1
|
He H, Yuan K, Pan J, Weng S, Li C, Chen Y, He J. Shrimp Virus Regulates ROS Dynamics via the Nrf2 Pathway to Facilitate Viral Replication. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407695. [PMID: 40091388 PMCID: PMC12079347 DOI: 10.1002/advs.202407695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Reactive oxygen species (ROS) of hosts are widely involved in intracellular signaling and against pathogens. Viruses manipulate ROS homeostasis of hosts as a strategy to evade ROS-mediated negative effects of their infection, but the mechanisms remain unclear. The economically important aquaculture shrimp, Litopenaeus vannamei, is selected to investigate the molecular mechanism of how white spot syndrome virus (WSSV) regulates ROS dynamics and enhances viral replication. WSSV protein wsv220 binds to the repressor of shrimp nuclear factor erythroid 2-related factor 2 (LvNrf2), called Kelch-like ECH-associated protein 1 (LvKeap1), disrupting LvNrf2/LvKeap1 complex and facilitating LvNrf2 nuclear translocation. This activation of LvNrf2 causes up-regulation of antioxidant genes, including glucose-6-phosphate dehydrogenase (LvG6PDH), which increases nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH) production, effectively eliminating excessive ROS. Moreover, WSSV exploits LvNrf2 to establish a positive feedback loop by up-regulating viral immediate early gene wsv051, which further enhances wsv220 expression. Knockdown of LvNrf2 or LvG6PDH reduces WSSV replication and increases host ROS levels. Therefore, WSSV hijacks LvNrf2 pathway to maintain ROS homeostasis and establishes a positive feedback loop to facilitate WSSV replication. These findings reveal a novel molecular mechanism of viral manipulation of host ROS dynamics and suggest potential antiviral strategies targeting LvNrf2 pathway.
Collapse
Affiliation(s)
- Honghui He
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Kai Yuan
- School of Life SciencesHuizhou UniversityHuizhouGuangdong516007China
| | - Junming Pan
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Shaoping Weng
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Chaozheng Li
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Yihong Chen
- Institute of Modern Aquaculture Science and EngineeringCollege of Life SciencesSouth China Normal UniversityGuangzhouGuangdong510631China
| | - Jianguo He
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| |
Collapse
|
2
|
Chen CY, Chen CL, Ng YS, Lee DY, Lin SS, Huang CK, Kumar R, Wang HC. Glucose- and glutamine-driven de novo nucleotide synthesis facilitates WSSV replication in shrimp. Cell Commun Signal 2025; 23:191. [PMID: 40264189 PMCID: PMC12012963 DOI: 10.1186/s12964-025-02186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/30/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Viruses rely on host metabolism to complete their replication cycle. White spot syndrome virus (WSSV), a major pathogen in shrimp aquaculture, hijacks host metabolic pathways to fulfill its biosynthetic and energetic needs. Previous studies have demonstrated that WSSV promotes aerobic glycolysis (Warburg effect) and glutaminolysis during its replication stage (12 hpi). Therefore, glucose and glutamine serve as crucial metabolites for viral replication. Additionally, de novo nucleotide synthesis, including the pentose phosphate pathway and purine/pyrimidine synthesis, is significantly activated during WSSV infection. However, the precise association between WSSV and host glucose and glutamine metabolism in driving de novo nucleotide synthesis remains unclear. This study aimed to investigate the involvement of glucose and glutamine in nucleotide metabolism during WSSV replication and to elucidate how WSSV reprograms these pathways to facilitate its pathogenesis. METHODS To assess changes in metabolic flux during WSSV replication, LC-ESI-MS-based isotopically labeled glucose ([U-13C] glucose) and glutamine ([A-15N] glutamine) were used as metabolic tracers in in vivo experiments with white shrimp (Litopenaeus vannamei). The in vivo experiments were also conducted to measure the expression and enzymatic activity of genes involved in nucleotide metabolism. Additionally, in vivo dsRNA-mediated gene silencing was employed to evaluate the roles of these genes in WSSV replication. Pharmacological inhibitors targeting the Ras-PI3K-Akt-mTOR pathway were also applied to investigate its regulatory role in WSSV-induced nucleotide metabolic reprogramming. RESULTS The metabolite tracking analysis confirmed that de novo nucleotide synthesis was significantly activated at the WSSV replication stage (12 hpi). Glucose metabolism is preferentially reprogrammed to support purine synthesis, while glutamine uptake is significantly increased and contributes to both purine and pyrimidine synthesis. Consistently, gene expression and enzymatic activity analyses, along with gene silencing experiments, indicated the critical role of de novo nucleotide synthesis in supporting viral replication. However, while the inhibition of the Ras-PI3K-Akt-mTOR pathway suggested its involvement in regulating nucleotide metabolism, no consistent effect on WSSV replication was observed, suggesting the presence of alternative regulatory mechanisms. CONCLUSION This study demonstrates that WSSV infection induces specific metabolic reprogramming of glucose and glutamine utilization to facilitate de novo nucleotide synthesis in shrimp. These metabolic changes provide the necessary precursors for nucleotide synthesis, supporting WSSV replication and pathogenesis. The findings offer novel insights into the metabolic strategies employed by WSSV and suggest potential targets for controlling WSSV outbreaks in shrimp aquaculture.
Collapse
Affiliation(s)
- Cong-Yan Chen
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Ling Chen
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan
| | - Yen Siong Ng
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Shun Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chien-Kang Huang
- Department of Engineering Science and Ocean Engineering, National Taiwan University, Taipei, Taiwan
| | - Ramya Kumar
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan
| | - Han-Ching Wang
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan.
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
3
|
Pérez SE, Gooz M, Maldonado EN. Mitochondrial Dysfunction and Metabolic Disturbances Induced by Viral Infections. Cells 2024; 13:1789. [PMID: 39513896 PMCID: PMC11545457 DOI: 10.3390/cells13211789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Viruses are intracellular parasites that utilize organelles, signaling pathways, and the bioenergetics machinery of the cell to replicate the genome and synthesize proteins to build up new viral particles. Mitochondria are key to supporting the virus life cycle by sustaining energy production, metabolism, and synthesis of macromolecules. Mitochondria also contribute to the antiviral innate immune response. Here, we describe the different mechanisms involved in virus-mitochondria interactions. We analyze the effects of viral infections on the metabolism of glucose in the Warburg phenotype, glutamine, and fatty acids. We also describe how viruses directly regulate mitochondrial function through modulation of the activity of the electron transport chain, the generation of reactive oxygen species, the balance between fission and fusion, and the regulation of voltage-dependent anion channels. In addition, we discuss the evasion strategies used to avoid mitochondrial-associated mechanisms that inhibit viral replication. Overall, this review aims to provide a comprehensive view of how viruses modulate mitochondrial function to maintain their replicative capabilities.
Collapse
Affiliation(s)
- Sandra E. Pérez
- Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil CC7000, Buenos Aires, Argentina;
| | - Monika Gooz
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
| | - Eduardo N. Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
4
|
Zhang Y, Zhang X. Virus-Induced Histone Lactylation Promotes Virus Infection in Crustacean. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401017. [PMID: 38874057 PMCID: PMC11321649 DOI: 10.1002/advs.202401017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/04/2024] [Indexed: 06/15/2024]
Abstract
As "non-cellular organisms", viruses need to infect living cells to survive themselves. The virus infection must alter host's metabolisms. However, the influence of the metabolites from the altered metabolisms of virus-infected host cells on virus-host interactions remains largely unclear. To address this issue, shrimp, a representative species of crustaceans, is challenged with white spot syndrome virus (WSSV) in this study. The in vivo results presented that the WSSV infection enhanced shrimp glycolysis, leading to the accumulation of lactate. The lactate accumulation in turn promoted the site-specific histone lactylation (H3K18la and H4K12la) in a p300/HDAC1/HDAC3-dependent manner. H3K18la and H4K12la are enriched in the promoters of 75 target genes, of which the H3K18la and H4K12la modification upregulated the expression of ribosomal protein S6 kinases 2 (S6K2) in the virus-infected hosts to promote the virus infection. Further data revealed that the virus-encoded miR-N20 targeted hypoxia inducible factor-1α (HIF-1α) to inhibit the host glycolysis, leading to the suppression of H3K18la and H4K12la. Therefore, the findings contributed novel insights into the effects and the underlying mechanism of the virus-induced histone lactylation on the virus-host interactions, providing new targets for the control of virus infection.
Collapse
Affiliation(s)
- Yu Zhang
- College of Life SciencesZhejiang UniversityHangzhou310058P. R. China
- Department of Clinical PharmacologyKey Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang ProvinceAffiliated Hangzhou First People's HospitalCancer CenterWestlake University School of MedicineHangzhou310006P. R. China
| | - Xiaobo Zhang
- College of Life SciencesZhejiang UniversityHangzhou310058P. R. China
- Laboratory for Marine Biology and Biotechnology of Pilot National Laboratory for Marine Science and Technology (Qingdao)Qingdao266003P. R. China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Zhuhai519000P. R. China
| |
Collapse
|
5
|
Hafner A, Meurs N, Garner A, Azar E, Kannan A, Passalacqua KD, Nagrath D, Wobus CE. Norovirus NS1/2 protein increases glutaminolysis for efficient viral replication. PLoS Pathog 2024; 20:e1011909. [PMID: 38976719 PMCID: PMC11257395 DOI: 10.1371/journal.ppat.1011909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/18/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Viruses are obligate intracellular parasites that rely on host cell metabolism for successful replication. Thus, viruses rewire host cell pathways involved in central carbon metabolism to increase the availability of building blocks for successful propagation. However, the underlying mechanisms of virus-induced alterations to host metabolism are largely unknown. Noroviruses (NoVs) are highly prevalent pathogens that cause sporadic and epidemic viral gastroenteritis. In the present study, we uncovered several strain-specific and shared host cell metabolic requirements of three murine norovirus (MNV) strains, MNV-1, CR3, and CR6. While all three strains required glycolysis, glutaminolysis, and the pentose phosphate pathway for optimal infection of macrophages, only MNV-1 relied on host oxidative phosphorylation. Furthermore, the first metabolic flux analysis of NoV-infected cells revealed that both glycolysis and glutaminolysis are upregulated during MNV-1 infection of macrophages. Glutamine deprivation affected the viral lifecycle at the stage of genome replication, resulting in decreased non-structural and structural protein synthesis, viral assembly, and egress. Mechanistic studies further showed that MNV infection and overexpression of the non-structural protein NS1/2 increased the enzymatic activity of the rate-limiting enzyme glutaminase. In conclusion, the inaugural investigation of NoV-induced alterations to host glutaminolysis identified NS1/2 as the first viral molecule for RNA viruses that regulates glutaminolysis either directly or indirectly. This increases our fundamental understanding of virus-induced metabolic alterations and may lead to improvements in the cultivation of human NoVs.
