1
|
Schmalzriedt DL, Aurubin CA, Rahlf CR, Brown MA, Bobek JM, Lange PT, Bradeen XG, Sahoo D, Tarakanova VL. Apolipoprotein E selectively supports gammaherpesvirus replication in macrophages. J Virol 2025; 99:e0048025. [PMID: 40439406 DOI: 10.1128/jvi.00480-25] [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: 03/17/2025] [Accepted: 04/17/2025] [Indexed: 06/18/2025] Open
Abstract
Gammaherpesviruses establish lifelong infections in over 90% of adults worldwide and contribute to the development of several cancers. Endogenous lipid synthesis pathways support lytic and latent life cycles of several gammaherpesviruses. However, the role of circulating lipoproteins and the corresponding apolipoproteins in gammaherpesvirus infection remains unknown. Apolipoprotein E (ApoE) is a protein that associates with lipoproteins in circulation and supports bidirectional lipid transport to maintain lipid homeostasis. Interestingly, ApoE differentially affects several virus families, but its role in gammaherpesvirus infection has not been evaluated. In this study, we demonstrate that ApoE expression was increased in murine gammaherpesvirus 68 (MHV68)-infected macrophages in a type-I interferon (IFN)-dependent manner. Intriguingly, ApoE expression was usurped to support MHV68 lytic replication and expression of lytic viral genes. The proviral effects of ApoE in macrophages were independent of the conventional functions of ApoE in the regulation of endogenous lipid synthesis and type I IFN signaling. Finally, the proviral effects of ApoE were limited to the lytic life cycle, as the establishment of MHV68 latency in macrophages was not altered by the ApoE genotype of chronically infected mice. Thus, our study defines a viral life cycle-specific proviral role of ApoE in gammaherpesvirus infection. IMPORTANCE ApoE is an apolipoprotein that mediates lipid transport and exchange between tissues and the circulation. ApoE differentially affects several virus families, but its role in gammaherpesvirus infection remains unknown. Here, we show that ApoE supported lytic gammaherpesvirus replication in primary macrophages and that infected macrophages increased expression of ApoE in an interferon-dependent manner. However, ApoE expression did not affect viral latency in vivo, implying a novel viral life cycle-specific proviral role for ApoE in gammaherpesvirus infection.
Collapse
Affiliation(s)
- Damon L Schmalzriedt
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Carlie A Aurubin
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cade R Rahlf
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Matthew A Brown
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jordan M Bobek
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Philip T Lange
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Xander G Bradeen
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Daisy Sahoo
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vera L Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
2
|
Lei Z, Wei W, Wang M, Xu Y, Bai L, Gao Y, Jiang C, Li F, Tian N, Kuang L, Zhu R, Pang G, Lan K, Feng S, Liang X. PINLYP-mediated phospholipid metabolism reprogramming contributes to chronic herpesvirus infection. PLoS Pathog 2025; 21:e1013146. [PMID: 40373067 PMCID: PMC12080810 DOI: 10.1371/journal.ppat.1013146] [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: 01/07/2025] [Accepted: 04/18/2025] [Indexed: 05/17/2025] Open
Abstract
Many viruses alter the phospholipid metabolism to benefit their own life cycles. It is unclear whether the host or the virus is driving phospholipid metabolism reprogramming, and how virus infections are affected by the metabolic status. Here we report that phospholipase A2 inhibitor and LY6/PLAUR domain-containing protein (PINLYP) inhibits Kaposi's sarcoma-associated herpesvirus (KSHV) lytic reactivation by remodeling phospholipid metabolism and especially triacylglycerol (TAG) biosynthesis. PINLYP deficiency led to increased phospholipase cPLA2α activity, cPLA2α-mediated AKT phosphorylation, and KSHV lytic reactivation. Analyses of RNA-seq and lipidomics reveal that PINLYP regulates long-chain fatty acid CoA ligase ACSL5 expression and TAG production. The inhibition of ACSL5 activity or TAG biosynthesis suppresses AKT phosphorylation and KSHV lytic reactivation, restoring the phenotype of PINLYP deficiency. This finding underscores the pivotal role of PINLYP in remodeling phospholipid metabolism and promoting viral latency, which sheds new light on how phospholipid metabolism is regulated by herpesvirus and provides a potential target for controlling chronic herpesvirus infection.
