamkonda N, Maria A. Long-Term Neurodevelopmental Outcomes of Neonatal Chikungunya: Follow-Up of a Series of Cases Till 1 Year. J Trop Pediatr 2021; 67:5881350. [PMID: 32756979 DOI: 10.1093/tropej/fmaa053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Short- or long-term neurodevelopmental outcomes of Neonatal Chikungunya infection haven't been well described. In this study, we describe neurodevelopmental outcomes of a series of 13 infants. METHODS Study was conducted over a period of 1 year in high-risk follow-up clinic. Follow-up visits were conducted at 3, 6, 9, 12 ± 3 months. PRIMARY OUTCOME Development Quotient by Developmental Assessment Scales for Indian Infants. Secondary outcome: growth/anthropometric assessment, neuromotor/neurosensory and re-hospitalization rate. RESULTS DASII was <85 in three infants. Two other had expressive speech delay. Two, three and four babies had head circumference, length and weight below third centile, respectively. Two infants had persistent hypertonia and one had hypotonia. Two infants developed strabismus. Two infants had seizures persisting at 12 months requiring re-hospitalization. CONCLUSION Neonatal Chikungunya infection portends a poor neurodevelopmental outcome at 1 year of age. Careful neurodevelopmental follow-up is required for identification and management of sequelae.
Collapse
Affiliation(s)
- Amlin Shukla
- Department of Neonatology, ABVIMS and Dr RML Hospital, New Delhi 110001, India
| | - Tapas Bandyopadhyay
- Department of Neonatology, ABVIMS and Dr RML Hospital, New Delhi 110001, India
| | | | - Arti Maria
- Department of Neonatology, ABVIMS and Dr RML Hospital, New Delhi 110001, India
| |
Collapse
|
10
|
Alves-Leon SV, Ferreira CDS, Herlinger AL, Fontes-Dantas FL, Rueda-Lopes FC, Francisco RDS, Gonçalves JPDC, de Araújo AD, Rêgo CCDS, Higa LM, Gerber AL, Guimarães APDC, de Menezes MT, de Paula Tôrres MC, Maia RA, Nogueira BMG, França LC, da Silva MM, Naurath C, Correia ASDS, Vasconcelos CCF, Tanuri A, Ferreira OC, Cardoso CC, Aguiar RS, de Vasconcelos ATR. Exome-Wide Search for Genes Associated With Central Nervous System Inflammatory Demyelinating Diseases Following CHIKV Infection: The Tip of the Iceberg. Front Genet 2021; 12:639364. [PMID: 33815474 PMCID: PMC8010313 DOI: 10.3389/fgene.2021.639364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/08/2021] [Indexed: 12/31/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emergent arbovirus that causes a disease characterized primarily by fever, rash and severe persistent polyarthralgia, although <1% of cases develop severe neurological manifestations such as inflammatory demyelinating diseases (IDD) of the central nervous system (CNS) like acute disseminated encephalomyelitis (ADEM) and extensive transverse myelitis. Genetic factors associated with host response and disease severity are still poorly understood. In this study, we performed whole-exome sequencing (WES) to identify HLA alleles, genes and cellular pathways associated with CNS IDD clinical phenotype outcomes following CHIKV infection. The cohort includes 345 patients of which 160 were confirmed for CHIKV. Six cases presented neurological manifestation mimetizing CNS IDD. WES data analysis was performed for 12 patients, including the CNS IDD cases and 6 CHIKV patients without any neurological manifestation. We identified 29 candidate genes harboring rare, pathogenic, or probably pathogenic variants in all exomes analyzed. HLA alleles were also determined and patients who developed CNS IDD shared a common signature with diseases such as Multiple sclerosis (MS) and Neuromyelitis Optica Spectrum Disorders (NMOSD). When these genes were included in Gene Ontology analyses, pathways associated with CNS IDD syndromes were retrieved, suggesting that CHIKV-induced CNS outcomesmay share a genetic background with other neurological disorders. To our knowledge, this study was the first genome-wide investigation of genetic risk factors for CNS phenotypes in CHIKV infection. Our data suggest that HLA-DRB1 alleles associated with demyelinating diseases may also confer risk of CNS IDD outcomes in patients with CHIKV infection.
Collapse
Affiliation(s)
- Soniza Vieira Alves-Leon
- Translational Neuroscience Laboratory, Rio de Janeiro State Federal University, Rio de Janeiro, Brazil
- Department of Neurology/Reference and Research Center for Multiple Sclerosis and Other Central Nervous System Idiopathic Demyelinating Inflammatory Diseases, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | - João Paulo da Costa Gonçalves
- Translational Neuroscience Laboratory, Rio de Janeiro State Federal University, Rio de Janeiro, Brazil
- Department of Neurology/Reference and Research Center for Multiple Sclerosis and Other Central Nervous System Idiopathic Demyelinating Inflammatory Diseases, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amanda Dutra de Araújo
- Translational Neuroscience Laboratory, Rio de Janeiro State Federal University, Rio de Janeiro, Brazil
- Department of Neurology/Reference and Research Center for Multiple Sclerosis and Other Central Nervous System Idiopathic Demyelinating Inflammatory Diseases, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cláudia Cecília da Silva Rêgo
- Translational Neuroscience Laboratory, Rio de Janeiro State Federal University, Rio de Janeiro, Brazil
- Department of Neurology/Reference and Research Center for Multiple Sclerosis and Other Central Nervous System Idiopathic Demyelinating Inflammatory Diseases, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza Mendonça Higa
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Richard Araújo Maia
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Laise Carolina França
- Translational Neuroscience Laboratory, Rio de Janeiro State Federal University, Rio de Janeiro, Brazil
| | - Marcos Martins da Silva
- Department of Clinical Medicine, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christian Naurath
- Federal Hospital Cardoso Fontes, Ministry of Health, Rio de Janeiro, Brazil
| | | | | | - Amilcar Tanuri
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Orlando Costa Ferreira
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Renato Santana Aguiar
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
11
|
Islam MS, Khan MAAK. Computational analysis revealed miRNAs produced by Chikungunya virus target genes associated with antiviral immune responses and cell cycle regulation. Comput Biol Chem 2021; 92:107462. [PMID: 33640797 DOI: 10.1016/j.compbiolchem.2021.107462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/11/2021] [Accepted: 02/17/2021] [Indexed: 11/18/2022]
Abstract
Chikungunya virus (CHIKV) that causes chikungunya fever, is an alphavirus that belongs to the Togaviridae family containing a single-stranded RNA genome. Mosquitoes of the Aedes species act as the vectors for this virus and can be found in the blood, which can be passed from an infected person to a mosquito through mosquito bites. CHIKV has drawn much attention recently because of its potential of causing an epidemic. As the detailed mechanism of its pathogenesis inside the host system is still lacking, in this in silico research we have hypothesized that CHIKV might create miRNAs, which would target the genes associated with host cellular regulatory pathways, thereby providing the virus with prolonged refuge. Using bioinformatics approaches we found several putative miRNAs produced by CHIKV. Then we predicted the genes of the host targeted by these miRNAs. Functional enrichment analysis of these targeted genes shows the involvement of several biological pathways regulating antiviral immune stimulation, cellular proliferation, and cell cycle, thereby provide themselves with prolonged refuge and facilitate their pathogenesis, which in turn may lead to disease conditions. Finally, we analyzed a publicly available microarray dataset (GSE49985) to determine the altered expression levels of the targeted genes and found genes associated with pathways such as cell differentiation, phagocytosis, T-cell activation, response to cytokine, autophagy, Toll-like receptor signaling, RIG-I like receptor signaling and apoptosis. Our finding presents novel miRNAs and their targeted genes, which upon experimental validation could facilitate in developing new therapeutics to combat CHIKV infection and minimize CHIKV mediated diseases.
Collapse
Affiliation(s)
- Md Sajedul Islam
- Department of Biochemistry & Biotechnology, University of Barishal, Barishal, 8254, Bangladesh.
| | | |
Collapse
|
12
|
Ves-Urai P, Krobthong S, Thongsuk K, Roytrakul S, Yokthongwattana C. Comparative secretome analysis between salinity-tolerant and control Chlamydomonas reinhardtii strains. PLANTA 2021; 253:68. [PMID: 33594587 DOI: 10.1007/s00425-021-03583-7] [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: 07/05/2020] [Accepted: 01/30/2021] [Indexed: 06/12/2023]
Abstract
Secretome analysis of a salt-tolerant and control Chlamydomonas reinhardtii revealed 514 differentially expressed proteins. Membrane transport and trafficking, signal transduction and channel proteins were up-regulated in the ST secretome. Salinity is a major abiotic stress that limits crop production worldwide. Multiple adverse effects have been reported in many living organisms exposed to high-saline concentrations. Chlamydomonas reinhardtii is known for secreting proteins in response to many environmental stresses. A salinity-tolerant (ST) strain of Chlamydomonas has been developed, whose cells were able to grow at 300 mM NaCl. The current study analyzed the secretomes of ST grown in TAP medium supplemented with 300 mM NaCl and the laboratory strain CC-503 grown in TAP medium without NaCl supplement. In total, 514 secreted proteins were identified of which 203 were up-regulated and 110 were down-regulated. Bioinformatic analysis predicted 168 proteins to be secreted or in the conventional secretory pathway. Out of these, 70 were up-regulated, while 51 proteins were down-regulated. Proteins involved in membrane transport and trafficking, signal transduction and channel proteins were altered in their expression in the ST secretome, suggesting the response of saline stress acts toward not only the intracellular pool of proteins but also the extracellular proteins. This also suggested that the secreted proteins might have roles in the extracellular space. Signal peptide (SP) prediction revealed that almost 40% of the predicted secreted proteins contained a signal peptide; however, a high proportion of proteins lacked an SP, suggesting that these proteins might be secreted through an unconventional protein secretion pathway.