Collapse
Affiliation(s)
- Adam Hafner
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Noah Meurs
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ari Garner
- Department of Microbiology, Immunology, and Inflammation, University of Illinois, Chicago, Illinois, United States of America
| | - Elaine Azar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Aditya Kannan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Karla D. Passalacqua
- Graduate Medical Education, Henry Ford Health, Detroit, Michigan, United States of America
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
6
|
Yasir M, Park J, Chun W. Discovery of Novel Aldose Reductase Inhibitors via the Integration of Ligand-Based and Structure-Based Virtual Screening with Experimental Validation. ACS OMEGA 2024; 9:20338-20349. [PMID: 38737046 PMCID: PMC11079907 DOI: 10.1021/acsomega.4c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
Aldose reductase plays a central role in diabetes mellitus (DM) associated complications by converting glucose to sorbitol, resulting in a harmful increase of reactive oxygen species (ROS) in various tissues, such as the heart, vasculature, neurons, eyes, and kidneys. We employed a comprehensive approach, integrating both ligand- and structure-based virtual screening followed by experimental validation. Initially, candidate compounds were extracted from extensive drug and chemical libraries using the DeepChem's GraphConvMol algorithm, leveraging its capacity for robust molecular feature representation. Subsequent refinement employed molecular docking and molecular dynamics (MD) simulations, which are crucial for understanding compound-receptor interactions and dynamic behavior in a simulated physiological environment. Finally, the candidate compounds were subjected to experimental validation of their biological activity using an aldose reductase inhibitor screening kit. The comprehensive approach led to the identification of a promising compound, demonstrating significant potential as an aldose reductase inhibitor. This comprehensive approach not only yields a potential therapeutic intervention for DM-related complications but also establishes an integrated protocol for drug development, setting a new benchmark in the field.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Republic of Korea
| |
Collapse
|
7
|
Cui C, Tang X, Xing J, Sheng X, Chi H, Zhan W. Single-cell RNA-seq revealed heterogeneous responses and functional differentiation of hemocytes against white spot syndrome virus infection in Litopenaeus vannamei. J Virol 2024; 98:e0180523. [PMID: 38323810 PMCID: PMC10949519 DOI: 10.1128/jvi.01805-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Shrimp hemocytes are the vital immune cells participating in innate immune response to defend against viruses. However, the lack of specific molecular markers for shrimp hemocyte hindered the insightful understanding of their functional clusters and differential roles in combating microbial infections. In this study, we used single-cell RNA sequencing to map the transcriptomic landscape of hemocytes from the white spot syndrome virus (WSSV)-infected Litopenaeus vannamei and conjointly analyzed with our previous published single-cell RNA sequencing technology data from the healthy hemocytes. A total of 16 transcriptionally distinct cell clusters were identified, which occupied different proportions in healthy and WSSV-infected hemocytes and exerted differential roles in antiviral immune response. Following mapping of the sequencing data to the WSSV genome, we found that all types of hemocytes could be invaded by WSSV virions, especially the cluster 8, which showed the highest transcriptional levels of WSSV genes and exhibited a cell type-specific antiviral response to the viral infection. Further evaluation of the cell clusters revealed the delicate dynamic balance between hemocyte immune response and viral infestation. Unsupervised pseudo-time analysis of hemocytes showed that the hemocytes in immune-resting state could be significantly activated upon WSSV infection and then functionally differentiated to different hemocyte subsets. Collectively, our results revealed the differential responses of shrimp hemocytes and the process of immune-functional differentiation post-WSSV infection, providing essential resource for the systematic insight into the synergistic immune response mechanism against viral infection among hemocyte subtypes. IMPORTANCE Current knowledge of shrimp hemocyte classification mainly comes from morphology, which hinder in-depth characterization of cell lineage development, functional differentiation, and different immune response of hemocyte types during pathogenic infections. Here, single-cell RNA sequencing was used for mapping hemocytes during white spot syndrome virus (WSSV) infection in Litopenaeus vannamei, identifying 16 cell clusters and evaluating their potential antiviral functional characteristics. We have described the dynamic balance between viral infestation and hemocyte immunity. And the functional differentiation of hemocytes under WSSV stimulation was further characterized. Our results provided a comprehensive transcriptional landscape and revealed the heterogeneous immune response in shrimp hemocytes during WSSV infection.
Collapse
Affiliation(s)
- Chuang Cui
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
8
|
Dong F, Wang X, Li J, Zhao D, Li J. Causal relationship between lactate dehydrogenase and risk of developing ischemic stroke: A Mendelian randomized study. Brain Behav 2024; 14:e3352. [PMID: 38376049 PMCID: PMC10757901 DOI: 10.1002/brb3.3352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/31/2023] [Accepted: 11/26/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Ischemic stroke (IS) is one of the major global health problems. It is not clear whether there is a causal relationship between lactate dehydrogenase (LDH) and the risk of IS attacks. The purpose of this study was to investigate whether LDH has a causal relationship with the development of IS. METHODS The genome-wide association data of LDH and IS were obtained through a Mendelian randomization-based platform. Single nucleotide polymorphisms (SNP) that were significantly associated with LDH were identified and used as instrumental variables, and a two-sample Mendelian randomization study was used to examine the causal relationship between LDH and IS. The statistical methods included Inverse-variance weighted approach, MR-Egger regression, and weighted median estimator. RESULTS We selected 15 SNPs of genome-wide significance from Genome-wide association study database with LDH as instrumental variables. A consistent causal association between LDH and IS was observed by different assessment methods. The results of the inverse-variance weighted method suggested an inverse association between LDH and higher genetic predictability of IS risk (OR, 0.997; 95%CI 0.995-0.999). The weighted median estimate showed consistent results with the MR-Egger method (weighted median estimate: OR, 0.995; 95%CI 0.992-0.999; MR-Egger method: OR, 0.996; 95%CI 0.992-0.999). The inverse-variance weighted method indicates a causal association between LDH and IS (β = -0.002563, SE = 0.00128, p = .0453). MR-Egger analysis (β = -0.004498, SE = 0.001877, p = .03) and the weighted median method suggested that LDH and IS also existed causal relationship (β = -0.004861, SE = 0.001801, p = .00695). CONCLUSIONS Our Mendelian randomization results suggest that LDH is inversely associated with the risk of developing IS, and are contrary to the results of previous observational studies.
Collapse
Affiliation(s)
- Fuxiang Dong
- College of Traditional Chinese MedicineChangchun University of Chinese MedicineChangchunJilinChina
| | - Xu Wang
- College of Traditional Chinese MedicineChangchun University of Chinese MedicineChangchunJilinChina
- School of Public HealthJilin UniversityChangchunJilinChina
| | - Jinjian Li
- College of Traditional Chinese MedicineChangchun University of Chinese MedicineChangchunJilinChina
| | - Dexi Zhao
- College of Traditional Chinese MedicineChangchun University of Chinese MedicineChangchunJilinChina
| | - Jinhua Li
- School of Public HealthJilin UniversityChangchunJilinChina
| |
Collapse
|
9
|
Hafner A, Meurs N, Garner A, Azar E, Passalacqua KD, Nagrath D, Wobus CE. Norovirus NS1/2 protein increases glutaminolysis for efficient viral replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572316. [PMID: 38187600 PMCID: PMC10769279 DOI: 10.1101/2023.12.19.572316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Viruses are obligate intracellular parasites that rely on host cell metabolism for successful replication. Thus, viruses rewire host cell pathways involved in central carbon metabolism to increase the availability of building blocks for replication. However, the underlying mechanisms of virus-induced alterations to host metabolism are largely unknown. Noroviruses (NoVs) are highly prevalent pathogens that cause sporadic and epidemic viral gastroenteritis. In the present study, we uncovered several strain-specific and shared host cell metabolic requirements of three murine norovirus (MNV) strains, the acute MNV-1 strain and the persistent CR3 and CR6 strains. While all three strains required glycolysis, glutaminolysis, and the pentose phosphate pathway for optimal infection of macrophages, only MNV-1 relied on host oxidative phosphorylation. Furthermore, the first metabolic flux analysis of NoV-infected cells revealed that both glycolysis and glutaminolysis are upregulated during MNV-1 infection of macrophages. Glutamine deprivation affected the MNV lifecycle at the stage of genome replication, resulting in decreased non-structural and structural protein synthesis, viral assembly, and egress. Mechanistic studies further showed that MNV infection and overexpression of the MNV non-structural protein NS1/2 increased the enzymatic activity of the rate-limiting enzyme glutaminase. In conclusion, the inaugural investigation of NoV-induced alterations to host glutaminolysis identified the first viral regulator of glutaminolysis for RNA viruses, which increases our fundamental understanding of virus-induced metabolic alterations.
Collapse
Affiliation(s)
- Adam Hafner
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Meurs
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ari Garner
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Elaine Azar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Cao Z, Chen C, Wang C, Li T, Chang L, Si L, Yan D. Enterocytozoon hepatopenaei (EHP) Infection Alters the Metabolic Processes and Induces Oxidative Stress in Penaeus vannamei. Animals (Basel) 2023; 13:3661. [PMID: 38067012 PMCID: PMC10705197 DOI: 10.3390/ani13233661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 09/10/2024] Open
Abstract
Enterocytozoon hepatopenaei (EHP) is highly contagious and can cause hepatopancreatic microsporidiosis (HPM), which is typically characterized by the slow growth of shrimp. In this study, the differences in histology, metabolism, oxidative stress and growth between healthy and EHP-infected Penaeus vannamei were analyzed using an EHP challenge experiment. Histology showed that EHP caused lesions in the hepatic tubules of P. vannamei, such as hepatic tubular atrophy and epithelial cell shedding, with mature spores. Meanwhile, white feces may appear when the infection is severe. Furthermore, the content of total protein, glycogen, ATP and glucose in the EHP challenge group was significantly reduced. The qPCR results showed that EHP infection changed the expression of key genes in glucose metabolism, among which hexokinase (HK), phosphofructokinase (PFK), pyruvatekinase (PK), citrate synthase (CS) and isocitric dehydrogenase (IDH) were significantly down-regulated, while phosphoenolpyruvate carboxykinase (PEPCK), fructose bisphosphatase (FBP) and glucose-6-phosphatase (G6P) were significantly up-regulated. Obviously, the expression of growth-related genes was disordered. Simultaneously, the antioxidant genes manganese superoxide dismutase (MnSOD), catalase (CAT), glutathione peroxidase (GPX), glutathione-S-transferases (GST) and nuclear factor E2-related factor2 (Nrf2) were up-regulated to varying degrees in the EHP challenge group, and EHP infection induced significant increases in the oxidative damage products lipid peroxide (LPO) and malondialdehyde (MDA). Ultimately, the shrimp weight of the challenge group was 6.85 ± 0.86 g, which was significantly lower than that of the control group (8.95 ± 0.75 g). Taken together, we speculate that EHP changes the substance metabolism and growth process by causing oxidative damage to the hepatopancreas, which may lead to the growth retardation of P. vannamei.