Collapse
Affiliation(s)
- Zhangmengxue Lei
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Wendi Wei
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Mingyu Wang
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Yun Xu
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Lei Bai
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Gao
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Congwei Jiang
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fangxia Li
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Na Tian
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Linlin Kuang
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Ruiliang Zhu
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Gang Pang
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Ke Lan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Suihan Feng
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Xiaozhen Liang
- University of Chinese Academy of Sciences, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
3
|
Albano C, Trifirò L, Hewelt-Belka W, Cairns DM, Pasquero S, Griffante G, Gugliesi F, Bajetto G, Garwolińska D, Rossi M, Vallino M, Malerba M, De Andrea M, Kaplan DL, Dell'Oste V, Biolatti M. The impact of fatty acid synthase on HSV-1 infection dynamics. PLoS Pathog 2025; 21:e1013068. [PMID: 40327680 DOI: 10.1371/journal.ppat.1013068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 05/16/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Herpes simplex virus type-1 (HSV-1) is a widespread human pathogen that relies on host cell pathways, including those involved in metabolism to support replication. Here, we demonstrate that de novo lipogenesis is essential for HSV-1 infectivity. Specifically, HSV-1 infection upregulates fatty acid synthase (FASN) expression, accompanied by a marked increase in lipids and a differential lipid species distribution. Conversely, silencing FASN or applying FASN inhibitors (i.e., CMS121 and C75) markedly reduces the infectivity of newly released HSV-1 virions, suggesting that, while initial replication remains unaffected, FASN is crucial for maintaining virion structure and facilitating entry into host cells. Additionally, we show that a source of lipid-rich external factors provided by fetal bovine serum significantly increases HSV-1 infectivity. Specifically, HSV-1 infection enhanced CD36-mediated fatty acid uptake, especially in FASN-depleted cells, compensating for reduced lipogenesis. Blocking CD36 function with SSO further decreased viral infectivity, demonstrating the critical role of lipid uptake in HSV-1 life cycle. Altogether, our findings reveal how HSV-1 manipulates lipid metabolism, offering insights into its association with chronic disease and therapeutic intervention.
Collapse
Affiliation(s)
- Camilla Albano
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Linda Trifirò
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | | | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Selina Pasquero
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Gloria Griffante
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Francesca Gugliesi
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Greta Bajetto
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
- CAAD Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Dorota Garwolińska
- Department of Analytical Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Marika Rossi
- Institute for Sustainable Plant Protection, National Research Council of Italy, Turin, Italy
| | - Marta Vallino
- Institute for Sustainable Plant Protection, National Research Council of Italy, Turin, Italy
| | - Mario Malerba
- INRIM Istituto Nazionale di Ricerca Metrologica, Turin, Italy
| | - Marco De Andrea
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
- CAAD Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Valentina Dell'Oste
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Matteo Biolatti
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Feng S, Fu W, Li S, Yeh W, She R, Brenner C, Chen C, Feng P. Oral Neisseria gonorrhoeae Promotes KSHV Lytic Replication. J Med Virol 2025; 97:e70304. [PMID: 40171956 DOI: 10.1002/jmv.70304] [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: 01/02/2025] [Revised: 02/13/2025] [Accepted: 03/08/2025] [Indexed: 04/04/2025]