Collapse
Affiliation(s)
- Parthompong Ves-Urai
- Interdisciplinary Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
| | - Sucheewin Krobthong
- National Center for Genetic Engineering and Biotechnology, 113 Thailand Science Park, Phahonyothin Rd., Pathumthani, 12120, Thailand
| | - Karnpitcha Thongsuk
- Department of Biochemistry, Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Bangkok, 10900, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, 113 Thailand Science Park, Phahonyothin Rd., Pathumthani, 12120, Thailand
| | - Chotika Yokthongwattana
- Department of Biochemistry, Faculty of Science, Kasetsart University, 50 Ngamwongwan Rd., Bangkok, 10900, Thailand.
- Omics Center for Agriculture, Bioresources, Food and Health, Kasetsart University (OmiKU), Kasetsart University, 50 Ngam Wong Wan Road, Chatuchak, Bangkok, 10900, Thailand.
| |
Collapse
|
13
|
Current Understanding of the Role of Cholesterol in the Life Cycle of Alphaviruses. Viruses 2020; 13:v13010035. [PMID: 33383613 PMCID: PMC7823518 DOI: 10.3390/v13010035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/28/2022] Open
Abstract
Enveloped viruses rely on different lipid classes present in cell membranes to accomplish several steps of their life cycle in the host. Particularly for alphaviruses, a medically important group of arboviruses, which are part of the Togaviridae family, cholesterol seems to be a critical lipid exploited during infection, although its relevance may vary depending on which stage of the virus life cycle is under consideration and whether infection takes place in vertebrate or invertebrate hosts. In this review, the role of cholesterol in both early and late events of alphavirus infection and how viral replication may affect cholesterol metabolism are summarized, taking into account studies on Old World and New World alphaviruses in different cell lines. Moreover, the importance of cholesterol for the structural stability of alphavirus particles is also discussed, shedding light on the role played by this lipid when they leave the host cell.
Collapse
|
14
|
Fei Y, Han M, Chu X, Feng Z, Yu L, Luo Y, Lu L, Xu D. Transcriptomic and proteomic analyses reveal new insights into the regulation of immune pathways during cyprinid herpesvirus 2 infection in vitro. FISH & SHELLFISH IMMUNOLOGY 2020; 106:167-180. [PMID: 32717324 DOI: 10.1016/j.fsi.2020.07.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/17/2020] [Accepted: 07/21/2020] [Indexed: 06/11/2023]
Abstract
Carassius auratus gibelio is susceptible to the herpesviral hematopoietic necrosis (HVHN) disease caused by cyprinid herpesvirus 2 (CyHV-2) infection during the breeding process. Nevertheless, the report on biological response of CyHV-2 with C. auratus gibelio was limited, especially in vitro. In this study, host gene expression profiling was mostly analyzed in caudal fin cells of Carassius auratus gibelio (GiCF) underlying CyHV-2 infection. Transcriptomics and proteomics were employed to study the differential expression gene and revealed the host genes involved in pathway during the CyHV-2 infection. Transcriptome analysis revealed that compared with the control group, there were 11 335 and 19 421 differentially expressed unigenes at 48 h and at 96 h, respectively. Furthermore, proteome analysis showed that there were a total of 9008 proteins, among which 169 proteins were differential expression in the 48 h group and 502 proteins in the 96 h group. Notably, 10 and 158 differentially co-expressed genes at mRNA and protein levels (cDEGs) were reliably quantified at 48 h and 96 h, respectively. Interestingly, significantly different expressed genes both in the transcriptome and the proteome were identified, including GNG7, Hsp90a, THBS1 and RRM2. The result suggested that PI3k-AKT pathway was activated, but the p53 signaling pathway was suppressed. The above result will lay the foundation for understanding the mechanisms of host defense virus invasion during CyHV-2 infection.
Collapse
Affiliation(s)
- Yueyue Fei
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Minzhen Han
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Xin Chu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Zizhao Feng
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Lu Yu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Yang Luo
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China
| | - Liqun Lu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China; National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, PR China
| | - Dan Xu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, PR China; Key Laboratory of Aquaculture Ministry for Freshwater Aquatic Genetic Resources, Shanghai Ocean University, Shanghai, PR China; National Experimental Teaching Demonstration Center for Fishery Sciences, Shanghai Ocean University, Shanghai, PR China; Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, PR China.
| |
Collapse
|
15
|
Khodadadi E, Zeinalzadeh E, Taghizadeh S, Mehramouz B, Kamounah FS, Khodadadi E, Ganbarov K, Yousefi B, Bastami M, Kafil HS. Proteomic Applications in Antimicrobial Resistance and Clinical Microbiology Studies. Infect Drug Resist 2020; 13:1785-1806. [PMID: 32606829 PMCID: PMC7305820 DOI: 10.2147/idr.s238446] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/23/2020] [Indexed: 12/11/2022] Open
Abstract
Sequences of the genomes of all-important bacterial pathogens of man, plants, and animals have been completed. Still, it is not enough to achieve complete information of all the mechanisms controlling the biological processes of an organism. Along with all advances in different proteomics technologies, proteomics has completed our knowledge of biological processes all around the world. Proteomics is a valuable technique to explain the complement of proteins in any organism. One of the fields that has been notably benefited from other systems approaches is bacterial pathogenesis. An emerging field is to use proteomics to examine the infectious agents in terms of, among many, the response the host and pathogen to the infection process, which leads to a deeper knowledge of the mechanisms of bacterial virulence. This trend also enables us to identify quantitative measurements for proteins extracted from microorganisms. The present review study is an attempt to summarize a variety of different proteomic techniques and advances. The significant applications in bacterial pathogenesis studies are also covered. Moreover, the areas where proteomics may lead the future studies are introduced.
Collapse
Affiliation(s)
- Ehsaneh Khodadadi
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Zeinalzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Taghizadeh
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahareh Mehramouz
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fadhil S Kamounah
- Department of Chemistry, University of Copenhagen, Copenhagen, DK 2100, Denmark
| | - Ehsan Khodadadi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
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
|
17
|
Vairo F, Haider N, Kock R, Ntoumi F, Ippolito G, Zumla A. Chikungunya: Epidemiology, Pathogenesis, Clinical Features, Management, and Prevention. Infect Dis Clin North Am 2020; 33:1003-1025. [PMID: 31668189 DOI: 10.1016/j.idc.2019.08.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Chikungunya, a zoonotic disease caused by the Chikungunya virus (CHIKV), is transmitted by infected Aedes spp mosquitoes. CHIKV has now spread to more than 100 countries and is listed on the WHO Blueprint priority pathogens. After an incubation period of 1 to 12 days, symptoms similar to other febrile infections appear, with a sudden onset of high fever, nausea, polyarthralgia, myalgia, widespread skin rash, and conjunctivitis. Serious complications include myocarditis, uveitis, retinitis, hepatitis, acute renal disease, severe bullous lesions, meningoencephalitis, Guillain-Barré syndrome, myelitis, and cranial nerve palsies. Treatment is supportive; there is no specific antiviral treatment and no effective vaccine.
Collapse
Affiliation(s)
- Francesco Vairo
- National Institute for Infectious Diseases, "Lazzaro Spallanzani"Istituto di ricovero e cura a carattere scientifico - IRCCS, Via Portuense 292, 00149, Rome, Italy.
| | - Najmul Haider
- The Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - Richard Kock
- The Royal Veterinary College, University of London, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Médicale (FCRM), Brazzaville, Congo; Faculty of Sciences and Techniques, University Marien Ngouabi, PO Box 69, Brazzaville, Congo; Institute for Tropical Medicine, University of Tübingen, Wilhelmstraße 27 72074, Tübingen, Germany
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases, "Lazzaro Spallanzani"Istituto di ricovero e cura a carattere scientifico - IRCCS, Via Portuense 292, 00149, Rome, Italy
| | - Alimuddin Zumla
- Center for Clinical Microbiology, University College London, Royal Free Campus 2nd Floor, Rowland Hill Street, London NW3 2PF, United Kingdom
| |
Collapse
|
18
|
SAMHD1 Enhances Chikungunya and Zika Virus Replication in Human Skin Fibroblasts. Int J Mol Sci 2019; 20:ijms20071695. [PMID: 30959732 PMCID: PMC6480247 DOI: 10.3390/ijms20071695] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/23/2019] [Accepted: 04/03/2019] [Indexed: 01/19/2023] Open
Abstract
Chikungunya virus (CHIKV) and Zika virus (ZIKV) are emerging arboviruses that pose a worldwide threat to human health. Currently, neither vaccine nor antiviral treatment to control their infections is available. As the skin is a major viral entry site for arboviruses in the human host, we determined the global proteomic profile of CHIKV and ZIKV infections in human skin fibroblasts using Stable Isotope Labelling by Amino acids in Cell culture (SILAC)-based mass-spectrometry analysis. We show that the expression of the interferon-stimulated proteins MX1, IFIT1, IFIT3 and ISG15, as well as expression of defense response proteins DDX58, STAT1, OAS3, EIF2AK2 and SAMHD1 was significantly up-regulated in these cells upon infection with either virus. Exogenous expression of IFITs proteins markedly inhibited CHIKV and ZIKV replication which, accordingly, was restored following the abrogation of IFIT1 or IFIT3. Overexpression of SAMHD1 in cutaneous cells, or pretreatment of cells with the virus-like particles containing SAMHD1 restriction factor Vpx, resulted in a strong increase or inhibition, respectively, of both CHIKV and ZIKV replication. Moreover, silencing of SAMHD1 by specific SAMHD1-siRNA resulted in a marked decrease of viral RNA levels. Together, these results suggest that IFITs are involved in the restriction of replication of CHIKV and ZIKV and provide, as yet unreported, evidence for a proviral role of SAMHD1 in arbovirus infection of human skin cells.