Collapse
Affiliation(s)
| | | | | | | | | | - Lingjun Si
- Laboratory of Disease Research of Aquatic Animal, School of Agriculture, Ludong University, Yantai 264025, China; (Z.C.); (C.C.); (C.W.); (T.L.); (L.C.)
| | - Dongchun Yan
- Laboratory of Disease Research of Aquatic Animal, School of Agriculture, Ludong University, Yantai 264025, China; (Z.C.); (C.C.); (C.W.); (T.L.); (L.C.)
| |
Collapse
|
11
|
Moraes B, Martins R, Lopes C, Martins R, Arcanjo A, Nascimento J, Konnai S, da Silva Vaz I, Logullo C. G6PDH as a key immunometabolic and redox trigger in arthropods. Front Physiol 2023; 14:1287090. [PMID: 38046951 PMCID: PMC10693429 DOI: 10.3389/fphys.2023.1287090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
The enzyme glucose-6-phosphate dehydrogenase (G6PDH) plays crucial roles in glucose homeostasis and the pentose phosphate pathway (PPP), being also involved in redox metabolism. The PPP is an important metabolic pathway that produces ribose and nicotinamide adenine dinucleotide phosphate (NADPH), which are essential for several physiologic and biochemical processes, such as the synthesis of fatty acids and nucleic acids. As a rate-limiting step in PPP, G6PDH is a highly conserved enzyme and its deficiency can lead to severe consequences for the organism, in particular for cell growth. Insufficient G6PDH activity can lead to cell growth arrest, impaired embryonic development, as well as a reduction in insulin sensitivity, inflammation, diabetes, and hypertension. While research on G6PDH and PPP has historically focused on mammalian models, particularly human disorders, recent studies have shed light on the regulation of this enzyme in arthropods, where new functions were discovered. This review will discuss the role of arthropod G6PDH in regulating redox homeostasis and immunometabolism and explore potential avenues for further research on this enzyme in various metabolic adaptations.
Collapse
Affiliation(s)
- Bruno Moraes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Renato Martins
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Cintia Lopes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Ronald Martins
- Programa de Computação Científica, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
| | - Angélica Arcanjo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Jhenifer Nascimento
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Satoru Konnai
- Laboratory of Infectious Diseases, Hokkaido University, Sapporo, Japan
| | - Itabajara da Silva Vaz
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
- Centro de Biotecnologia and Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos Logullo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Zhong Y, He Z, Long X, Hou D, Hu X, Sun C. Transcriptome analysis of Fenneropenaeus merguiensis in response to Vibrio proteolyticus infection. JOURNAL OF FISH DISEASES 2023; 46:1207-1224. [PMID: 37589383 DOI: 10.1111/jfd.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
In recent years, due to the destruction of the culture environment and serious ecological pressure, especially in the process of culture, residual bait, faeces and fishery drug abuse will lead to the accumulation of harmful metabolites such as ammonia nitrogen and nitrite, and biological denitrification is the most economical and effective method to remove the single. Therefore, in this study, a nitrite removal strain XA19 was isolated and screened from a shrimp biofloc culture pond. This strain was identified as a clade of Vibrio proteolyticus because the homology between XA19 and V. proteolyticus WDVP was as high as 99.86% by using 16S rDNA gene sequence analysis and NCBI database comparison. Scanning electron microscopy images showed that V. proteolyticus is short-rod-shaped with a curved body and no budding spores, pods and flagella. Antimicrobial susceptibility test proved that V. proteolyticus was resistant to ampicillin, oxacillin, penicillin, vancomycin and clindamycin. In the median lethal concentration 50 (LC50 ) test, at 7-day post-infection (dpi), LC50 of V. proteolyticus for Fenneropenaeus merguiensis was 1.69 × 104 CFU/mL. Transcriptome sequencing analysis was carried out on hepatopancreas of F. merguiensis at 24 and 48 hpi. A total of 176 differentially expressed genes (DEGs) were screened at 24 hpi, including 104 up-regulated DEGs and 72 down-regulated DEGs, and a total of 52 DEGs were screened at 48 hpi, including 32 up-regulated DEGs and 20 down-regulated DEGs. In the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs, many immune-related signalling pathways were significantly enriched, including Hippo signalling pathway, phagosome, Toll and Imd signalling pathways and Wnt signalling pathway. In addition, some pathways related to Warburg effect were also enriched, including Glycolysis/Gluconeogenesis, Biosynthesis of amino acids, amino sugar and nucleotide sugar metabolism and so on. In this study, the toxicity and drug sensitivity of V. proteolyticus were systematically studied, and the immune response of hepatopancreas of F. merguiensis to V. proteolyticus infection was preliminarily revealed from the molecular level. The results may provide a reference for the prevention and control of V. proteolyticus.
Collapse
Affiliation(s)
- Yunqi Zhong
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Zihao He
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Xinxin Long
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Danqing Hou
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Xianye Hu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Chengbo Sun
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| |
Collapse
|
13
|
Alam MS, Islam MN, Das M, Islam SF, Rabbane MG, Karim E, Roy A, Alam MS, Ahmed R, Kibria ASM. RNAi-Based Therapy: Combating Shrimp Viral Diseases. Viruses 2023; 15:2050. [PMID: 37896827 PMCID: PMC10612085 DOI: 10.3390/v15102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/18/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Shrimp aquaculture has become a vital industry, meeting the growing global demand for seafood. Shrimp viral diseases have posed significant challenges to the aquaculture industry, causing major economic losses worldwide. Conventional treatment methods have proven to be ineffective in controlling these diseases. However, recent advances in RNA interference (RNAi) technology have opened new possibilities for combating shrimp viral diseases. This cutting-edge technology uses cellular machinery to silence specific viral genes, preventing viral replication and spread. Numerous studies have shown the effectiveness of RNAi-based therapies in various model organisms, paving the way for their use in shrimp health. By precisely targeting viral pathogens, RNAi has the potential to provide a sustainable and environmentally friendly solution to combat viral diseases in shrimp aquaculture. This review paper provides an overview of RNAi-based therapy and its potential as a game-changer for shrimp viral diseases. We discuss the principles of RNAi, its application in combating viral infections, and the current progress made in RNAi-based therapy for shrimp viral diseases. We also address the challenges and prospects of this innovative approach.
Collapse
Affiliation(s)
- Md. Shahanoor Alam
- Department of Genetics and Fish Breeding, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | - Mohammad Nazrul Islam
- Department of Biotechnology, Sher-e-Bangla Agricultural University, Dhaka 1207, Bangladesh;
| | - Mousumi Das
- Department of Aquaculture, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | - Sk. Farzana Islam
- Department of Fisheries (DoF), Government of the People’s Republic of Bangladesh, Matshya Bhaban, Ramna, Dhaka 1000, Bangladesh; (S.F.I.); (R.A.)
| | - Md. Golam Rabbane
- Department of Fisheries, Faculty of Biological Sciences, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Ehsanul Karim
- Bangladesh Fisheries Research Institute, Mymensingh 2201, Bangladesh;
| | - Animesh Roy
- Department of Fisheries Biology and Aquatic Environment, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | - Mohammad Shafiqul Alam
- Department of Genetics and Fish Breeding, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur 1706, Bangladesh;
| | - Raju Ahmed
- Department of Fisheries (DoF), Government of the People’s Republic of Bangladesh, Matshya Bhaban, Ramna, Dhaka 1000, Bangladesh; (S.F.I.); (R.A.)
| | - Abu Syed Md. Kibria
- Department of Aquaculture, Hajee Mohammad Danesh Science and Technology University, Dinajpur 5200, Bangladesh;
| |
Collapse
|
14
|
Zhang P, Fu HJ, Lv LX, Liu CF, Han C, Zhao XF, Wang JX. WSSV exploits AMPK to activate mTORC2 signaling for proliferation by enhancing aerobic glycolysis. Commun Biol 2023; 6:361. [PMID: 37012372 PMCID: PMC10070494 DOI: 10.1038/s42003-023-04735-z] [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: 01/13/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
AMPK plays significant roles in the modulation of metabolic reprogramming and viral infection. However, the detailed mechanism by which AMPK affects viral infection is unclear. The present study aims to determine how AMPK influences white spot syndrome virus (WSSV) infection in shrimp (Marsupenaeus japonicus). Here, we find that AMPK expression and phosphorylation are significantly upregulated in WSSV-infected shrimp. WSSV replication decreases remarkably after knockdown of Ampkα and the shrimp survival rate of AMPK-inhibitor injection shrimp increases significantly, suggesting that AMPK is beneficial for WSSV proliferation. Mechanistically, WSSV infection increases intracellular Ca2+ level, and activates CaMKK, which result in AMPK phosphorylation and partial nuclear translocation. AMPK directly activates mTORC2-AKT signaling pathway to phosphorylate key enzymes of glycolysis in the cytosol and promotes expression of Hif1α to mediate transcription of key glycolytic enzyme genes, both of which lead to increased glycolysis to provide energy for WSSV proliferation. Our findings reveal a novel mechanism by which WSSV exploits the host CaMKK-AMPK-mTORC2 pathway for its proliferation, and suggest that AMPK might be a target for WSSV control in shrimp aquaculture.
Collapse
Affiliation(s)
- Peng Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Hai-Jing Fu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Li-Xia Lv
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Chen-Fei Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Chang Han
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China.