Abstract
The human oral cavity contains highly diverse microbes, including bacteria, fungi, and viruses. Human herpesviruses are ubiquitous pathogens, and the oral cavity is conducive to the replication, dissemination, and pathogenesis of human herpesviruses. Herpesviruses are generally pathogenic in immunodeficient individuals, such as AIDS patients and organ transplant recipients. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma and two types of rare lymphoma, that is, primary effusion lymphoma and multicentric Castleman's disease. Mounting evidence indicates that KSHV viral load positively correlates with ongoing bacterial infection in the oral cavity, suggesting that bacteria potentially stimulate KSHV replication. However, the mechanism by which oral bacteria may promote KSHV lytic replication is poorly understood. In this study, we performed DNA sequencing and 16S ribosomal RNA analysis of saliva samples of AIDS-KS patients. A correlation analysis identified a panel of oral residential bacteria and uncommon ones that paralleled with KSHV viral load. Performing functional assays, we discovered that the sexually transmitted Neisseria gonorrhoeae (N.g.) significantly increased KSHV lytic replication. Increased KSHV lytic replication was evidenced by elevated levels of mRNA and proteins of viral lytic genes. N.g. stimulation increases the expression of RTA that drives viral lytic replication. Metabolomic analysis reveals the synergistic effect of KSHV and N.g. on cellular metabolism, including the glycolysis and purine and pyrimidine synthesis, that likely underpins the elevated KSHV lytic replication. Findings from our study shed light on the molecular detail of bacteria-virus interaction in the oral cavity and provide references to develop an innovative strategy to treat diseases associated with KSHV.
Collapse
Affiliation(s)
- Shu Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Wen Fu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Shutong Li
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Wayne Yeh
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Rosemary She
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
- Department of Pathology, Duarte, California, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Casey Chen
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
5
|
Alfaez A, Christopher MW, Garrett TJ, Papp B. Analysis of Metabolomic Reprogramming Induced by Infection with Kaposi's Sarcoma-Associated Herpesvirus Using Untargeted Metabolomic Profiling. Int J Mol Sci 2025; 26:3109. [PMID: 40243754 PMCID: PMC11988554 DOI: 10.3390/ijms26073109] [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: 01/28/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus. There are no vaccines or antiviral therapies for KSHV. Identifying the cellular metabolic pathways that KSHV manipulates can broaden the knowledge of how these pathways contribute to sustaining lytic infection, which can be targeted in future therapies to prevent viral spread. In this study, we performed an untargeted metabolomic analysis of KSHV infected telomerase-immortalized gingival keratinocytes (TIGK) cells at 4 h post-infection compared to mock-infected cells. We found that the metabolomic landscape of KSHV-infected TIGK differed from that of the mock. Specifically, a total of 804 differential metabolic features were detected in the two groups, with 741 metabolites that were significantly upregulated, and 63 that were significantly downregulated in KSHV-infected TIGK cells. The differential metabolites included ornithine, arginine, putrescine, dimethylarginine, orotate, glutamate, and glutamine, and were associated with pathways, such as the urea cycle, polyamine synthesis, dimethylarginine synthesis, and de novo pyrimidine synthesis. Overall, our untargeted metabolomics analysis revealed that KSHV infection results in marked rapid alterations in the metabolic profile of the oral epithelial cells. We envision that a subset of these rapid metabolic changes might result in altered cellular functions that can promote viral lytic replication and transmission in the oral cavity.