Collapse
|
19
|
Comparative Proteome-Wide Analysis of Bone Marrow Microenvironment of β-Thalassemia/Hemoglobin E. Proteomes 2019; 7:proteomes7010008. [PMID: 30813444 PMCID: PMC6473223 DOI: 10.3390/proteomes7010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/16/2019] [Accepted: 02/19/2019] [Indexed: 12/17/2022] Open
Abstract
β-thalassemia/Hb E is a global health issue, which is characterized by a range of clinical symptoms from a mild and asymptomatic anemia to severe disorders that require transfusions from infancy. Pathological mechanisms of the disease involve the excess of unmatched alpha globin and iron overload, leading to ineffective erythropoiesis and ultimately to the premature death of erythroid precursors in bone marrow (BM) and peripheral organs. However, it is unclear as to how BM microenvironment factors contribute to the defective erythropoiesis in β-thalassemia/Hb E patients. Here, we employed mass spectrometry-based comparative proteomics to analyze BM plasma that was collected from six β-thalassemia/Hb E patients and four healthy donors. We identified that the differentially expressed proteins are enriched in secretory or exosome-associated proteins, many of which have putative functions in the oxidative stress response. Using Western blot assay, we confirmed that atypical lipoprotein, Apolipoprotein D (APOD), belonging to the Lipocalin transporter superfamily, was significantly decreased in BM plasma of the tested pediatric β-thalassemia/Hb E patients. Our results highlight that the disease condition of ineffective erythropoiesis and oxidative stress found in BM microenvironment of β-thalassemia/Hb E patients is associated with the impaired expression of APOD protein.
Collapse
|
20
|
Matusali G, Colavita F, Bordi L, Lalle E, Ippolito G, Capobianchi MR, Castilletti C. Tropism of the Chikungunya Virus. Viruses 2019; 11:v11020175. [PMID: 30791607 PMCID: PMC6410217 DOI: 10.3390/v11020175] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/16/2019] [Accepted: 02/17/2019] [Indexed: 12/12/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne virus that displays a large cell and organ tropism, and causes a broad range of clinical symptoms in humans. It is maintained in nature through both urban and sylvatic cycles, involving mosquito vectors and human or vertebrate animal hosts. Although CHIKV was first isolated in 1953, its pathogenesis was only more extensively studied after its re-emergence in 2004. The unexpected spread of CHIKV to novel tropical and non-tropical areas, in some instances driven by newly competent vectors, evidenced the vulnerability of new territories to this infectious agent and its associated diseases. The comprehension of the exact CHIKV target cells and organs, mechanisms of pathogenesis, and spectrum of both competitive vectors and animal hosts is pivotal for the design of effective therapeutic strategies, vector control measures, and eradication actions.
Collapse
Affiliation(s)
- Giulia Matusali
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Francesca Colavita
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Licia Bordi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Eleonora Lalle
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Maria R Capobianchi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Concetta Castilletti
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| |
Collapse
|
21
|
Dello Russo C, Cappoli N, Coletta I, Mezzogori D, Paciello F, Pozzoli G, Navarra P, Battaglia A. The human microglial HMC3 cell line: where do we stand? A systematic literature review. J Neuroinflammation 2018; 15:259. [PMID: 30200996 PMCID: PMC6131758 DOI: 10.1186/s12974-018-1288-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/21/2018] [Indexed: 02/08/2023] Open
Abstract
Microglia, unique myeloid cells residing in the brain parenchyma, represent the first line of immune defense within the central nervous system. In addition to their immune functions, microglial cells play an important role in other cerebral processes, including the regulation of synaptic architecture and neurogenesis. Chronic microglial activation is regarded as detrimental, and it is considered a pathogenic mechanism common to several neurological disorders. Microglial activation and function have been extensively studied in rodent experimental models, whereas the characterization of human cells has been limited due to the restricted availability of primary sources of human microglia. To overcome this problem, human immortalized microglial cell lines have been developed. The human microglial clone 3 cell line, HMC3, was established in 1995, through SV40-dependent immortalization of human embryonic microglial cells. It has been recently authenticated by the American Type Culture Collection (ATCC®) and distributed under the name of HMC3 (ATCC®CRL-3304). The HMC3 cells have been used in six research studies, two of which also indicated by ATCC® as reference articles. However, a more accurate literature revision suggests that clone 3 was initially distributed under the name of CHME3. In this regard, several studies have been published, thus contributing to a more extensive characterization of this cell line. Remarkably, the same cell line has been used in different laboratories with other denominations, i.e., CHME-5 cells and C13-NJ cells. In view of the fact that "being now authenticated by ATCC®" may imply a wider distribution of the cells, we aimed at reviewing data obtained with the human microglia cell line clone 3, making the readers aware of this complicated nomenclature. In addition, we also included original data, generated in our laboratory with the HMC3 (ATCC®CRL-3304) cells, providing information on the current state of the culture together with supplementary details on the culturing procedures to obtain and maintain viable cells.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy. .,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Natalia Cappoli
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy
| | - Isabella Coletta
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A., Rome, Italy
| | - Daniele Mezzogori
- Institute of Human Physiology, Università Cattolica del S. Cuore, Rome, Italy
| | - Fabiola Paciello
- Institute of Otolaryngology, Università Cattolica del S. Cuore, Rome, Italy
| | - Giacomo Pozzoli
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy.,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Pierluigi Navarra
- Institute of Pharmacology, Università Cattolica del S. Cuore, L.go F Vito 1, 00168, Rome, Italy.,Pharmacology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandra Battaglia
- Immunology Laboratory, Department of Oncological Gynecology, Università Cattolica del S. Cuore, Rome, Italy
| |
Collapse
|
22
|
Mutthi P, Theerawatanasirikul S, Roytrakul S, Paemanee A, Lekcharoensuk C, Hansoongnern P, Petcharat N, Thangthamniyom N, Lekcharoensuk P. Interferon gamma induces cellular protein alteration and increases replication of porcine circovirus type 2 in PK-15 cells. Arch Virol 2018; 163:2947-2957. [DOI: 10.1007/s00705-018-3944-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/28/2018] [Indexed: 01/01/2023]
|
23
|
Jirakkakul J, Roytrakul S, Srisuksam C, Swangmaneecharern P, Kittisenachai S, Jaresitthikunchai J, Punya J, Prommeenate P, Senachak J, So L, Tachaleat A, Tanticharoen M, Cheevadhanarak S, Wattanachaisaereekul S, Amnuaykanjanasin A. Culture degeneration in conidia of Beauveria bassiana and virulence determinants by proteomics. Fungal Biol 2017; 122:156-171. [PMID: 29458719 DOI: 10.1016/j.funbio.2017.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/04/2017] [Accepted: 12/15/2017] [Indexed: 01/18/2023]
Abstract
The quality of Beauveria bassiana conidia directly affects the virulence against insects. In this study, continuous subculturing of B. bassiana on both rice grains and potato dextrose agar (PDA) resulted in 55 and 49 % conidial yield reduction after 12 passages and 68 and 60 % virulence reduction after 20 and 12 passages at four d post-inoculation, respectively. The passage through Tenebrio molitor and Spodoptera exigua restored the virulence of rice and PDA subcultures, respectively. To explore the molecular mechanisms underlying the conidial quality and the decline of virulence after multiple subculturing, we investigated the conidial proteomic changes. Successive subculturing markedly increased the protein levels in oxidative stress response, autophagy, amino acid homeostasis, and apoptosis, but decreased the protein levels in DNA repair, ribosome biogenesis, energy metabolism, and virulence. The nitro blue tetrazolium assay verified that the late subculture's colony and conidia had a higher oxidative stress level than the early subculture. A 2A-type protein phosphatase and a Pleckstrin homology domain protein Slm1, effector proteins of the target of rapamycin (TOR) complex 1 and 2, respectively, were dramatically increased in the late subculture. These results suggest that TOR signalling might be associated with ageing in B. bassiana late subculture, in turn affecting its physiological characteristics and virulence.
Collapse
Affiliation(s)
- Jiraporn Jirakkakul
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkhuntien, Bangkok 10150, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Chettida Srisuksam
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Pratchya Swangmaneecharern
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Suthathip Kittisenachai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Janthima Jaresitthikunchai
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Juntira Punya
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Peerada Prommeenate
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Jittisak Senachak
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand
| | - Laihong So
- Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Kowloon, Hong Kong
| | - Anuwat Tachaleat
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkhuntien, Bangkok 10150, Thailand
| | - Morakot Tanticharoen
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkhuntien, Bangkok 10150, Thailand
| | - Supapon Cheevadhanarak
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkhuntien, Bangkok 10150, Thailand
| | - Songsak Wattanachaisaereekul
- Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (KMUTT), Bangkhuntien, Bangkok 10150, Thailand.
| | - Alongkorn Amnuaykanjanasin
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Paholyothin Road, Klong 1, Klong Luang, Pathumthani 12120, Thailand.
| |
Collapse
|
24
|
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
|
25
|
Jean Beltran PM, Federspiel JD, Sheng X, Cristea IM. Proteomics and integrative omic approaches for understanding host-pathogen interactions and infectious diseases. Mol Syst Biol 2017; 13:922. [PMID: 28348067 PMCID: PMC5371729 DOI: 10.15252/msb.20167062] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Organisms are constantly exposed to microbial pathogens in their environments. When a pathogen meets its host, a series of intricate intracellular interactions shape the outcome of the infection. The understanding of these host–pathogen interactions is crucial for the development of treatments and preventive measures against infectious diseases. Over the past decade, proteomic approaches have become prime contributors to the discovery and understanding of host–pathogen interactions that represent anti‐ and pro‐pathogenic cellular responses. Here, we review these proteomic methods and their application to studying viral and bacterial intracellular pathogens. We examine approaches for defining spatial and temporal host–pathogen protein interactions upon infection of a host cell. Further expanding the understanding of proteome organization during an infection, we discuss methods that characterize the regulation of host and pathogen proteomes through alterations in protein abundance, localization, and post‐translational modifications. Finally, we highlight bioinformatic tools available for analyzing such proteomic datasets, as well as novel strategies for integrating proteomics with other omic tools, such as genomics, transcriptomics, and metabolomics, to obtain a systems‐level understanding of infectious diseases.