- State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, Shandong, China.
| |
Collapse
|
15
|
Qian Y, Yang Y, Qing W, Li C, Kong M, Kang Z, Zuo Y, Wu J, Yu M, Yang Z. Coxsackievirus B3 infection induces glycolysis to facilitate viral replication. Front Microbiol 2022; 13:962766. [PMID: 36569097 PMCID: PMC9780277 DOI: 10.3389/fmicb.2022.962766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is a leading cause of viral myocarditis, but no effective treatment strategy against CVB3 is available. Viruses lack an inherent metabolic system and thus depend on host cellular metabolism for their benefit. In this study, we observed that CVB3 enhanced glycolysis in H9c2 rat cardiomyocytes and HL-1 mouse cardiomyocytes. Therefore, three key glycolytic enzymes, namely, hexokinase 2 (HK2), muscle phosphofructokinase (PFKM), and pyruvate kinase M2 (PKM2), were measured in CVB3-infected H9c2 and HL-1 cells. Expression levels of HK2 and PFKM, but not PKM2, were increased in CVB3-infected H9c2 cells. All three key glycolytic enzymes showed elevated expression in CVB3-infected HL-1 cells. To further investigate this, we used 2 deoxyglucose, sodium citrate, and shikonin as glycolysis inhibitors for HK2, PFKM, and PKM2, respectively. Glycolysis inhibitors significantly reduced CVB3 replication, while the glycolysis enhancer dramatically promoted it. In addition, glycolysis inhibitors decreased autophagy and accelerated autophagosome degradation. The autophagy inducer eliminated partial inhibition effects of glycolysis inhibitors on CVB3 replication. These results demonstrate that CVB3 infection enhances glycolysis and thus benefits viral replication.
Collapse
Affiliation(s)
- Yujie Qian
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yeyi Yang
- Department of Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wenxiang Qing
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chunyun Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Min Kong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhijuan Kang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuanbojiao Zuo
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiping Wu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Meng Yu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zuocheng Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Zuocheng Yang
| |
Collapse
|
16
|
Xu Y, Xu W, Hu X, Su H, Wen G, Yang K, Cao Y. Toxicity of the microcystin-producing cyanobacteria Microcystis aeruginosa to shrimp Litopenaeus vannamei. ECOTOXICOLOGY (LONDON, ENGLAND) 2022; 31:1403-1412. [PMID: 36223040 DOI: 10.1007/s10646-022-02597-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Microcystis aeruginosa is reported to cause cyanobacterial blooms in shrimp breeding ponds, which can result in significant shrimp mortality. However, the toxic effects of M. aeruginosa on Litopenaeus vannamei are still not completely understood. In this paper, the toxicity of M. aeruginosa cells to L. vannamei was examined, and the toxic components in the cells were analyzed through high-pressure liquid chromatography (HLPC). In addition, the immune response of shrimp to the M. aeruginosa cell extract was assessed by measuring the activity of immune-related enzymes, as well as the transcription of the relevant genes. The results showed that M. aeruginosa cells, extract and cell-free cultured medium resulted in a 100%, 98.3%, and 1.7% mortality rate in shrimp, respectively. HPLC analysis results revealed the presence of microcystin-LR (MC-LR) at a concentration of 190.40 mg/kg of cells. In addition, the activity and gene transcription of two immune related enzymes, SOD and LZM, were both significantly reduced in shrimp hepatopancreas (p < 0.05) after injection with extract. However, reduced glutathione (GSH) content was slightly increased, but the ratio of GSH to GSSG decreased. The transcription of gst gene function as detoxification, was significantly downregulated (p < 0.05). The results demonstrated that M. aeruginosa cell extract was highly toxic to L. vannamei, and exerted a negative effect on shrimp immunity including reduction of antioxidant capacity, antibacterial activity and detoxification activity, due to toxins including microcystin-LR.
Collapse
Affiliation(s)
- Yu Xu
- Sanya Tropical Fisheries Research Institute, 572018, Sanya, China
- Key Laboratory of Efficient Utilization and Processing of Marine Fishery Resources of Hainan Province, 572018, Sanya, China
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Wujie Xu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Xiaojuan Hu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Haochang Su
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Guoliang Wen
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Keng Yang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China
| | - Yucheng Cao
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, China.
| |
Collapse
|
17
|
Zakirova NF, Kondrashova AS, Golikov MV, Ivanova ON, Ivanov AV, Isaguliants MG, Bayurova EO. Expression of HIV-1 Reverse Transcriptase in Murine Cancer Cells Increases Mitochondrial Respiration. Mol Biol 2022; 56:723-734. [DOI: 10.1134/s0026893322050168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 04/17/2025]
|
18
|
Ng YS, Lee DY, Liu CH, Tung CY, He ST, Wang HC. White Spot Syndrome Virus Triggers a Glycolytic Pathway in Shrimp Immune Cells (Hemocytes) to Benefit Its Replication. Front Immunol 2022; 13:901111. [PMID: 35860260 PMCID: PMC9289281 DOI: 10.3389/fimmu.2022.901111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
White spot syndrome virus (WSSV) is the causative agent of a shrimp disease that inflicts in huge economic losses in shrimp-farming industry. WSSV triggers aerobic glycolysis in shrimp immune cells (hemocytes), but how this virus regulates glycolytic enzymes or pathway is yet to be characterized. Therefore, mRNA levels and activity of four important glycolytic enzymes, Hexokinase (HK), Phosphofructokinase (PFK), Pyruvate kinase (PK), and Lactate dehydrogenase (LDH), were measured in WSSV-infected shrimp hemocytes. Gene expression of HK and PFK, but not LDH or PK, was increased at the viral genome replication stage (12 hpi); furthermore, activity of these enzymes, except HK, was concurrently increased. However, there was no increased enzyme activity at the viral late stage (24 hpi). In vivo dsRNA silencing and glycolysis disruption by 2-DG further confirmed the role of glycolysis in virus replication. Based on tracing studies using stable isotope labeled glucose, glycolysis was activated at the viral genome replication stage, but not at the viral late stage. This study demonstrated that WSSV enhanced glycolysis by activating glycolytic enzyme at the viral genome replication stage, providing energy and biomolecules for virus replication.
Collapse
Affiliation(s)
- Yen Siong Ng
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hung Liu
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Cheng-Yi Tung
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ting He
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Han-Ching Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- International Center for the Scientific Development of Shrimp Aquaculture, National Cheng Kung University, Tainan, Taiwan
- *Correspondence: Han-Ching Wang,
| |
Collapse
|
19
|
Tang X, Liu T, Li X, Sheng X, Xing J, Chi H, Zhan W. Protein phosphorylation in hemocytes of Fenneropenaeus chinensis in response to white spot syndrome virus infection. FISH & SHELLFISH IMMUNOLOGY 2022; 122:106-114. [PMID: 35092807 DOI: 10.1016/j.fsi.2022.01.038] [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: 11/08/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Protein phosphorylation and dephosphorylation are the most common and important regulatory mechanisms in signal transduction, which play a vital role in immune defense response. Our previous study has found the level of tyrosine phosphorylation was significantly changed in the hemocytes of Fenneropenaeus chinensis upon white spot syndrome virus (WSSV) infection. In order to explore the relationship between protein phosphorylation and WSSV infection, the quantitative phosphoproteomics was employed to identify differential phosphorylated proteins in hemocytes of F. chinensis before and after WSSV infection, and elucidate the role of key differential phosphorylated proteins in WSSV infection process. The results showed that a total of 147 differential phosphorylated proteins were identified in the hemocytes, including 64 phosphorylated proteins and 83 dephosphorylated proteins, which were mostly enriched in pyruvate metabolism, TCA cycle, glycolysis, and ribosomal biosynthesis. Functional analysis of differential phosphorylated proteins showed that they were involved in cell apoptosis, cell phagocytosis, cell metabolism and antiviral infection. A total of 236 differential phosphorylation sites were found, including 91 modified sites in the phosphorylation proteins and 145 modified sites in the dephosphorylation proteins. Motif analysis showed that these phosphorylation sites could activate mitogen-activated protein kinase, P70 S6 kinase and other kinases in hemocytes. Moveover, the phosphorylation levels of eukaryotic protein initiation factor 4E binding proteins and histone H3 were further determined by ELISA and Western blotting, which both exhibited a significant increase post WSSV infection and reach their peak levels at 6 and 12 h, respectively. Moreover, we found that lactate, a metabolite closely related to pyruvate metabolism, TCA cycle and glycolysis, was significantly increased in the hemocytes after WSSV infection. This study revealed the protein phosphorylation response in hemocytes of F. chinensis to WSSV infection, which help to clarify the response characteristics and virus resistance mechanism of hemocytes in F. chinensis, and also facilitate further understanding of the interaction between WSSV and shrimp hemocytes.
Collapse
Affiliation(s)
- Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| | - Ting Liu
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China
| | - Xiaoai Li
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
20
|
Chen YH, Lian YY, Lu KC, Chen GL, Fan JQ, Li BB, He JG. Litopenaeus vannamei Sma and Mad related protein 5 gene is involved in stress response and white spot syndrome virus infection. FISH & SHELLFISH IMMUNOLOGY 2021; 117:104-112. [PMID: 34333126 DOI: 10.1016/j.fsi.2021.07.021] [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: 07/04/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 06/13/2023]
Abstract
Cell survival is based on the stability of intracellular state. It was well known that biochemical reactions in cells require specific intracellular environments, such as pH and calcium concentration. While the mechanism of stabilizing the intracellular environment is complex and far from clear. In this study, a Sma and Mad related protein 5 gene (LvSmad5) of Litopenaeus vannamei was cloned. LvSmad5 was located to both cytoplasm and nucleus. And subcellular localization of LvSmad5 was responsed to the changing of cells internal and external environment. Besides, it was found that subcellular localization of LvSmad5 was also regulated by unfolded protein response. Moreover, it was proved that nucleic localization of LvSmad5 could significantly increase the white spot syndrome virus (WSSV) infection in shrimp, and knockdown expression of LvSmad5 decreased the cumulative mortality of WSSV infection shrimp. Further investigation revealed that cytoplasm LvSmad5 could interplay with shrimp hexokinase 1, and contribute to glycolysis. These results indicated that LvSmad5 played a role in L. vannamei environmental stress response, and was used by WSSV for its replication.