Collapse
Affiliation(s)
- Abdulkarim Alfaez
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, USA;
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | | | - Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610, USA;
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA;
| | - Bernadett Papp
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610, USA
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
- Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Informatics Institute, University of Florida, Gainesville, FL 32610, USA
- Center for Orphaned Autoimmune Disorders, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
6
|
Ma YX, Han YQ, Wang PZ, Wang MY, Yang GY, Li JL, Wang J, Chu BB. Porcine reproductive and respiratory syndrome virus activates lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs axis. Int J Biol Macromol 2024; 282:136720. [PMID: 39433189 DOI: 10.1016/j.ijbiomac.2024.136720] [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: 07/27/2024] [Revised: 09/27/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is a highly contagious pathogen in pigs. This study aimed to investigate the impact of PRRSV infection on cellular metabolism, particularly focusing on lipid metabolism to understand its role in promoting viral replication. We conducted a metabolic analysis on MARC-145 cells before and after PRRSV infection. Our results demonstrated that the most significant alterations in cellular metabolism, accounting for 40.8 % of total changes, were related to lipid metabolism. These changes were primarily driven by the activation of sterol regulatory-element binding proteins (SREBPs), critical regulators of lipid biosynthesis. To understand the mechanisms behind SREBPs activation by PRRSV, we investigated the involvement of upstream effectors, specifically protein kinase B (AKT) and phosphoenolpyruvate carboxykinase 1 (PCK1). Our findings indicated that PRRSV infection triggered AKT activation, leading to the subsequent activation of PCK1. Activated PCK1 then phosphorylated insulin-induced genes (INSIGs), resulting in their degradation. This degradation facilitated the translocation of SREBPs from the endoplasmic reticulum to the nucleus. Additionally, we observed that PRRSV infection stimulated the production of reactive oxygen species (ROS), which played a critical role in activating AKT. Collectively, our findings demonstrate that PRRSV enhances lipid synthesis through a ROS-dependent AKT/PCK1/INSIG/SREBPs signaling axis, which provides new insights into the metabolic strategies employed by PRRSV.
Collapse
Affiliation(s)
- Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ya-Qi Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Pei-Zhu Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Ming-Yang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China
| | - Jian-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China.
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou 450046, Henan Province, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Longhu Advanced Immunization Laboratory, Zhengzhou 450046, Henan Province, China; International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, Henan Province, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
7
|
Singh RK, Torne AS, Robertson ES. Hypoxic reactivation of Kaposi's sarcoma associated herpesvirus. CELL INSIGHT 2024; 3:100200. [PMID: 39391006 PMCID: PMC11466537 DOI: 10.1016/j.cellin.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024]
Abstract
Hypoxic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) refers to the phenomenon under low oxygen where the virus goes from latent to lytic replication. Typically, healthy cells generally cease cell division and DNA replication under hypoxic conditions due to limited resources, and the presence of physiological inhibitors. This restricted replication under hypoxic conditions is considered an employed strategy of the cell to minimize energy consumption. However, cancerous cells continuously replicate and divide in hypoxic conditions by reprogramming several aspects of their cell physiology, including but not limited to metabolism, cell cycle, DNA replication, transcription, translation, and the epigenome. KSHV infection, similar to cancerous cells, is known to bypass hypoxia-induced restrictions and undergo reactivation to produce progeny viruses. In previous studies we have mapped several aspects of cell physiology that are manipulated by KSHV through its latent antigens during hypoxic conditions, which allows for a permissive environment for its replication. We discuss the major strategies utilized by KSHV to bypass hypoxia-induced repression. We also describe the KSHV-encoded antigens responsible for modulating these cellular processes important for successful viral replication and persistence in hypoxia.
Collapse
Affiliation(s)
- Rajnish Kumar Singh
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Atharva S Torne
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| |
Collapse
|
8
|
Choi UY, Lee SH. Understanding Metabolic Pathway Rewiring by Oncogenic Gamma Herpesvirus. J Microbiol Biotechnol 2024; 34:2143-2152. [PMID: 39403716 PMCID: PMC11637867 DOI: 10.4014/jmb.2407.07039] [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: 07/22/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 11/29/2024]
Abstract
Gamma herpesviruses, including Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV), are key contributors to the development of various cancers through their ability to manipulate host cellular pathways. This review explores the intricate ways these viruses rewire host metabolic pathways to sustain viral persistence and promote tumorigenesis. We look into how EBV and KSHV induce glycolytic reprogramming, alter mitochondrial function, and remodel nucleotide and amino acid metabolism, highlighting the crucial role of lipid metabolism in these oncogenic processes. By understanding these metabolic alterations, which confer proliferative and survival advantages to the virus-infected cells, we can identify potential therapeutic targets and develop innovative treatment strategies for gamma herpesvirus-associated malignancies. Ultimately, this review underscores the critical role of metabolic reprogramming in gamma herpesvirus oncogenesis and its implications for precision medicine in combating virus-driven cancers.