Collapse
Affiliation(s)
- Pierre M Jean Beltran
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Joel D Federspiel
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Xinlei Sheng
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| |
Collapse
|
26
|
Le Sage V, Cinti A, Amorim R, Mouland AJ. Adapting the Stress Response: Viral Subversion of the mTOR Signaling Pathway. Viruses 2016; 8:v8060152. [PMID: 27231932 PMCID: PMC4926172 DOI: 10.3390/v8060152] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a central regulator of gene expression, translation and various metabolic processes. Multiple extracellular (growth factors) and intracellular (energy status) molecular signals as well as a variety of stressors are integrated into the mTOR pathway. Viral infection is a significant stress that can activate, reduce or even suppress the mTOR signaling pathway. Consequently, viruses have evolved a plethora of different mechanisms to attack and co-opt the mTOR pathway in order to make the host cell a hospitable environment for replication. A more comprehensive knowledge of different viral interactions may provide fruitful targets for new antiviral drugs.
Collapse
Affiliation(s)
- Valerie Le Sage
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
| | - Alessandro Cinti
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| | - Raquel Amorim
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| | - Andrew J Mouland
- HIV-1 RNA Trafficking Laboratory, Lady Davis Institute at the Jewish General Hospital, Montréal, QC H3T 1E2, Canada.
- Department of Medicine, McGill University, Montréal, QC H3A 0G4, Canada.
| |
Collapse
|
27
|
Schistosoma mekongi cathepsin B and its use in the development of an immunodiagnosis. Acta Trop 2016; 155:11-9. [PMID: 26655041 DOI: 10.1016/j.actatropica.2015.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/17/2015] [Accepted: 11/27/2015] [Indexed: 11/24/2022]
Abstract
Schistosomiasis mekongi is one of the most important human parasitic diseases caused by Schistosoma mekongi in South-east Asia. The endemic area is the Mekong River sub-region from Laos to Cambodia. This parasite also infects dogs and pigs which are its alternative host species. Currently, the lack of reliable rapid diagnosis makes it difficult to monitor the infection and spreading of the disease. In this study, we screened the antigens of the parasite with sera of infected mice using Western blotting and identified proteins of interest with LC-MS/MS to obtain potential candidate proteins for diagnostic development. This assay yielded 2 immunoreactive bands at molecular masses of 31 and 22kDa. The 31kDa protein was the major band identified as cathepsin B, and its gene was cloned to obtain a full cDNA sequence (SmekCatB). The cDNA consisted of 1123bp and its longest reading frame encoded for 342 amino acids with some putative post translation modifications. The recombinant SmekCatB (rSmekCatB) with hexahistidine tag at the C-terminus was expressed in Escherichia coli and purified by Ni-NTA resin under denaturing conditions. The rSmekCatB reacted with sera of S. mekongi-infected mice. Indirect ELISA using rSmekCatB as the antigen to detect mouse antibodies, revealed a sensitivity of 91.67% for schistosomiasis mekongi and the specificity of 100%. Our data suggested that SmekCatB is one of the most promising parasitic antigens that could be used for the diagnosis of S. mekongi infection.
Collapse
|
28
|
Paemanee A, Wikan N, Roytrakul S, Smith DR. Application of GelC-MS/MS to Proteomic Profiling of Chikungunya Virus Infection: Preparation of Peptides for Analysis. Methods Mol Biol 2016; 1426:179-93. [PMID: 27233271 DOI: 10.1007/978-1-4939-3618-2_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Gel-enhanced liquid chromatography coupled with tandem mass spectrometry (GeLC-MS/MS) is a labor intensive, but relatively straightforward methodology that generates high proteome coverage which can be applied to the proteome analysis of a range of starting materials such as cells or patient specimens. Sample proteins are resolved electrophoretically in one dimension through a sodium dodecyl sulfate (SDS) polyacrylamide gel after which the lanes are sliced into sections. The sections are further diced and the gel cubes generated are subjected to in-gel tryptic digestion. The resultant peptides can then be analyzed by tandem mass spectroscopy to identify the proteins by database searching. The methodology can routinely detect several thousand proteins in one analysis. The protocol we describe here has been used with both cells in culture that have been infected with chikungunya virus and specimens from Chikungunya fever patients. This protocol details the process for generating peptides for subsequent mass spectroscopic and bioinformatic analysis.
Collapse
Affiliation(s)
- Atchara Paemanee
- Institute of Molecular Biosciences, Mahidol University, Salaya Campus, 25/25 Phuttamontol Sai 4, Salaya, Nakorn Pathom, Thailand
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Nitwara Wikan
- Institute of Molecular Biosciences, Mahidol University, Salaya Campus, 25/25 Phuttamontol Sai 4, Salaya, Nakorn Pathom, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya Campus, 25/25 Phuttamontol Sai 4, Salaya, Nakorn Pathom, Thailand.
- Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
29
|
Deeba F, Islam A, Kazim SN, Naqvi IH, Broor S, Ahmed A, Parveen S. Chikungunya virus: recent advances in epidemiology, host pathogen interaction and vaccine strategies. Pathog Dis 2015; 74:ftv119. [PMID: 26657109 DOI: 10.1093/femspd/ftv119] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2015] [Indexed: 12/22/2022] Open
Abstract
The Chikungunya virus is a re-emerging alphavirus that belongs to the family Togaviridae. The symptoms include fever, rashes, nausea and joint pain that may last for months. The laboratory diagnosis of the infection is based on the serologic assays, virus isolation and molecular methods. The pathogenesis of the Chikungunya viral infection is not completely understood. Some of the recent investigations have provided information on replication of the virus in various cells and organs. In addition, some recent reports have indicated that the severity of the disease is correlated with the viral load and cytokines. The Chikungunya virus infection re-emerged as an explosive epidemic during 2004-09 affecting millions of people in the Indian Ocean. Subsequent global attention was given to research on this viral pathogen due to its broad area of geographical distribution during this epidemic. Chikungunya viral infection has become a challenge for the public health system because of the absence of a vaccine as well as antiviral drugs. A number of potential vaccine candidates have been tested on humans and animal models during clinical and preclinical trials. In this review, we mainly discuss the host-pathogen relationship, epidemiology and recent advances in the development of drugs and vaccines for the Chikungunya viral infection.
Collapse
Affiliation(s)
- Farah Deeba
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Syed Naqui Kazim
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | | | - Shobha Broor
- Department of Microbiology, SGT University, Gurgaon 122001, Haryana, India
| | - Anwar Ahmed
- Protein Research Chair, Department of Biochemistry, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shama Parveen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
30
|
Sphingosine kinase 2 is a chikungunya virus host factor co-localized with the viral replication complex. Emerg Microbes Infect 2015; 4:e61. [PMID: 26576339 PMCID: PMC4631929 DOI: 10.1038/emi.2015.61] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 07/14/2015] [Accepted: 07/27/2015] [Indexed: 02/04/2023]
Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus which causes severe and prolonged arthralgic febrile illness. The recent global spread of the virus and lack of approved therapeutic options makes it imperative to gain greater insight into the molecular mechanisms underlying CHIKV pathogenesis, in particular host factors recruited by the virus. In the current study, we identify sphingosine kinase 2 (SK2) as a CHIKV host factor co-localized with the viral replication complex (VRC) during infection. SK2 was demonstrated to co-localize with viral RNA and nonstructural proteins. Targeted impairment of SK2 expression or function significantly inhibited CHIKV infection. Furthermore, affinity purification-mass spectrometry studies revealed that SK2 associates with a number of proteins involved in cellular gene expression specifically during viral infection, suggesting a role in replication. Collectively these results identify SK2 as a novel CHIKV host factor.
Collapse
|
31
|
Joubert PE, Stapleford K, Guivel-Benhassine F, Vignuzzi M, Schwartz O, Albert ML. Inhibition of mTORC1 Enhances the Translation of Chikungunya Proteins via the Activation of the MnK/eIF4E Pathway. PLoS Pathog 2015; 11:e1005091. [PMID: 26317997 PMCID: PMC4552638 DOI: 10.1371/journal.ppat.1005091] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/16/2015] [Indexed: 01/20/2023] Open
Abstract
Chikungunya virus (CHIKV), the causative agent of a major epidemic spanning five continents, is a positive stranded mRNA virus that replicates using the cell's cap-dependent translation machinery. Despite viral infection inhibiting mTOR, a metabolic sensor controls cap-dependent translation, viral proteins are efficiently translated. Rapalog treatment, silencing of mtor or raptor genes, but not rictor, further enhanced CHIKV infection in culture cells. Using biochemical assays and real time imaging, we demonstrate that this effect is independent of autophagy or type I interferon production. Providing in vivo evidence for the relevance of our findings, mice treated with mTORC1 inhibitors exhibited increased lethality and showed a higher sensitivity to CHIKV. A systematic evaluation of the viral life cycle indicated that inhibition of mTORC1 has a specific positive effect on viral proteins, enhancing viral replication by increasing the translation of both structural and nonstructural proteins. Molecular analysis defined a role for phosphatidylinositol-3 kinase (PI3K) and MAP kinase-activated protein kinase (MnKs) activation, leading to the hyper-phosphorylation of eIF4E. Finally, we demonstrated that in the context of CHIKV inhibition of mTORC1, viral replication is prioritized over host translation via a similar mechanism. Our study reveals an unexpected bypass pathway by which CHIKV protein translation overcomes viral induced mTORC1 inhibition.