Collapse
Affiliation(s)
- Yi-Hong Chen
- Institute of Modern Aquaculture Science and Engineering (IMASE) / Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), 519000, China.
| | - Yu-Ying Lian
- State Key Laboratory for Biocontrol/School of Life Sciences, SunYat-senUniversity, 135 Xingang Road West, Guangzhou, 510275, PR China
| | - Ke-Cheng Lu
- Institute of Modern Aquaculture Science and Engineering (IMASE) / Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Guo-Lian Chen
- Institute of Modern Aquaculture Science and Engineering (IMASE) / Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Jin-Quan Fan
- Institute of Modern Aquaculture Science and Engineering (IMASE) / Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Bin-Bin Li
- Institute of Modern Aquaculture Science and Engineering (IMASE) / Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou, 510631, PR China
| | - Jian-Guo He
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), 519000, China; State Key Laboratory for Biocontrol/School of Life Sciences, SunYat-senUniversity, 135 Xingang Road West, Guangzhou, 510275, PR China.
| |
Collapse
|
21
|
Wang C, Ruan L, Shi H, Lin W, Liu L, Li S. Phosphorylation of Shrimp Tcf by a Viral Protein Kinase WSV083 Suppresses Its Antiviral Effect. Front Immunol 2021; 12:698697. [PMID: 34408747 PMCID: PMC8365339 DOI: 10.3389/fimmu.2021.698697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Nuclear DNA-binding TCF proteins, which act as the main downstream effectors of Wnt signaling, are essential for the regulation of cell fate and innate immunity. However, their role during viral infection in shrimp remains unknown. Herein, we demonstrated that Litopenaeus vannamei TCF (LvTcf) acts independently of Lvβ-catenin to promote interferon-like protein LvVago1 production, thus mounting the response to WSSV infection. Further, we observed that WSV083, a WSSV serine/threonine protein kinase, bound to LvTcf and phosphorylated it. Phosphorylated LvTcf was then recognized and degraded via the ubiquitin-proteasome pathway. Moreover, mass spectrometry analyses indicated that the T39 and T104 residues of LvTcf were target sites phosphorylated by WSV083. Point mutation analyses suggested that additional sites of LvTcf may undergo phosphorylation via WSV083. Taken together, the current work provides valuable insights into host immunity and viral pathogenesis. LvTcf is not only a modulator of shrimp innate immunity but is also an important target for WSSV immune evasion. Thus, the current findings will help improve disease control in shrimps.
Collapse
Affiliation(s)
- Chuanqi Wang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Lingwei Ruan
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Hong Shi
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Wenyang Lin
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Linmin Liu
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Sujie Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| |
Collapse
|
22
|
Immunometabolism Modulation in Therapy. Biomedicines 2021; 9:biomedicines9070798. [PMID: 34356862 PMCID: PMC8301471 DOI: 10.3390/biomedicines9070798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
The study of cancer biology should be based around a comprehensive vision of the entire tumor ecosystem, considering the functional, bioenergetic and metabolic state of tumor cells and those of their microenvironment, and placing particular importance on immune system cells. Enhanced understanding of the molecular bases that give rise to alterations of pathways related to tumor development can open up new therapeutic intervention opportunities, such as metabolic regulation applied to immunotherapy. This review outlines the role of various oncometabolites and immunometabolites, such as TCA intermediates, in shaping pro/anti-inflammatory activity of immune cells such as MDSCs, T lymphocytes, TAMs and DCs in cancer. We also discuss the extraordinary plasticity of the immune response and its implication in immunotherapy efficacy, and highlight different therapeutic intervention possibilities based on controlling the balanced systems of specific metabolites with antagonistic functions.
Collapse
|
23
|
Millard RS, Bickley LK, Bateman KS, Farbos A, Minardi D, Moore K, Ross SH, Stentiford GD, Tyler CR, van Aerle R, Santos EM. Global mRNA and miRNA Analysis Reveal Key Processes in the Initial Response to Infection with WSSV in the Pacific Whiteleg Shrimp. Viruses 2021; 13:v13061140. [PMID: 34199268 PMCID: PMC8231841 DOI: 10.3390/v13061140] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
White Spot Disease (WSD) presents a major barrier to penaeid shrimp production. Mechanisms underlying White Spot Syndrome Virus (WSSV) susceptibility in penaeids are poorly understood due to limited information related to early infection. We investigated mRNA and miRNA transcription in Penaeus vannamei over 36 h following infection. Over this time course, 6192 transcripts and 27 miRNAs were differentially expressed—with limited differential expression from 3–12 h post injection (hpi) and a more significant transcriptional response associated with the onset of disease symptoms (24 hpi). During early infection, regulated processes included cytoskeletal remodelling and alterations in phagocytic activity that may assist WSSV entry and translocation, novel miRNA-induced metabolic shifts, and the downregulation of ATP-dependent proton transporter subunits that may impair cellular recycling. During later infection, uncoupling of the electron transport chain may drive cellular dysfunction and lead to high mortalities in infected penaeids. We propose that post-transcriptional silencing of the immune priming gene Dscam (downregulated following infections) by a novel shrimp miRNA (Pva-pmiR-78; upregulated) as a potential mechanism preventing future recognition of WSSV that may be suppressed in surviving shrimp. Our findings improve our understanding of WSD pathogenesis in P. vannamei and provide potential avenues for future development of prophylactics and treatments.
Collapse
Affiliation(s)
- Rebecca S. Millard
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK; (L.K.B.); (C.R.T.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Correspondence: (R.S.M.); (E.M.S.); Tel.: +44-(0)-1392-724607 (E.M.S.)
| | - Lisa K. Bickley
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK; (L.K.B.); (C.R.T.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
| | - Kelly S. Bateman
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Cefas Weymouth Laboratory, International Centre of Excellence for Aquatic Animal Health, Weymouth DT4 8UB, UK;
| | - Audrey Farbos
- Exeter Sequencing Service, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK; (A.F.); (K.M.)
| | - Diana Minardi
- Cefas Weymouth Laboratory, International Centre of Excellence for Aquatic Animal Health, Weymouth DT4 8UB, UK;
| | - Karen Moore
- Exeter Sequencing Service, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK; (A.F.); (K.M.)
| | - Stuart H. Ross
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Cefas Weymouth Laboratory, International Centre of Excellence for Aquatic Animal Health, Weymouth DT4 8UB, UK;
| | - Grant D. Stentiford
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Cefas Weymouth Laboratory, International Centre of Excellence for Aquatic Animal Health, Weymouth DT4 8UB, UK;
| | - Charles R. Tyler
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK; (L.K.B.); (C.R.T.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
| | - Ronny van Aerle
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Cefas Weymouth Laboratory, International Centre of Excellence for Aquatic Animal Health, Weymouth DT4 8UB, UK;
| | - Eduarda M. Santos
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX4 4QD, UK; (L.K.B.); (C.R.T.)
- Centre for Sustainable Aquaculture Futures, University of Exeter, Exeter EX4 4QD, UK; (K.S.B.); (S.H.R.); (G.D.S.); (R.v.A.)
- Correspondence: (R.S.M.); (E.M.S.); Tel.: +44-(0)-1392-724607 (E.M.S.)
| |
Collapse
|
24
|
Alfaro AC, Nguyen TV, Bayot B, Rodriguez Leon JA, Domínguez-Borbor C, Sonnenholzner S. Metabolic responses of whiteleg shrimp to white spot syndrome virus (WSSV). J Invertebr Pathol 2021; 180:107545. [PMID: 33571511 DOI: 10.1016/j.jip.2021.107545] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 01/12/2023]
Abstract
Outbreaks of white spot syndrome virus (WSSV) have caused serious damage to penaeid shrimp aquaculture worldwide. Despite great efforts to characterize the virus, the conditions that lead to infection and the infection mechanisms, there is still a lack of understanding regarding these complex virus-host interactions, which is needed to develop consistent and effective treatment methods for WSSV. In this study, we used a gas chromatography - mass spectrometry (GC-MS)-based metabolomics approach to compare the metabolite profiles of gills, haemolymph and hepatopancreas from whiteleg shrimp (Penaeus vannamei) exposed to WSSV and corresponding controls. The results revealed clear discriminations between metabolite profiles of WSSV-challenged shrimp and controlled shrimp in each tissue. The responses of shrimp gills to WSSV infection were characterized by increases of many fatty acids and amino acids in WSSV-challenged shrimp compared to the controls. Changes in haemolymph metabolite profiles include the increased levels of itaconic acid, energy-related metabolites, metabolites in glutathione cycle and decrease of amino acids. The WSSV challenge led to the decreases of several fatty acids and amino acids and increases of other amino acids, lactic acid and other organic compounds (levulinic acid, malonic acid and putrescine) in hepatopancreas. These alterations of shrimp metabolites suggest several immune responses of shrimp to WSSV in a tissue-specific manner, including upregulation of osmoregulation, antimicrobial activity, metabolic rate, gluconeogenesis, glutathione pathway in control of oxidative stress and shift from aerobic to anaerobic metabolism in shrimp which indicates the Warburg effect. The findings from this study provide a better understanding of molecular process of shrimp response against WSSV invasion which may be useful for development of disease management strategies.
Collapse
Affiliation(s)
- Andrea C Alfaro
- Aquaculture Biotechnology Research Group, School of Science, Auckland University of Technology, Auckland, New Zealand.
| | - Thao V Nguyen
- Aquaculture Biotechnology Research Group, School of Science, Auckland University of Technology, Auckland, New Zealand; NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Bonny Bayot
- Escuela Superior Politécnica del Litoral, ESPOL, Centro Nacional de Acuicultura e Investigaciones Marinas, CENAIM, Campus Gustavo Galindo Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| | - Jenny A Rodriguez Leon
- Escuela Superior Politécnica del Litoral, ESPOL, Centro Nacional de Acuicultura e Investigaciones Marinas, CENAIM, Campus Gustavo Galindo Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| | - Cristóbal Domínguez-Borbor
- Escuela Superior Politécnica del Litoral, ESPOL, Centro Nacional de Acuicultura e Investigaciones Marinas, CENAIM, Campus Gustavo Galindo Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| | - Stanislaus Sonnenholzner
- Escuela Superior Politécnica del Litoral, ESPOL, Centro Nacional de Acuicultura e Investigaciones Marinas, CENAIM, Campus Gustavo Galindo Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| |
Collapse
|
25
|
Yi C, Lv X, Chen D, Sun B, Guo L, Wang S, Ru Y, Wang H, Zeng Q. Transcriptome analysis of the Macrobrachium nipponense hepatopancreas provides insights into immunoregulation under Aeromonas veronii infection. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111503. [PMID: 33120268 DOI: 10.1016/j.ecoenv.2020.111503] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 06/11/2023]
Abstract
The oriental river prawn Macrobrachium nipponense is a commercially important freshwater shrimp that is widely farmed in China. Aeromonas veronii is a conditional pathogen of farmed shrimp, which has caused huge economic losses to the industry. Therefore, there is urgency to study the host-pathogen interactions between M. nipponense and A. veronii to screen individuals with antimicrobial resistance. In this study, we examined the hepatopancreas of moribund M. nipponense infected with A. veronii and healthy individuals at both the histopathological and transcriptomic levels. We showed that A. veronii infection resulted in tubular necrosis of the M. nipponense hepatopancreas. Such changes likely affect assimilation, storage, and excretion by the hepatopancreas, which could ultimately affect the survival and growth of infected individuals. Among the 61,345 unigenes obtained through RNA sequencing and de novo transcriptome assembly, 232 were differentially expressed between the two groups. KEGG and GO analyses revealed that these differentially expressed genes were implicated in pathways, including PPAR, PI3K/AKT, and AMPK signaling. The results of this study will contribute to an analysis of the immune response of M. nipponense to A. veronii infection at the transcriptomic level. Furthermore, the RNA-seq data generated here provide an important genomic resource for research on M. nipponense in the absence of a reference genome.