Collapse
Affiliation(s)
- Un Yung Choi
- Department of Microbiology, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
- KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| | - Seung Hyun Lee
- Department of Microbiology, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
- KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, Republic of Korea
| |
Collapse
|
9
|
Feng H, Zhang K, Zhang J, Wang X, Guo Z, Wang L, Chen F, Han S, Li J. The alleviating effect of Phillygenin on the regulation of respiratory microbiota and its metabolites in IBV-infected broilers by inhibiting the TLR7/MyD88/NF-κB axis. FASEB J 2024; 38:e23882. [PMID: 39143727 DOI: 10.1096/fj.202400168rr] [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: 01/22/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
Phillygenin (PHI) is an active ingredient derived from the leaf of Forsythia suspensa that has been found to alleviate inflammation and peroxidation response. Avian infectious bronchitis (IB) is a major threat to poultry industry viral respiratory tract disease that infected with infectious bronchitis virus (IBV). This study investigated the protection of PHI to CEK cell and broiler's tracheal injury triggered by avian infectious bronchitis virus (IBV). The results showed that IBV infection did not cause serious clinical symptoms and slowing-body weight in PHI-treated broilers. The expression of virus loads, pro-inflammation factors (IL-6, TNF-α, and IL-1β) in CEK cell, and tracheas were decreased compared to the IBV group, exhibiting its potent anti-inflammation. Mechanistically, the study demonstrated that the inhibition of TLR7/MyD88/NF-κB pathway was mainly involved in the protection effect of PHI to inflammation injury. Interestingly, a higher abundance of Firmicutes and Lactobacillus in respiratory tract was observed in PHI-treated broilers than in the IBV group. Significant differences were observed between the IBV group and PHI-treated group in the Ferroptosis, Tryptophan metabolism, and Glutathione metabolism pathways. PHI exhibited potent protection effect on IBV infection and alleviated inflammation injury, mainly through inhibiting TLR7/MyD88/NF-κB pathway. The study encourages further development of PHI, paving the way to its clinical use as a new candidate drug to relieve IBV-induced respiratory symptoms.
Collapse
Affiliation(s)
- Haipeng Feng
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Kang Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jingyan Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xuezhi Wang
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhiting Guo
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Lei Wang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Fubing Chen
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Songwei Han
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jianxi Li
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
10
|
Zhang J, Wang P, Li T, Liu W, Shi T, Wang M, Kong W, Huang X, Aihemaijiang K, Ding Y, Gao F, Kang X. Association between human herpesvirus 8 and lipid profile in northwest China: A cross-sectional study. J Med Virol 2024; 96:e29794. [PMID: 39101375 DOI: 10.1002/jmv.29794] [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: 03/25/2024] [Revised: 06/08/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024]
Abstract
Human herpesvirus 8 (HHV-8) infection shows obvious regional and ethnic differences. Although studies have shown that these differences may be associated with lipid metabolism, to date, no large-scale studies have explored this. This study explored the seropositivity rate of HHV-8 among 2516 residents from 10 regions of northwest China and then the correlates of HHV-8 infection with lipid profile. The HHV-8 serological positivity rate was 15.6% among all residents. The HHV-8 seroprevalence ranged 11.2-27.6% among different ethnicities. Across different BMI levels, the positive rates of HHV-8 were 27.6%, 16.9%, and 13.6% for a BMI < 18.5, 18.5-24.9, and ≥25, respectively. HHV-8 seropositivity rate was lower for hypertensive people (12.6%) than for non-hypertensive people (16.7%). Univariate logistic regression analyses revealed that age, hypertension, systolic blood pressure, BMI, total cholesterol, and high-density lipoprotein cholesterol (HDL-C) significantly correlated with HHV-8 seropositivity (p < 0.05). Multivariate logistic regression analysis after adjusting for confounding factors showed that HDL-C (odds ratio [OR]: 0.132, 95% confidence interval [CI], 0.082-0.212; p < 0.001) and BMI (OR: 0.959, 95% CI 0.933-0.986; p = 0.003) were associated with HHV-8 seropositivity. Subgroup analyses concerning ethnicity, sex, or age demonstrated a consistent relationship with HDL-C. The results of HHV-8 seropositivity and BMI were inconsistent in the subgroups. However, Spearman's correlation analysis between HHV-8 serum antibody titer and HDL-C levels showed no linear relationship among HHV-8 seropositive individuals (ρ = -0.080, p = 0.058). HHV-8 serum antibody titers were also not significantly correlated with BMI (ρ = -0.015, p = 0.381). Low HDL-C levels may be an independent risk factor for HHV-8 infection, but there is no significant correlation between HDL-C levels and HHV-8 antibody titers.