Collapse
Affiliation(s)
- Pierre-Emmanuel Joubert
- Unité Immunobiologie des Cellules Dendritiques, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
- INSERM U818, Paris, France
| | - Kenneth Stapleford
- Unité des populations virales et Pathogenèse, Département de Virologie, Institut Pasteur, Paris, Cedex 15, France
| | | | - Marco Vignuzzi
- Unité des populations virales et Pathogenèse, Département de Virologie, Institut Pasteur, Paris, Cedex 15, France
| | - Olivier Schwartz
- Unité Virus et Immunité, Département de Virologie Institut Pasteur, Paris, Cedex 15, France
| | - Matthew L. Albert
- Unité Immunobiologie des Cellules Dendritiques, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
- INSERM U818, Paris, France
- Centre d’Immunologie Humaine, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
| |
Collapse
|
32
|
Sun Y, Hu B, Fan C, Jia L, Zhang Y, Du A, Zheng X, Zhou J. iTRAQ-based quantitative subcellular proteomic analysis of Avibirnavirus-infected cells. Electrophoresis 2015; 36:1596-611. [PMID: 25929241 PMCID: PMC7163642 DOI: 10.1002/elps.201500014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/22/2015] [Accepted: 03/25/2015] [Indexed: 11/19/2022]
Abstract
Infectious bursal disease virus (IBDV) enters the host cells via endocytic pathway to achieve viral replication in the cytoplasm. Here, we performed LC-MS/MS coupled with isobaric tags for relative and absolute quantification labeling of differentially abundant proteins of IBDV-infected cells using a subcellular fractionation strategy. We show that the viral infection regulates the abundance and/or subcellular localization of 3211 proteins during early infection. In total, 23 cellular proteins in the cytoplasmic proteome and 34 in the nuclear proteome were significantly altered after virus infection. These differentially abundant proteins are involved in such biological processes as immune response, signal transduction, RNA processing, macromolecular biosynthesis, energy metabolism, virus binding, and cellular apoptosis. Moreover, transcriptional profiles of the 25 genes corresponding to the identified proteins were analyzed by quantitative real-time RT-PCR. Ingenuity Pathway Analysis clustered the differentially abundant proteins primarily into the mTOR pathway, PI3K/Akt pathway, and interferon-β signaling cascades. Confocal microscopy showed colocalization of the viral protein VP3 with host proteins heterogeneous nuclear ribonucleoprotein H1, nuclear factor 45, apoptosis inhibitor 5, nuclear protein localization protein 4 and DEAD-box RNA helicase 42 during the virus infection. Together, these identified subcellular constituents provide important information for understanding host-IBDV interactions and underlying mechanisms of IBDV infection and pathogenesis.
Collapse
Affiliation(s)
- Yanting Sun
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
| | - Boli Hu
- College of Veterinary MedicineNanjing Agricultural UniversityNanjingP. R. China
| | - Chengfei Fan
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
| | - Lu Jia
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
| | - Yina Zhang
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
| | - Aifang Du
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
| | - Xiaojuan Zheng
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
- State Key Laboratory and Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang UniversityHangzhouP. R. China
| | - Jiyong Zhou
- Key Laboratory of Animal Virology of Ministry of AgricultureZhejiang UniversityHangzhouP. R. China
- College of Veterinary MedicineNanjing Agricultural UniversityNanjingP. R. China
- State Key Laboratory and Collaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated Hospital, Zhejiang UniversityHangzhouP. R. China
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
Proteomics profiling of chikungunya-infected Aedes albopictus C6/36 cells reveal important mosquito cell factors in virus replication. PLoS Negl Trop Dis 2015; 9:e0003544. [PMID: 25738640 PMCID: PMC4349740 DOI: 10.1371/journal.pntd.0003544] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/16/2015] [Indexed: 12/18/2022] Open
Abstract
Chikungunya virus (CHIKV) is the only causative agent of CHIKV fever with persistent arthralgia, and in some cases may lead to neurological complications which can be highly fatal, therefore it poses severe health issues in many parts of the world. CHIKV transmission can be mediated via the Aedes albopictus mosquito; however, very little is currently known about the involvement of mosquito cellular factors during CHIKV-infection within the mosquito cells. Unravelling the neglected aspects of mosquito proteome changes in CHIKV-infected mosquito cells may increase our understanding on the differences in the host factors between arthropod and mammalian cells for successful replication of CHIKV. In this study, the CHIKV-infected C6/36 cells with differential cellular proteins expression were profiled using two-dimensional gel electrophoresis (2DE) coupled with the use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). 2DE analysis on CHIKV-infected C6/36 cells has shown 23 mosquito cellular proteins that are differentially regulated, and which are involved diverse biological pathways, such as protein folding and metabolic processes. Among those identified mosquito proteins, spermatogenesis-associated factor, enolase phosphatase e-1 and chaperonin-60kD have been found to regulate CHIKV infection. Furthermore, siRNA-mediated gene knockdown of these proteins has demonstrated the biological importance of these host proteins that mediate CHIKV infection. These findings have provided an insight to the importance of mosquito host factors in the replication of CHIKV, thus providing a potential channel for developing novel antiviral strategies against CHIKV transmission. Chikungunya virus (CHIKV), a re-emerging mosquito-borne virus, is the main cause of CHIKV fever, persistent arthralgia and serious neurological complications which can be highly fatal; therefore, it poses serious health threats worldwide. Unraveling the underappreciated aspects of mosquito cellular factors that contribute to the replication processes of CHIKV was performed using two-dimensional gel electrophoresis (2DE) coupled with the use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). The 2DE proteomics profiling of CHIKV-infected mosquito C6/36 cells has revealed twenty-three proteins that were differentially regulated upon CHIKV infection. These proteins are shown to be involved in diverse biological pathways and cellular processes. Notably, upon selected genes knockdown, spermatogenesis-associated factor, enolase phosphatase e-1 and chaperonin-60kD are found to be important during the replication processes of CHIKV. This study illustrates the importance of mosquito cellular factors in association with CHIKV infection in mosquito cells and reveals an interesting portal for developing novel antiviral strategies in CHIKV studies.
Collapse
|
36
|
Dudha N, Rana J, Rajasekharan S, Gabrani R, Gupta A, Chaudhary VK, Gupta S. Host-pathogen interactome analysis of Chikungunya virus envelope proteins E1 and E2. Virus Genes 2015; 50:200-9. [PMID: 25563600 DOI: 10.1007/s11262-014-1161-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/16/2014] [Indexed: 02/05/2023]
Abstract
The envelope proteins of Chikungunya virus (CHIKV) are known to play crucial roles in viral infection and spread. Although the role of envelope proteins in viral infection has been studied, the cellular interactors of these proteins are still elusive. In the present study, the ectodomains of CHIKV envelope proteins (E1 and E2) have been used for a high throughput yeast two-hybrid (Y2H) screening to identify the interacting host protein partners. Following a comparative analysis between the viral-host protein interaction data generated from Y2H and computational approach, five host proteins interacting with E1 and three host proteins interacting with E2 common to both datasets were identified. These associations were further verified independently by pull down and protein interaction ELISA. The identified interactions shed light on the possible cellular machinery that CHIKV might be employing during viral entry, trafficking, and evasion of immune system.
Collapse
Affiliation(s)
- Namrata Dudha
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, 201307, UP, India
| | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Gérardin P, Sampériz S, Ramful D, Boumahni B, Bintner M, Alessandri JL, Carbonnier M, Tiran-Rajaoefera I, Beullier G, Boya I, Noormahomed T, Okoï J, Rollot O, Cotte L, Jaffar-Bandjee MC, Michault A, Favier F, Kaminski M, Fourmaintraux A, Fritel X. Neurocognitive outcome of children exposed to perinatal mother-to-child Chikungunya virus infection: the CHIMERE cohort study on Reunion Island. PLoS Negl Trop Dis 2014; 8:e2996. [PMID: 25033077 PMCID: PMC4102444 DOI: 10.1371/journal.pntd.0002996] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 05/22/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Little is known about the neurocognitive outcome in children exposed to perinatal mother-to-child Chikungunya virus (p-CHIKV) infection. METHODS The CHIMERE ambispective cohort study compared the neurocognitive function of 33 p-CHIKV-infected children (all but one enrolled retrospectively) at around two years of age with 135 uninfected peers (all enrolled prospectively). Psychomotor development was assessed using the revised Brunet-Lezine scale, examiners blinded to infectious status. Development quotients (DQ) with subscores covering movement/posture, coordination, language, sociability skills were calculated. Predictors of global neurodevelopmental delay (GND, DQ ≤ 85), were investigated using multivariate Poisson regression modeling. Neuroradiologic follow-up using magnetic resonance imaging (MRI) scans was proposed for most of the children with severe forms. RESULTS The mean DQ score was 86.3 (95%CI: 81.0-91.5) in infected children compared to 100.2 (95%CI: 98.0-102.5) in uninfected peers (P<0.001). Fifty-one percent (n = 17) of infected children had a GND compared to 15% (n = 21) of uninfected children (P<0.001). Specific neurocognitive delays in p-CHIKV-infected children were as follows: coordination and language (57%), sociability (36%), movement/posture (27%). After adjustment for maternal social situation, small for gestational age, and head circumference, p-CHIKV infection was found associated with GND (incidence rate ratio: 2.79, 95%CI: 1.45-5.34). Further adjustments on gestational age or breastfeeding did not change the independent effect of CHIKV infection on neurocognitive outcome. The mean DQ of p-CHIKV-infected children was lower in severe encephalopathic children than in non-severe children (77.6 versus 91.2, P<0.001). Of the 12 cases of CHIKV neonatal encephalopathy, five developed a microcephaly (head circumference <-2 standard deviations) and four matched the definition of cerebral palsy. MRI scans showed severe restrictions of white matter areas, predominant in the frontal lobes in these children. CONCLUSIONS The neurocognitive outcome of children exposed to perinatal mother-to-child CHIKV infection is poor. Severe CHIKV neonatal encephalopathy is associated with an even poorer outcome.