Collapse
Affiliation(s)
- Cao Yi
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Xiaoting Lv
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Duanduan Chen
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Bing Sun
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Leifeng Guo
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Shouquan Wang
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Yuanyuan Ru
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Hui Wang
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China.
| | - Qifan Zeng
- Ministry of Education, Key Laboratory of Marine Genetics and Breeding, College of Marine Science, Ocean University of China, Qingdao 266003, Shandong, China.
| |
Collapse
|
26
|
Romano A, Casazza M, Gonella F. Addressing Non-linear System Dynamics of Single-Strand RNA Virus-Host Interaction. Front Microbiol 2021; 11:600254. [PMID: 33519741 PMCID: PMC7843927 DOI: 10.3389/fmicb.2020.600254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
Positive single-strand ribonucleic acid [(+)ssRNA] viruses can cause multiple outbreaks, for which comprehensive tailored therapeutic strategies are still missing. Virus and host cell dynamics are tightly connected, generating a complex dynamics that conveys in virion assembly to ensure virus spread in the body. Starting from the knowledge of relevant processes in (+ss)RNA virus replication, transcription, translation, virions budding and shedding, and their respective energy costs, we built up a systems thinking (ST)-based diagram of the virus-host interaction, comprehensive of stocks, flows, and processes as well-described in literature. In ST approach, stocks and flows are expressed by a proxy of the energy embedded and transmitted, respectively, whereas processes are referred to the energy required for the system functioning. In this perspective, healthiness is just a particular configuration, in which stocks relevant for the system (equivalent but not limited to proteins, RNA, DNA, and all metabolites required for the survival) are constant, and the system behavior is stationary. At time of infection, the presence of additional stocks (e.g., viral protein and RNA and all metabolites required for virion assembly and spread) confers a complex network of feedbacks leading to new configurations, which can evolve to maximize the virions stock, thus changing the system structure, output, and purpose. The dynamic trajectories will evolve to achieve a new stationary status, a phenomenon described in microbiology as integration and symbiosis when the system is resilient enough to the changes, or the system may stop functioning and die. Application of external driving forces, acting on processes, can affect the dynamic trajectories adding a further degree of complexity, which can be captured by ST approach, used to address these new configurations. Investigation of system configurations in response to external driving forces acting is developed by computational analysis based on ST diagrams, with the aim at designing novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Romano
- Sezione di Ematologia, Dipartimento di Chirurgia Generale e Specialità Medico Chirurgiche (CHIRMED), Università degli Studi di Catania, Catania, Italy
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Marco Casazza
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Francesco Gonella
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca’ Foscari Venezia, Venezia, Italy
| |
Collapse
|
27
|
Wu W, Dai C, Duan X, Wang C, Lin X, Ke J, Wang Y, Zhang X, Liu H. miRNAs induced by white spot syndrome virus involve in immunity pathways in shrimp Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 93:743-751. [PMID: 31408731 DOI: 10.1016/j.fsi.2019.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 07/04/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023]
Abstract
White shrimp Litopenaeus vannamei are widely cultured in the world and white spot syndrome virus (WSSV) led to huge economic losses in the shrimp industry every year. In the present study, miRNAs involved in the response of shrimp L. vannamei to WSSV infection were obtained through the Illumina HiSeq 2500 high-throughput next-generation sequencing technique. A total number of 7 known miRNAs and 54 putative novel miRNAs were obtained. Among them, 14 DEMs were identified in the shrimp infected with WSSV. The putative target genes of these DEMs were related to host immune response or signaling pathways, indicating the importance of miRNAs in shrimp against WSSV infection. The results will provide information for further research on shrimp response to virus infection and contribute to the development of new strategies for effective protection against WSSV infections.
Collapse
Affiliation(s)
- Wenlin Wu
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Congjie Dai
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xunwei Duan
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Cuifang Wang
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xiaosi Lin
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Jiaying Ke
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Yixuan Wang
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xiaobo Zhang
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou, 362000, China; School of Life Science, Zhejiang University, Hangzhou, 310004, China.
| | - Haipeng Liu
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, 361102, Fujian, China; Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources (Xiamen University), State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen, 361102, Fujian, China.
| |
Collapse
|
28
|
Peruzza L, Shekhar MS, Kumar KV, Swathi A, Karthic K, Hauton C, Vijayan KK. Temporal changes in transcriptome profile provide insights of White Spot Syndrome Virus infection in Litopenaeus vannamei. Sci Rep 2019; 9:13509. [PMID: 31534145 PMCID: PMC6751192 DOI: 10.1038/s41598-019-49836-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/30/2019] [Indexed: 02/08/2023] Open
Abstract
Shrimp aquaculture is severely affected by WSSV. Despite an increasing effort to understand host/virus interaction by characterizing changes in gene expression (GE) following WSSV infection, the majority of published studies have focussed on a single time-point, providing limited insight on the development of host-pathogen interaction over the infection cycle. Using RNA-seq, we contrasted GE in gills of Litopenaeus vannamei at 1.5, 18 and 56 hours-post-infection (hpi), between WSSV-challenged and control shrimps. Time course analysis revealed 5097 differentially expressed genes: 63 DEGs were viral genes and their expression in WSSV group either peaked at 18 hpi (and decreased at 56 hpi) or increased linearly up to 56 hpi, suggesting a different role played by these genes during the course of infection. The remaining DEGs showed that WSSV altered the expression of metabolic, immune, apoptotic and cytoskeletal genes and was able to inhibit NF-κB and JAK/STAT pathways. Interestingly, GE changes were not consistent through the course of infection but were dynamic with time, suggesting the complexity of host-pathogen interaction. These data offer novel insights into the cellular functions that are affected during the course of infection and ultimately provide a valuable resource towards our understanding of the host-pathogen dynamics and its variation with time.
Collapse
Affiliation(s)
- Luca Peruzza
- School of Ocean and Earth Science, University of Southampton, Hampshire, SO14 3ZH, United Kingdom.
| | - M S Shekhar
- Genetics and Biotechnology Unit, Central Institute of Brackishwater Aquaculture, 75 Santhome High Road, R.A. Puram, Chennai, 600004, Tamil Nadu, India
| | - K Vinaya Kumar
- Genetics and Biotechnology Unit, Central Institute of Brackishwater Aquaculture, 75 Santhome High Road, R.A. Puram, Chennai, 600004, Tamil Nadu, India
| | - A Swathi
- Genetics and Biotechnology Unit, Central Institute of Brackishwater Aquaculture, 75 Santhome High Road, R.A. Puram, Chennai, 600004, Tamil Nadu, India
| | - K Karthic
- Genetics and Biotechnology Unit, Central Institute of Brackishwater Aquaculture, 75 Santhome High Road, R.A. Puram, Chennai, 600004, Tamil Nadu, India
| | - Chris Hauton
- School of Ocean and Earth Science, University of Southampton, Hampshire, SO14 3ZH, United Kingdom
| | - K K Vijayan
- Genetics and Biotechnology Unit, Central Institute of Brackishwater Aquaculture, 75 Santhome High Road, R.A. Puram, Chennai, 600004, Tamil Nadu, India
| |
Collapse
|
29
|
Godoy-Lugo JA, Miranda-Cruz MM, Rosas-Rodríguez JA, Adan-Bante NP, Icedo-García R, Soñanez-Organis JG. Hypoxia inducible factor -1 regulates WSSV-induced glycolytic genes in the white shrimp Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2019; 92:165-171. [PMID: 31146006 DOI: 10.1016/j.fsi.2019.05.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/04/2019] [Accepted: 05/20/2019] [Indexed: 06/09/2023]
Abstract
Hypoxia-inducible factor -1 (HIF-1) is a transcriptional factor that regulates the expression of several glycolytic genes. The white spot syndrome virus (WSSV) induces a shift in glycolysis that favors viral replication in white shrimp Litopenaeus vannamei. HIF-1 is related to the pathogenesis of the WSSV infection through the induction of metabolic changes in infected white shrimp. Although the WSSV infection is associated with metabolic changes, the role of HIF-1 on key glycolytic genes during the WSSV infection has not been examined. In this work, we evaluated the effect of HIF-1α silencing on expression and activity of glycolytic enzymes (Hexokinase-HK, phosphofructokinase-PFK and pyruvate kinase-PK) along with the glucose transporter 1 (Glut1), regulatory enzymes (glucose-6-phosphate dehydrogenase-G6PDH and pyruvate dehydrogenase-PDH), and metabolic intermediates of glycolysis (glucose-6-phosphate-G6P and pyruvate). The expression of Glut1 increased in each tissue evaluated after WSSV infection, while HK, PFK and PK gene expression and enzyme activities increased in a tissue-specific manner. G6PDH activity increased during WSSV infection, and its substrate G6P decreased, while PDH activity decreased and its substrate pyruvate increased. Silencing of HIF-1α blocked the WSSV-induced Glut1 and glycolytic genes upregulation and enzyme activity in a tissue-specific manner. We conclude that HIF-1 regulates the WSSV-induced glycolysis through induction of glycolytic genes contributing to glucose metabolism in tissues of infected shrimp. Also, the inhibition, and activation of regulatory genes are likely to decrease the availability of the raw materials essential for WSSV replication and increase oxidative metabolism.