Collapse
Affiliation(s)
- Jingzhan Zhang
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Dermatology and Venereology, Urumqi, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, China
| | - Peng Wang
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Dermatology and Venereology, Urumqi, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, China
| | - Tingting Li
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Dermatology and Venereology, Urumqi, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, China
| | - Weidong Liu
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Tian Shi
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Man Wang
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wenjie Kong
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaoling Huang
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Kuerbanjiang Aihemaijiang
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yuan Ding
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Dermatology and Venereology, Urumqi, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, China
| | - Feng Gao
- Xinjiang Clinical Research Center for Digestive Diseases, Urumqi, China
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaojing Kang
- Department of Dermatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Xinjiang Clinical Research Center for Dermatology and Venereology, Urumqi, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, China
| |
Collapse
|
11
|
Bhattacharjee P, Wang D, Anderson D, Buckler JN, de Geus E, Yan F, Polekhina G, Schittenhelm R, Creek DJ, Harris LD, Sadler AJ. The immune response to RNA suppresses nucleic acid synthesis by limiting ribose 5-phosphate. EMBO J 2024; 43:2636-2660. [PMID: 38778156 PMCID: PMC11217295 DOI: 10.1038/s44318-024-00100-w] [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: 09/06/2023] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 05/25/2024] Open
Abstract
During infection viruses hijack host cell metabolism to promote their replication. Here, analysis of metabolite alterations in macrophages exposed to poly I:C recognises that the antiviral effector Protein Kinase RNA-activated (PKR) suppresses glucose breakdown within the pentose phosphate pathway (PPP). This pathway runs parallel to central glycolysis and is critical to producing NADPH and pentose precursors for nucleotides. Changes in metabolite levels between wild-type and PKR-ablated macrophages show that PKR controls the generation of ribose 5-phosphate, in a manner distinct from its established function in gene expression but dependent on its kinase activity. PKR phosphorylates and inhibits the Ribose 5-Phosphate Isomerase A (RPIA), thereby preventing interconversion of ribulose- to ribose 5-phosphate. This activity preserves redox control but decreases production of ribose 5-phosphate for nucleotide biosynthesis. Accordingly, the PKR-mediated immune response to RNA suppresses nucleic acid production. In line, pharmacological targeting of the PPP during infection decreases the replication of the Herpes simplex virus. These results identify an immune response-mediated control of host cell metabolism and suggest targeting the RPIA as a potential innovative antiviral treatment.