Collapse
Affiliation(s)
- Patrick Gérardin
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
- INSERM CIC-EC (CIE2), Saint-Pierre, La Réunion, France
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
| | | | - Duksha Ramful
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
- INSERM CIC-EC (CIE2), Saint-Pierre, La Réunion, France
- GRI, Research Group on Immunopathology and Infection, EA4517, Université de La Réunion, INSERM UMRS 945 “Immunity and Infection” Saint-Denis, La Réunion, France
| | - Brahim Boumahni
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | - Marc Bintner
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | | | | | - Gilles Beullier
- Centre Hospitalier Gabriel Martin, Saint-Paul, La Réunion, France
| | - Irénée Boya
- Centre Hospitalier de l'Est Réunion, Saint-Benoît, La Réunion, France
| | | | - Jocelyn Okoï
- Clinique Durieux, Le Tampon, La Réunion, France
- Centre d'Action Médico-Sociale Précoce (CAMSP), Saint-Louis, La Réunion, France
| | | | - Liliane Cotte
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | - Alain Michault
- CHU de La Réunion Saint-Denis/Saint-Pierre, La Réunion, France
| | | | - Monique Kaminski
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
| | | | - Xavier Fritel
- INSERM UMRS 953, “Epidemiological Research Unit on Perinatal Health and Women and Children Health”, UPMC Université Paris 6, Paris, France
- Poitiers University Hospital, Poitiers, France
- INSERM CIC-P 0802, Poitiers, France
| |
Collapse
|
39
|
Issac THK, Tan EL, Chu JJH. Proteomic profiling of chikungunya virus-infected human muscle cells: reveal the role of cytoskeleton network in CHIKV replication. J Proteomics 2014; 108:445-64. [PMID: 24933005 DOI: 10.1016/j.jprot.2014.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/06/2014] [Accepted: 06/03/2014] [Indexed: 02/01/2023]
Abstract
UNLABELLED Chikungunya virus (CHIKV) is an arthropod-borne, positive-sense, single-stranded RNA virus belonging to genus Alphavirus and family Togaviridae. The clinical manifestations developed upon CHIKV-infection include fever, myositis, arthralgia and maculopapular rash. Thus, the re-emergence of CHIKV has posed serious health threats worldwide. Due to the fact that myositis is induced upon CHIKV-infection, we sought to understand the dynamic proteomic regulation in SJCRH30, a human rhabdomyosarcoma cell line, to gain insights on CHIKV pathogenesis. Two-dimensional gel electrophoresis (2DE) in combination of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was used to profile differential cellular proteins expression in CHIKV-infected SJCRH30 cells. 2DE analysis on CHIKV-infected cells has revealed 44 protein spots. These spots are found to be involved in various biological pathways such as biomolecules synthesis and metabolism, cell signaling and cellular reorganization. siRNA-mediated gene silencing on selected genes has elucidated the biological significance of these gene-translated host proteins involved in CHIKV-infection. More importantly, the interaction of vimentin with non-structural protein (nsP3) of CHIKV was shown, suggesting the role played by vimentin during CHIKV replication by forming an anchorage network with the CHIKV replication complexes (RCs). BIOLOGICAL SIGNIFICANCE Chikungunya virus (CHIKV) is a re-emerging virus that has caused various disease outbreaks in Africa and Asia. The clinical symptoms of CHIKV-infection include fever, skin rash, recurrent joint paint, and myositis. Neuronal implications and death may be resulted from the severe viral infection. Up to date, there are no effective treatments and vaccines against CHIKV-infection. More importantly, little is known about the differential regulation of host proteins upon CHIKV infection, hence deciphering the viral-host cell interactions during viral infection provide critical information on our understanding on the mechanisms of virus infection and its dependency of host proteins for replication. In light of the muscle-related clinical manifestations of myositis resulting from CHIKV-infection, human rhabdomyosarcoma cells, SJCRH30 were utilized in this protein profiling study, in order to decipher the pathogenesis of CHIKV. This study has identified an arrays of host proteins that are differentially regulated upon CHIKV infection including that of the cytoskeletal protein, vimentin that plays significant role in aiding the replication of CHIKV within the host cells through 2DE assay. Immunofluorescence assay further shows that the novel interaction between cytoskeleton structure and CHIKV replication complex by forming an intercalating network around the replication complexes and facilitating various stages of the virus life cycle. This novel finding has inevitably led to a deeper understanding of CHIKV pathogenesis in revealing the importance of host proteins during CHIKV replication, as well as contributing to the development of specific antiviral strategies against this medically important viral pathogen.
Collapse
Affiliation(s)
- Too Horng Khit Issac
- Laboratory of Molecular RNA Virology and Antiviral Strategies. Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, 5 Science Drive 2, National University of Singapore, Singapore 117597
| | - Eng Lee Tan
- Department of Paediatrics, University Children's Medical Institute, National University Hospital, Singapore, Singapore; Centre for Biomedical and Life Sciences, Singapore Polytechnic, 500 Dover Road, Singapore, Singapore
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies. Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, 5 Science Drive 2, National University of Singapore, Singapore 117597.
| |
Collapse
|
40
|
Priya R, Patro IK, Parida MM. TLR3 mediated innate immune response in mice brain following infection with Chikungunya virus. Virus Res 2014; 189:194-205. [PMID: 24905288 DOI: 10.1016/j.virusres.2014.05.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
Chikungunya virus (CHIKV) has received global attention due to the series of large-scale outbreaks in different parts of the world. Many unusual clinical severities including neurological complications and death were reported in recent outbreaks. The mechanism underlying the host immune response to CHIKV in the brain is poorly characterized. In this study, the neuropathogenesis of CHIKV with E1:A226V mutation was elucidated in 1 week old BALB/c mice. The virus was found to replicate in mice brain with peak titer of 10(4) on 6th day post infection. Immunohistochemical analysis revealed preferential virus localization in neuronal cells of cerebellum. The expression profiling of TLR, antiviral genes and cytokines in mice brain revealed significant up regulation of TLR3, TRAF-6, TICAM-1, MCP-1, CXCL-10, IL-6, IL-4, ISG-15, MX-2, IFN-β, OAS-3 genes that ultimately resulted in virus clearance from brain by day 9-10 suggesting activation of innate immune pathway. Further the effect of poly I: C (Polyinosinic: Polycytidylic acid), a TLR-3 agonist and potent IFN inducer on CHIKV neuropathogenesis was studied. Pretreatment of mice with Poly I: C caused reduction of CHIKV titer in brain and offered 100% protection of animals. The protection was mediated by an increased induction of TLR3, IFN-β and antiviral genes in mice brain. Our result demonstrates that pre immune stimulation of animals by Poly I: C is effective inhibitor of CHIKV replication and might be a promising prevention agent against this virus.
Collapse
Affiliation(s)
- Raj Priya
- Division of Virology, Defence Research & Development Establishment, Gwalior 474002, India
| | - I K Patro
- School of Studies in Neurosciences, Jiwaji University, Gwalior 474002, India
| | - M M Parida
- Division of Virology, Defence Research & Development Establishment, Gwalior 474002, India.
| |
Collapse
|
41
|
Wikan N, Khongwichit S, Phuklia W, Ubol S, Thonsakulprasert T, Thannagith M, Tanramluk D, Paemanee A, Kittisenachai S, Roytrakul S, Smith DR. Comprehensive proteomic analysis of white blood cells from chikungunya fever patients of different severities. J Transl Med 2014; 12:96. [PMID: 24721947 PMCID: PMC4022080 DOI: 10.1186/1479-5876-12-96] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/02/2014] [Indexed: 01/13/2023] Open
Abstract
Background Chikungunya fever (CHIKF) is a recently re-emerged mosquito transmitted viral disease caused by the chikungunya virus (CHIKV), an Alphavirus belonging to the family Togaviridae. Infection of humans with CHIKV can result in CHIKF of variable severity, although the factors mediating disease severity remain poorly defined. Methods White blood cells were isolated from blood samples collected during the 2009-2010 CHIKF outbreak in Thailand. Clinical presentation and viral load data were used to classify samples into three groups, namely non chikungunya fever (non-CHIKF), mild CHIKF, and severe CHIKF. Five samples from each group were analyzed for protein expression by GeLC-MS/MS. Results CHIKV proteins (structural and non-structural) were found only in CHIKF samples. A total of 3505 human proteins were identified, with 68 proteins only present in non-CHIKF samples. A total of 240 proteins were found only in CHIKF samples, of which 65 and 46 were found only in mild and severe CHIKF samples respectively. Proteins with altered expression mapped predominantly to cellular signaling pathways (including toll-like receptor and PI3K-Akt signaling) although many other processes showed altered expression as a result of CHIKV infection. Expression of proteins consistent with the activation of the inflammasome was detected, and quantitation of (pro)-caspase 1 at the protein and RNA levels showed an association with disease severity. Conclusions This study confirms the infection of at least a component of white blood cells by CHIKV, and shows that CHIKV infection results in activation of the inflammasome in a manner that is associated with disease severity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya Campus 25/25 Phuttamonthol Sai 4, Nakorn Pathom 73170, Thailand.