Collapse
Affiliation(s)
| | - Melissa M Miranda-Cruz
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, Mexico
| | - Jesús Alfredo Rosas-Rodríguez
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, Mexico
| | - Norma Patricia Adan-Bante
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, Mexico
| | - Ramona Icedo-García
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, Mexico
| | - José Guadalupe Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, Mexico.
| |
Collapse
|
30
|
Zhang YS, Li FX, Yao CL. Glycogen phosphorylase of shrimp (Litopenaeus vannamei): Structure, expression and anti-WSSV function. FISH & SHELLFISH IMMUNOLOGY 2019; 91:275-283. [PMID: 31125663 DOI: 10.1016/j.fsi.2019.05.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 06/09/2023]
Abstract
Glycogen phosphorylase (GP, EC 2.4.1.1) catalyze the rate-limiting step in glycogenolysis in animals, forming glucose-1-phosphate from the terminal alpha-1,4-glycosidic bond. Therefore, GP plays a crucial role in carbohydrate metabolism. In the present study, the full-length cDNA sequence of GP (LvGP) was cloned from shrimp, Litopenaeus vannamei. The obtained 3242-bp LvGP cDNA sequence included a 5'-terminal untranslated region (UTR) of 48 bp, an open reading frame (ORF) of 2559 bp encoding a polypeptide of 852 amino acids (aa) and a 3'-UTR of 635 bp. The predicted LvGP protein sequence contained a typical phosphorylase domain (113-829 aa) and shared 72%-97% identities with GP from other species. Phylogenetic analysis revealed that LvGP showed the closest relationship with GP from Marsupenaeus japonicus. Tissue expression profiles showed that LvGP existed in most examined tissues, with the most predominant expression in the brain, followed by the muscles and stomach. LvGP transcripts in hepatopancreas and hemocytes were up regulated after the WSSV challenge. Furthermore, the role of LvGP in shrimp defending against WSSV infection was investigated by RNA interference (RNAi). Our findings showed that WSSV proliferation and shrimp accumulative mortality increased significantly after LvGP RNAi (P < 0.01). The glycogen, glucose, and pyruvate content decreased in GP RNAi shrimp after WSSV injection, however, the lactate and ATP concentration enhanced. Moreover, lectin and anti-lipopolysaccharide factor2 (ALF2) were induced in LvGP silencing shrimp after WSSV infection, whereas the expression levels of crustin, ALF1 and ALF3 decreased. Our results suggested that the LvGP might play a crucial role in shrimp defending against WSSV infection by regulating metabolism and affecting the anti-infectious gene expression.
Collapse
Affiliation(s)
| | - Fei-Xiang Li
- Fisheries College, Jimei University, Xiamen, 361021, China
| | - Cui-Luan Yao
- Fisheries College, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
31
|
Abstract
Viruses depend on the host cells they infect to provide the machinery and substrates for replication. Host cells are highly dynamic systems that can alter their intracellular environment and metabolic behavior, which may be helpful or inhibitory for an infecting virus. In this study, we show that macrophages, a target cell of murine norovirus (MNV), increase glycolysis upon viral infection, which is important for early steps in MNV infection. Human noroviruses (hNoV) are a major cause of gastroenteritis globally, causing enormous morbidity and economic burden. Currently, no effective antivirals or vaccines exist for hNoV, mainly due to the lack of high-efficiency in vitro culture models for their study. Thus, insights gained from the MNV model may reveal aspects of host cell metabolism that can be targeted for improving hNoV cell culture systems and for developing effective antiviral therapies. The metabolic pathways of central carbon metabolism, glycolysis and oxidative phosphorylation (OXPHOS), are important host factors that determine the outcome of viral infections and can be manipulated by some viruses to favor infection. However, mechanisms of metabolic modulation and their effects on viral replication vary widely. Herein, we present the first metabolomics and energetic profiling of norovirus-infected cells, which revealed increases in glycolysis, OXPHOS, and the pentose phosphate pathway (PPP) during murine norovirus (MNV) infection. Inhibiting glycolysis with 2-deoxyglucose (2DG) in macrophages revealed that glycolysis is an important factor for optimal MNV infection, while inhibiting the PPP and OXPHOS showed a relatively minor impact of these pathways on MNV infection. 2DG affected an early stage in the viral life cycle after viral uptake and capsid uncoating, leading to decreased viral protein production and viral RNA. The requirement of glycolysis was specific for MNV (but not astrovirus) infection, independent of the type I interferon antiviral response, and unlikely to be due to a lack of host cell nucleotide synthesis. MNV infection increased activation of the protein kinase Akt, but not AMP-activated protein kinase (AMPK), two master regulators of cellular metabolism, implicating Akt signaling in upregulating host metabolism during norovirus infection. In conclusion, our findings suggest that the metabolic state of target cells is an intrinsic host factor that determines the extent of norovirus replication and implicates glycolysis as a virulence determinant. They further point to cellular metabolism as a novel therapeutic target for norovirus infections and improvements in current human norovirus culture systems.
Collapse
|
32
|
Li C, Weng S, He J. WSSV-host interaction: Host response and immune evasion. FISH & SHELLFISH IMMUNOLOGY 2019; 84:558-571. [PMID: 30352263 DOI: 10.1016/j.fsi.2018.10.043] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/15/2018] [Accepted: 10/20/2018] [Indexed: 06/08/2023]
Abstract
As invertebrates, shrimps rely on multiple innate defense reactions, including humoral immunity and cellular immunity to recognize and eliminate various invaders, such as viruses. White spot syndrome virus (WSSV) causes the most prevalent and devastating viral disease in penaeid shrimps, which are the most widely cultured species in the coastal waters worldwide. In the last couple of decades, studies about WSSV implicate a dual role of the immune system in protecting shrimps against the infection; these studies also explore on the pathogenesis of WSSV infection. Herein, we review our current knowledge of the innate immune responses of shrimps to WSSV, as well as the molecular mechanisms used by this virus to evade host immune responses or actively subvert them for its own benefit. Deciphering the interactions between WSSV and the shrimp host is paramount to understanding the mechanisms that regulate the balance between immune-mediated protection and pathogenesis during viral infection and to the development of a safe and effective WSSV defensive strategy.
Collapse
Affiliation(s)
- Chaozheng Li
- State Key Laboratory for Biocontrol / School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- State Key Laboratory for Biocontrol / School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- State Key Laboratory for Biocontrol / School of Marine Sciences, Sun Yat-sen University, Guangzhou, PR China; School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
33
|
Miranda-Cruz MM, Poom-Llamas JJ, Godoy-Lugo JA, Ortiz RM, Gómez-Jiménez S, Rosas-Rodríguez JA, Morán-Palacio EF, Soñanez-Organis JG. Silencing of HIF-1 in WSSV-infected white shrimp: Effect on viral load and antioxidant enzymes. Comp Biochem Physiol C Toxicol Pharmacol 2018; 213:19-26. [PMID: 30041062 DOI: 10.1016/j.cbpc.2018.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 01/23/2023]
Abstract
Hypoxia inducible factor-1 (HIF-1) is a transcriptional factor that induces genes involved in glucose metabolism. HIF-1 is formed by a regulatory α-subunit (HIF-1α) and a constitutive β-subunit (HIF-1β). The white spot syndrome virus (WSSV) induces a shift in glucose metabolism and oxidative stress. HIF-1α is associated with the induction of metabolic changes in tissues of WSSV-infected shrimp. However, the contributions of HIF-1 to viral load and antioxidant responses in WSSV-infected shrimp have been not examined. In this study, the effect of HIF-1 silencing on viral load and the expression and activity of antioxidant enzymes (superoxide dismutase-SOD, glutathione S-transferase-GST, and catalase) along with oxidative damage (lipid peroxidation and protein carbonyl) in tissues of white shrimp infected with the WSSV were studied. The viral load increased in hepatopancreas and muscle after WSSV infection, and the accumulative mortality was of 100% at 72 h post-infection. The expression and activity of SOD, catalase, and GST decreased in each tissue evaluated after WSSV infection. Protein carbonyl concentrations increased in each tissue after WSSV infection, while lipid peroxidation increased in hepatopancreas, but not in muscle. Silencing of HIF-1α decreased the WSSV viral load in hepatopancreas and muscle of infected shrimp along with shrimp mortality. Silencing of HIF-1α ameliorated the antioxidant response in a tissue-specific manner, which translated to a decrease in oxidative damage. These results suggest that HIF-1 is essential for restoring the antioxidant response, which counters the oxidative injury associated with WSSV infection.
Collapse
Affiliation(s)
- Melissa M Miranda-Cruz
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, México
| | - Jennifer J Poom-Llamas
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, México
| | - José A Godoy-Lugo
- School of Natural Sciences, University of California Merced, 5200 N Lake Road, Merced, CA 95343, USA
| | - Rudy M Ortiz
- School of Natural Sciences, University of California Merced, 5200 N Lake Road, Merced, CA 95343, USA
| | - Silvia Gómez-Jiménez
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6. Hermosillo, Sonora, C.P. 83304, México
| | - Jesús A Rosas-Rodríguez
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, México
| | - Edgar F Morán-Palacio
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, México
| | - José G Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico-Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Apartado Postal 85880, Navojoa, Sonora, México.
| |
Collapse
|
34
|
Yu L, Chen X, Wang L, Chen S. Oncogenic virus-induced aerobic glycolysis and tumorigenesis. J Cancer 2018; 9:3699-3706. [PMID: 30405839 PMCID: PMC6216013 DOI: 10.7150/jca.27279] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
Enhanced glycolysis under normoxic conditions is known as aerobic glycolysis or the Warburg effect and is a hallmark of many tumors. Viral infection may also induce aerobic glycolysis as it is required for replication and survival. Tumor viruses inducing aerobic glycolysis and lactate production during latent infection suggest a potential role of virus-induced glycolysis in tumorigenesis. Virus or virus-encoded proteins regulate glucose uptake and lactate export, increase the activity of glycolytic enzymes, and modulate glucose metabolic signals. Accumulating evidence suggests that virus-induced glycolysis may facilitate cell growth, transformation, migration, and invasion, but its significance in tumorigenesis remains unclear. We summarize the effects of oncogenic viruses on the metabolic shift to aerobic glycolysis and discuss the possible association of this metabolic reprogramming with tumor development and progression.