| |
Collapse
|
42
|
Dhanwani R, Khan M, Lomash V, Rao PVL, Ly H, Parida M. Characterization of chikungunya virus induced host response in a mouse model of viral myositis. PLoS One 2014; 9:e92813. [PMID: 24667237 PMCID: PMC3965460 DOI: 10.1371/journal.pone.0092813] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 02/25/2014] [Indexed: 11/30/2022] Open
Abstract
While a number of studies have documented the persistent presence of chikungunya virus (CHIKV) in muscle tissue with primary fibroblast as the preferable cell target, little is known regarding the alterations that take place in muscle tissue in response to CHIKV infection. Hence, in the present study a permissive mouse model of CHIKV infection was established and characterized in order to understand the pathophysiology of the disease. The two dimensional electrophoresis of muscle proteome performed for differential analysis indicated a drastic reprogramming of the proteins from various classes like stress, inflammation, cytoskeletal, energy and lipid metabolism. The roles of the affected proteins were explained in relation to virus induced myopathy which was further supported by the histopathological and behavioural experiments proving the lack of hind limb coordination and other loco-motor abnormalities in the infected mice. Also, the level of various pro-inflammatory mediators like IL-6, MCP-1, Rantes and TNF-α was significantly elevated in muscles of infected mice. Altogether this comprehensive study of characterizing CHIKV induced mouse myopathy provides many potential targets for further evaluation and biomarker study.
Collapse
Affiliation(s)
- Rekha Dhanwani
- Department of Virology, Defence Research & Development Establishment (DRDE), Gwalior, India
| | - Mohsin Khan
- Department of Virology, Defence Research & Development Establishment (DRDE), Gwalior, India
| | - Vinay Lomash
- Department of Pharmacology and Toxicology, Defence Research & Development Establishment (DRDE), Gwalior, India
| | | | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Saint Paul, Minnestoa, United States of America
| | - Manmohan Parida
- Department of Virology, Defence Research & Development Establishment (DRDE), Gwalior, India
| |
Collapse
|
43
|
Fraisier C, Koraka P, Belghazi M, Bakli M, Granjeaud S, Pophillat M, Lim SM, Osterhaus A, Martina B, Camoin L, Almeras L. Kinetic analysis of mouse brain proteome alterations following Chikungunya virus infection before and after appearance of clinical symptoms. PLoS One 2014; 9:e91397. [PMID: 24618821 PMCID: PMC3949995 DOI: 10.1371/journal.pone.0091397] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/10/2014] [Indexed: 01/13/2023] Open
Abstract
Recent outbreaks of Chikungunya virus (CHIKV) infection have been characterized by an increasing number of severe cases with atypical manifestations including neurological complications. In parallel, the risk map of CHIKV outbreaks has expanded because of improved vector competence. These features make CHIKV infection a major public health concern that requires a better understanding of the underlying physiopathological processes for the development of antiviral strategies to protect individuals from severe disease. To decipher the mechanisms of CHIKV infection in the nervous system, a kinetic analysis on the host proteome modifications in the brain of CHIKV-infected mice sampled before and after the onset of clinical symptoms was performed. The combination of 2D-DIGE and iTRAQ proteomic approaches, followed by mass spectrometry protein identification revealed 177 significantly differentially expressed proteins. This kinetic analysis revealed a dramatic down-regulation of proteins before the appearance of the clinical symptoms followed by the increased expression of most of these proteins in the acute symptomatic phase. Bioinformatic analyses of the protein datasets enabled the identification of the major biological processes that were altered during the time course of CHIKV infection, such as integrin signaling and cytoskeleton dynamics, endosome machinery and receptor recycling related to virus transport and synapse function, regulation of gene expression, and the ubiquitin-proteasome pathway. These results reveal the putative mechanisms associated with severe CHIKV infection-mediated neurological disease and highlight the potential markers or targets that can be used to develop diagnostic and/or antiviral tools.
Collapse
Affiliation(s)
- Christophe Fraisier
- Aix Marseille Université, Unité de Recherche en Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
| | - Penelope Koraka
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Maya Belghazi
- Aix-Marseille Université, CNRS, CRN2M UMR 7286, Marseille, France
| | - Mahfoud Bakli
- Aix Marseille Université, Unité de Recherche en Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
| | - Samuel Granjeaud
- CRCM, Marseille Protéomique, Inserm, U1068, Marseille, France
- Aix-Marseille Université, UM 105, Marseille, France
| | - Matthieu Pophillat
- CRCM, Marseille Protéomique, Inserm, U1068, Marseille, France
- Aix-Marseille Université, UM 105, Marseille, France
| | - Stephanie M. Lim
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Albert Osterhaus
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Byron Martina
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Luc Camoin
- CRCM, Marseille Protéomique, Inserm, U1068, Marseille, France
- Aix-Marseille Université, UM 105, Marseille, France
| | - Lionel Almeras
- Aix Marseille Université, Unité de Recherche en Maladies Infectieuses et Tropicales Emergentes, UM63, CNRS 7278, IRD 198, Inserm 1095, Marseille, France
- Unité de recherche en biologie et épidémiologie parasitaires (URBEP), Institut de Recherche Biomédicale des Armées (IRBA), Marseille, France
- * E-mail:
| |
Collapse
|
44
|
Cruz DJM, Bonotto RM, Gomes RGB, da Silva CT, Taniguchi JB, No JH, Lombardot B, Schwartz O, Hansen MAE, Freitas-Junior LH. Identification of novel compounds inhibiting chikungunya virus-induced cell death by high throughput screening of a kinase inhibitor library. PLoS Negl Trop Dis 2013; 7:e2471. [PMID: 24205414 PMCID: PMC3814572 DOI: 10.1371/journal.pntd.0002471] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 08/27/2013] [Indexed: 12/16/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne arthrogenic alphavirus that causes acute febrile illness in humans accompanied by joint pains and in many cases, persistent arthralgia lasting weeks to years. The re-emergence of CHIKV has resulted in numerous outbreaks in the eastern hemisphere, and threatens to expand in the foreseeable future. Unfortunately, no effective treatment is currently available. The present study reports the use of resazurin in a cell-based high-throughput assay, and an image-based high-content assay to identify and characterize inhibitors of CHIKV-infection in vitro. CHIKV is a highly cytopathic virus that rapidly kills infected cells. Thus, cell viability of HuH-7 cells infected with CHIKV in the presence of compounds was determined by measuring metabolic reduction of resazurin to identify inhibitors of CHIKV-associated cell death. A kinase inhibitor library of 4,000 compounds was screened against CHIKV infection of HuH-7 cells using the resazurin reduction assay, and the cell toxicity was also measured in non-infected cells. Seventy-two compounds showing ≥50% inhibition property against CHIKV at 10 µM were selected as primary hits. Four compounds having a benzofuran core scaffold (CND0335, CND0364, CND0366 and CND0415), one pyrrolopyridine (CND0545) and one thiazol-carboxamide (CND3514) inhibited CHIKV-associated cell death in a dose-dependent manner, with EC50 values between 2.2 µM and 7.1 µM. Based on image analysis, these 6 hit compounds did not inhibit CHIKV replication in the host cell. However, CHIKV-infected cells manifested less prominent apoptotic blebs typical of CHIKV cytopathic effect compared with the control infection. Moreover, treatment with these compounds reduced viral titers in the medium of CHIKV-infected cells by up to 100-fold. In conclusion, this cell-based high-throughput screening assay using resazurin, combined with the image-based high content assay approach identified compounds against CHIKV having a novel antiviral activity - inhibition of virus-induced CPE - likely by targeting kinases involved in apoptosis. Recent outbreaks and expanding global distribution of Chikungunya virus (CHIKV) in different regions of Asia, Africa and Europe necessitates the development of effective therapeutic interventions. At present, only two antiviral compounds (chloroquine and ribavirin) that inhibit viral infection in vitro have been used in clinical cases of chikungunya infections. However, neither of these compounds have shown strong efficacy in vivo. Recent attempts to identify new antiviral candidates for CHIKV using cell-based phenotypic approach have been reported. In this study, we developed a simple cell-based high-throughput assay using resazurin to identify potential anti-CHIKV compounds. This high-throughput assay is based on the metabolic reduction of resazurin to the highly fluorescent resorufin by viable cells as an indicator of activity against CHIKV-induced CPE. We screened 4,000 small molecules belonging to the BioFocus kinase inhibitor chemical library and found a cluster of related molecules with antiviral activity against CHIKV. Finally, we characterized the putative mode of action of these active compounds using an image-based high content assay and conventional virological methods (i.e., virus yield reduction assay, microneutralization assay).
Collapse
Affiliation(s)
- Deu John M. Cruz
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
| | - Rafaela M. Bonotto
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
- Universidade Feevale, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Rafael G. B. Gomes
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
- Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - Camila T. da Silva
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
- Universidade Estadual do Rio Grande do Sul - Campus Novo Hamburgo, Novo Hamburgo, Rio Grande do Sul, Brazil
| | - Juliana B. Taniguchi
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
- Universidade Estadual Paulista “Júlio de Mesquita Filho”-Campus Araraquara, Araraquara, São Paulo, Brazil
| | - Joo Hwan No
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
| | - Benoit Lombardot
- Image Mining Group (IMG), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, Paris, France
| | - Michael A. E. Hansen
- Image Mining Group (IMG), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
| | - Lucio H. Freitas-Junior
- Center for Neglected Diseases Drug Discovery (CND3), Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, South Korea
- * E-mail: ,
| |
Collapse
|
45
|
Jha MK, Kim JH, Suk K. Proteome of brain glia: the molecular basis of diverse glial phenotypes. Proteomics 2013; 14:378-98. [PMID: 24124134 DOI: 10.1002/pmic.201300236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/16/2013] [Accepted: 07/30/2013] [Indexed: 12/11/2022]
Abstract
Several different types of nonneuronal glial cells with diverse phenotypes are present in the CNS, and all have distinct indispensible functions. Although glial cells primarily provide neurons with metabolic and structural support in the healthy brain, they may switch phenotype from a "resting" to a "reactive" state in response to pathological insults. Furthermore, this reactive gliosis is an invariant feature of the pathogeneses of CNS maladies. The glial proteome serves as a signature of glial phenotype, and not only executes physiological functions, but also acts as a molecular mediator of the reactive glial phenotype. The glial proteome is also involved in intra- and intercellular communications as exemplified by glia-glia and neuron-glia interactions. The utilization of authoritative proteomic tools and the bioinformatic analyses have helped to profile the brain glial proteome and explore the molecular mechanisms of diverse glial phenotypes. Furthermore, technologic innovations have equipped the field of "glioproteomics" with refined tools for studies of the expression, interaction, and function of glial proteins in the healthy and in the diseased CNS. Glioproteomics is expected to contribute to the elucidation of the molecular mechanisms of CNS pathophysiology and to the discovery of biomarkers and theragnostic targets in CNS disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | |
Collapse
|
46
|
Priya R, Dhanwani R, Patro IK, Rao PVL, Parida MM. Differential regulation of TLR mediated innate immune response of mouse neuronal cells following infection with novel ECSA genotype of Chikungunya virus with and without E1:A226V mutation. INFECTION GENETICS AND EVOLUTION 2013; 20:396-406. [PMID: 24126361 DOI: 10.1016/j.meegid.2013.09.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 02/01/2023]
Abstract
Chikungunya virus (CHIKV) has received global attention due to the series of large-scale outbreaks in different parts of the world including Africa, Indian Ocean Islands, India and South-East Asia. The appearance of many unusual severe manifestations including neurological disorders was reported in post resurgence epidemics with implication of novel East Central South African (ECSA) genotype with E1:A226V mutation. The molecular mechanism of CHIKV neuropathogenesis is not yet understood and very little is known about the host-pathogen interactions. In the present study replication kinetics and innate immune response of ECSA genotype of CHIKV with and without A226V mutation were determined in mouse neuroblastoma cell line (N2a). The 226V mutant strain was more replication competent in N2a cells with a peak titer of 10(8)PFU/ml compared to 10(6)PFU/ml for A226 virus. Besides, the 226V mutant virus showed relatively less induction of antiviral genes i.e. IFN-β, OAS-3, MX-2, ISG-15 and Toll like receptors 3 and 7 as compared to non mutant strain (A226). Further pretreatment of N2a cells either with Poly I: C, IFN-β or TNF-α resulted in inhibition of CHIKV replication hence confirming the role of TLR mediated innate immune response in CHIKV pathogenesis. Differential regulation of TLRs and associated down stream antiviral genes might have attributed for increased pathogenesis of the 226V mutant novel ECSA genotype of CHIKV during the recent epidemics.
Collapse
Affiliation(s)
- Raj Priya
- Division of Virology, Defence Research & Development Establishment, Gwalior 474002, India
| | | | | | | | | |
Collapse
|
47
|
Abraham R, Mudaliar P, Padmanabhan A, Sreekumar E. Induction of cytopathogenicity in human glioblastoma cells by chikungunya virus. PLoS One 2013; 8:e75854. [PMID: 24086645 PMCID: PMC3783433 DOI: 10.1371/journal.pone.0075854] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 08/22/2013] [Indexed: 01/01/2023] Open
Abstract
Chikungunya virus (CHIKV), an arthritogenic old-world alphavirus, has been implicated in the central nervous system (CNS) infection in infants and elderly patients. Astrocytes are the major immune cells of the brain parenchyma that mediate inflammation. In the present study we found that a local isolate of CHIKV infect and activate U-87 MG cells, a glioblastoma cell line of human astrocyte origin. The infection kinetics were similar in infected U-87 MG cells and the human embryo kidney (HEK293) cells as indicated by immunofluorescence and plaque assays, 24h post-infection (p.i.). In infected U-87 MG cells, apoptosis was detectable from 48h p.i. evidenced by DNA fragmentation, PARP cleavage, loss of mitochondrial membrane potential, nuclear condensation and visible cytopathic effects in a dose and time-dependent manner. XBP1 mRNA splicing and eIF2α phosphorylation studies indicated the occurrence of endoplasmic reticulum stress in infected cells. In U-87 MG cells stably expressing a green fluorescent protein-tagged light chain-3 (GFP-LC3) protein, CHIKV infection showed increased autophagy response. The infection led to an enhanced expression of the mRNA transcripts of the pro-inflammatory cytokines IL-1β, TNF-α, IL-6 and CXCL9 within 24h p.i. Significant up-regulation of the proteins of RIG-I like receptor (RLR) pathway, such as RIG-I and TRAF-6, was observed indicating the activation of the cytoplasmic-cellular innate immune response. The overall results show that the U-87 MG cell line is a potential in vitro model for in depth study of these molecular pathways in response to CHIKV infection. The responses in these cells of CNS origin, which are inherently defective in Type I interferon response, could be analogous to that occurring in infants and very old patients who also have a compromised interferon-response. The results also point to the intriguing possibility of using this virus for studies to develop oncolytic virus therapy approaches against glioblastoma, a highly aggressive malignancy.
Collapse
Affiliation(s)
- Rachy Abraham
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Prashant Mudaliar
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Aiswaria Padmanabhan
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| | - Easwaran Sreekumar
- Viral Disease Biology Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India
| |
Collapse
|
48
|
Differential proteome analysis of chikungunya virus infection on host cells. PLoS One 2013; 8:e61444. [PMID: 23593481 PMCID: PMC3622599 DOI: 10.1371/journal.pone.0061444] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 03/10/2013] [Indexed: 11/19/2022] Open
Abstract
Background Chikungunya virus (CHIKV) is an emerging mosquito-borne alphavirus that has caused multiple unprecedented and re-emerging outbreaks in both tropical and temperate countries. Despite ongoing research efforts, the underlying factors involved in facilitating CHIKV replication during early infection remains ill-characterized. The present study serves to identify host proteins modulated in response to early CHIKV infection using a proteomics approach. Methodology and Principal Findings The whole cell proteome profiles of CHIKV-infected and mock control WRL-68 cells were compared and analyzed using two-dimensional gel electrophoresis (2-DGE). Fifty-three spots were found to be differentially modulated and 50 were successfully identified by MALDI-TOF/TOF. Eight were significantly up-regulated and 42 were down-regulated. The mRNA expressions of 15 genes were also found to correlate with the corresponding protein expression. STRING network analysis identified several biological processes to be affected, including mRNA processing, translation, energy production and cellular metabolism, ubiquitin-proteasome pathway (UPP) and cell cycle regulation. Conclusion/Significance This study constitutes a first attempt to investigate alteration of the host cellular proteome during early CHIKV infection. Our proteomics data showed that during early infection, CHIKV affected the expression of proteins that are involved in mRNA processing, host metabolic machinery, UPP, and cyclin-dependent kinase 1 (CDK1) regulation (in favour of virus survival, replication and transmission). While results from this study complement the proteomics results obtained from previous late host response studies, functional characterization of these proteins is warranted to reinforce our understanding of their roles during early CHIKV infection in humans.
Collapse
|
49
|
Wintachai P, Wikan N, Kuadkitkan A, Jaimipuk T, Ubol S, Pulmanausahakul R, Auewarakul P, Kasinrerk W, Weng WY, Panyasrivanit M, Paemanee A, Kittisenachai S, Roytrakul S, Smith DR. Identification of prohibitin as a Chikungunya virus receptor protein. J Med Virol 2013; 84:1757-70. [PMID: 22997079 DOI: 10.1002/jmv.23403] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Chikungunya virus (CHIKV) has recently re-emerged causing millions of infections in countries around the Indian Ocean. While CHIKV has a broad host cell range and productively infects a number of different cell types, macrophages have been identified as a potential viral reservoir serving to increase the duration of symptoms. To date no CHIKV interacting protein has been characterized and this study sought to identify CHIKV binding proteins expressed on target cell membranes. Two-dimensional virus overlay identified prohibitin (PHB) as a microglial cell expressed CHIKV binding protein. Co-localization, co-immunoprecipitation as well as antibody and siRNA mediated infection inhibition studies all confirmed a role for PHB in mediating internalization of CHIKV into microglial cells. PHB is the first identified CHIKV receptor protein, and this study is evidence that PHB may play a role in the internalization of multiple viruses.
Collapse
|
50
|
Affiliation(s)
- G Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Sapienza University, Rome, Italy.
| |
Collapse
|