Collapse
Affiliation(s)
- Li Yu
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Xun Chen
- Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, People's Republic of China
| | - Liantang Wang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Shangwu Chen
- Guangdong Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People's Republic of China
| |
Collapse
|
35
|
Zhao C, Fu H, Sun S, Qiao H, Zhang W, Jin S, Jiang S, Xiong Y, Gong Y. A transcriptome study on Macrobrachium nipponense hepatopancreas experimentally challenged with white spot syndrome virus (WSSV). PLoS One 2018; 13:e0200222. [PMID: 29979781 PMCID: PMC6034857 DOI: 10.1371/journal.pone.0200222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/21/2018] [Indexed: 12/13/2022] Open
Abstract
White spot syndrome virus (WSSV) is one of the most devastating pathogens of cultured shrimp, responsible for massive loss of its commercial products worldwide. The oriental river prawn Macrobrachium nipponense is an economically important species that is widely farmed in China and adult prawns can be infected by WSSV. However, the molecular mechanisms of the host pathogen interaction remain unknown. There is an urgent need to learn the host pathogen interaction between M. nipponense and WSSV which will be able to offer a solution in controlling the spread of WSSV. Next Generation Sequencing (NGS) was used in this study to determin the transcriptome differences by the comparison of control and WSSV-challenged moribund samples, control and WSSV-challenged survived samples of hepatopancreas in M. nipponense. A total of 64,049 predicted unigenes were obtained and classified into 63 functional groups. Approximately, 4,311 differential expression genes were identified with 3,308 genes were up-regulated when comparing the survived samples with the control. In the comparison of moribund samples with control, 1,960 differential expression genes were identified with 764 genes were up-regulated. In the contrast of two comparison libraries, 300 mutual DEGs with 95 up-regulated genes and 205 down-regulated genes. All the DEGs were performed GO and KEGG analysis, overall a total of 85 immune-related genes were obtained and these gene were groups into 13 functions and 4 KEGG pathways, such as protease inhibitors, heat shock proteins, oxidative stress, pathogen recognition immune receptors, PI3K/AKT/mTOR pathway, MAPK signaling pathway and Ubiquitin Proteasome Pathway. Ten genes that valuable in immune responses against WSSV were selected from those DEGs to furture discuss the response of host to WSSV. Results from this study contribute to a better understanding of the immune response of M. nipponense to WSSV, provide information for identifying novel genes in the absence of genome of M. nipponense. Furthermore, large number of transcripts obtained from this study could provide a strong basis for future genomic research on M. nipponense.
Collapse
Affiliation(s)
- Caiyuan Zhao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, PR China
| | - Hongtuo Fu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, PR China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
- * E-mail:
| | - Shengming Sun
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Wenyi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Shubo Jin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Yiwei Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| | - Yongsheng Gong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, PR China
| |
Collapse
|
36
|
Hernández-Palomares MLE, Godoy-Lugo JA, Gómez-Jiménez S, Gámez-Alejo LA, Ortiz RM, Muñoz-Valle JF, Peregrino-Uriarte AB, Yepiz-Plascencia G, Rosas-Rodríguez JA, Soñanez-Organis JG. Regulation of lactate dehydrogenase in response to WSSV infection in the shrimp Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2018; 74:401-409. [PMID: 29337249 DOI: 10.1016/j.fsi.2018.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/17/2017] [Accepted: 01/11/2018] [Indexed: 06/07/2023]
Abstract
Lactate dehydrogenase (LDH) is key for anaerobic glycolysis. LDH is induced by the hypoxia inducible factor -1 (HIF-1). HIF-1 induces genes involved in glucose metabolism and regulates cellular oxygen homeostasis. HIF-1 is formed by a regulatory α-subunit (HIF-1α) and a constitutive β-subunit (HIF-1β). The white spot syndrome virus (WSSV) induces anaerobic glycolysis in shrimp hemocytes, associated with lactate accumulation. Although infection and lactate production are associated, the LDH role in WSSV-infected shrimp has not been examined. In this work, the effects of HIF-1 silencing on the expression of two LDH subunits (LDHvan-1 and LDHvan-2) in shrimp infected with the WSSV were studied. HIF-1α transcripts increased in gills, hepatopancreas, and muscle after WSSV infection, while HIF-1β remained constitutively expressed. The expression for both LDH subunits increased in each tissue evaluated during the WSSV infection, translating into increased enzyme activity. Glucose concentration increased in each tissue evaluated, while lactate increased in gills and hepatopancreas, but not in muscle. Silencing of HIF-1α blocked the increase of LDH expression and enzyme activity, along with glucose (all tissues) and lactate (gills and hepatopancreas) concentrations produced by WSSV infection. These results demonstrate that HIF-1 up regulates the expression of LDH subunits during WSSV infection, and that this induction contributes to substrate metabolism in energetically active tissues of infected shrimp.
Collapse
Affiliation(s)
- M L E Hernández-Palomares
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6, Hermosillo, Sonora, C.P. 83304, Mexico
| | - J A Godoy-Lugo
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Lázaro Cárdenas #100, Col. Francisco Villa, Apartado Postal 85390, Navojoa, Sonora, Mexico
| | - S Gómez-Jiménez
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6, Hermosillo, Sonora, C.P. 83304, Mexico
| | - L A Gámez-Alejo
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6, Hermosillo, Sonora, C.P. 83304, Mexico
| | - R M Ortiz
- School of Natural Sciences, University of California Merced, 5200 N Lake Road, Merced, CA, 95343, USA
| | - J F Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - A B Peregrino-Uriarte
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6, Hermosillo, Sonora, C.P. 83304, Mexico
| | - G Yepiz-Plascencia
- Centro de Investigación en Alimentación y Desarrollo (CIAD), Carretera a la Victoria KM. 0.6, Hermosillo, Sonora, C.P. 83304, Mexico
| | - J A Rosas-Rodríguez
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Lázaro Cárdenas #100, Col. Francisco Villa, Apartado Postal 85390, Navojoa, Sonora, Mexico
| | - J G Soñanez-Organis
- Universidad de Sonora, Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Lázaro Cárdenas #100, Col. Francisco Villa, Apartado Postal 85390, Navojoa, Sonora, Mexico.
| |
Collapse
|
37
|
He HH, Chi YM, Yuan K, Li XY, Weng SP, He JG, Chen YH. Functional characterization of a reactive oxygen species modulator 1 gene in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2017; 70:270-279. [PMID: 28889015 DOI: 10.1016/j.fsi.2017.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/30/2017] [Accepted: 09/06/2017] [Indexed: 06/07/2023]
Abstract
Reactive oxygen species (ROS) imparts a dual effect on multicellular organisms, wherein high levels are usually harmful, and low levels could facilitate in combating pathogenic microorganisms; therefore, the regulation of ROS production is critical. Previous studies have suggested that ROS contributes to resistance to the white spot syndrome virus (WSSV) or Vibrio alginolyticus in Litopenaeus vannamei. However, the regulation of ROS metabolism in L. vannamei remains elusive. In the present study, we proved that the overexpression of L. vannamei reactive oxygen species modulator 1 (LvROMO1) increases ROS production in Drosophila Schneider 2 (S2) cells. Real-time RT-PCR analysis indicated that LvROMO1 is induced by WSSV or V. alginolyticus infection and β-glucan or microcystin (MC-LR) injection. Further investigation showed that LvROMO1 responding to MC-LR, thereby inducing hemocytes to undergo apoptosis, and ultimately resulting in hepatopancreatic damage. And LvROMO1 downregulation induced an increase in the cumulative mortality of WSSV-infected shrimp by reducing ROS production and suppressing the expression of antimicrobial peptides genes. The findings of present study suggest that LvROMO1 plays an important role in ROS production in L. vannamei and is involved in innate immunity.
Collapse
Affiliation(s)
- Hong-Hui He
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Yi-Miao Chi
- Key Laboratory of Marine Resources and Coastal Engineering in Guangdong Province, South China Sea Bio-Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), School of Marine Sciences, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Kai Yuan
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Xiao-Yun Li
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Shao-Ping Weng
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Jian-Guo He
- Key Laboratory of Marine Resources and Coastal Engineering in Guangdong Province, South China Sea Bio-Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), School of Marine Sciences, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China; School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China
| | - Yi-Hong Chen
- Key Laboratory of Marine Resources and Coastal Engineering in Guangdong Province, South China Sea Bio-Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), School of Marine Sciences, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China; State Key Laboratory for Biocontro, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, SunYat-senUniversity, 135 Xingang Road West, Guangzhou 510275, PR China.
| |
Collapse
|
38
|
Zielinski DC, Jamshidi N, Corbett AJ, Bordbar A, Thomas A, Palsson BO. Systems biology analysis of drivers underlying hallmarks of cancer cell metabolism. Sci Rep 2017; 7:41241. [PMID: 28120890 PMCID: PMC5264163 DOI: 10.1038/srep41241] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 12/19/2016] [Indexed: 12/18/2022] Open
Abstract
Malignant transformation is often accompanied by significant metabolic changes. To identify drivers underlying these changes, we calculated metabolic flux states for the NCI60 cell line collection and correlated the variance between metabolic states of these lines with their other properties. The analysis revealed a remarkably consistent structure underlying high flux metabolism. The three primary uptake pathways, glucose, glutamine and serine, are each characterized by three features: (1) metabolite uptake sufficient for the stoichiometric requirement to sustain observed growth, (2) overflow metabolism, which scales with excess nutrient uptake over the basal growth requirement, and (3) redox production, which also scales with nutrient uptake but greatly exceeds the requirement for growth. We discovered that resistance to chemotherapeutic drugs in these lines broadly correlates with the amount of glucose uptake. These results support an interpretation of the Warburg effect and glutamine addiction as features of a growth state that provides resistance to metabolic stress through excess redox and energy production. Furthermore, overflow metabolism observed may indicate that mitochondrial catabolic capacity is a key constraint setting an upper limit on the rate of cofactor production possible. These results provide a greater context within which the metabolic alterations in cancer can be understood.
Collapse
Affiliation(s)
- Daniel C Zielinski
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92093-0412, USA
| | - Neema Jamshidi
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92093-0412, USA.,Institute of Engineering in Medicine, University of California, San Diego, La Jolla CA 92093-0412, USA
| | - Austin J Corbett
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92093-0412, USA
| | - Aarash Bordbar
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92093-0412, USA
| | - Alex Thomas
- Department of Bioinformatics and Systems Biology, University of California, San Diego, La Jolla CA 92093-0412, USA.,The Novo Nordisk Center for Biosustainability at the University of California San Diego School of Medicine, University of California, San Diego, La Jolla CA 92093-0412, USA.,Department of Pediatrics, University of California, San Diego, La Jolla CA 92093-0412, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla CA 92093-0412, USA.,Department of Pediatrics, University of California, San Diego, La Jolla CA 92093-0412, USA
| |
Collapse
|