Collapse
Affiliation(s)
- Pushpak Bhattacharjee
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Die Wang
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Dovile Anderson
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Joshua N Buckler
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, 5010, New Zealand
| | - Eveline de Geus
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Feng Yan
- Australian Centre for Blood Diseases, Department of Clinical Hematology, Monash University, Clayton, VIC, 3004, Australia
| | - Galina Polekhina
- Department of Epidemiology & Preventive Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Ralf Schittenhelm
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Lawrence D Harris
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, 5010, New Zealand
| | - Anthony J Sadler
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
12
|
Wang G, Cao Y, Xu C, Zhang S, Huang Y, Zhang S, Bao W. Comprehensive transcriptomic and metabolomic analysis of porcine intestinal epithelial cells after PDCoV infection. Front Vet Sci 2024; 11:1359547. [PMID: 38855411 PMCID: PMC11160942 DOI: 10.3389/fvets.2024.1359547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Introduction Porcine deltacoronavirus (PDCoV), an emerging swine enteropathogenic coronavirus with worldwide distribution, mainly infects newborn piglets with severe diarrhea, vomiting, dehydration, and even death, causing huge economic losses to the pig industry. However, the underlying pathogenic mechanisms of PDCoV infection and the effects of PDCoV infection on host transcripts and metabolites remain incompletely understood. Methods This study investigated a combined transcriptomic and metabolomic analysis of porcine intestinal epithelial cells (IPEC-J2) following PDCoV infection by LC/MS and RNA-seq techniques. A total of 1,401 differentially expressed genes and 254 differentially accumulated metabolites were detected in the comparison group of PDCoV-infected vs. mock-infected. Results and discussion We found that PDCoV infection regulates gene sets associated with multiple signaling pathways, including the neuroactive ligand-receptor interaction, cytokine-cytokine receptor interaction, MAPK signaling pathway, chemokine signaling pathway, ras signaling pathway and so on. Besides, the metabolomic results showed that biosynthesis of cofactors, nucleotide metabolism, protein digestion and absorption, and biosynthesis of amino acid were involved in PDCoV infection. Moreover, integrated transcriptomics and metabolomics analyses revealed the involvement of ferroptosis in PDCoV infection, and exogenous addition of the ferroptosis activator erastin significantly inhibited PDCoV replication. Overall, these unique transcriptional and metabolic reprogramming features may provide a better understanding of PDCoV-infected IPEC-J2 cells and potential targets for antiviral treatment.
Collapse
Affiliation(s)
- Guangzheng Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chao Xu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuoshuo Zhang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuai Zhang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| |
Collapse
|
13
|
Hsia JZ, Liu D, Haynes L, Cruz-Cosme R, Tang Q. Lipid Droplets: Formation, Degradation, and Their Role in Cellular Responses to Flavivirus Infections. Microorganisms 2024; 12:647. [PMID: 38674592 PMCID: PMC11051834 DOI: 10.3390/microorganisms12040647] [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: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Lipid droplets (LDs) are cellular organelles derived from the endoplasmic reticulum (ER), serving as lipid storage sites crucial for maintaining cellular lipid homeostasis. Recent attention has been drawn to their roles in viral replication and their interactions with viruses. However, the precise biological functions of LDs in viral replication and pathogenesis remain incompletely understood. To elucidate the interaction between LDs and viruses, it is imperative to comprehend the biogenesis of LDs and their dynamic interactions with other organelles. In this review, we explore the intricate pathways involved in LD biogenies within the cytoplasm, encompassing the uptake of fatty acid from nutrients facilitated by CD36-mediated membranous protein (FABP/FATP)-FA complexes, and FA synthesis via glycolysis in the cytoplasm and the TCL cycle in mitochondria. While LD biogenesis primarily occurs in the ER, matured LDs are intricately linked to multiple organelles. Viral infections can lead to diverse consequences in terms of LD status within cells post-infection, potentially involving the breakdown of LDs through the activation of lipophagy. However, the exact mechanisms underlying LD destruction or accumulation by viruses remain elusive. The significance of LDs in viral replication renders them effective targets for developing broad-spectrum antivirals. Moreover, considering that reducing neutral lipids in LDs is a strategy for anti-obesity treatment, LD depletion may not pose harm to cells. This presents LDs as promising antiviral targets for developing therapeutics that are minimally or non-toxic to the host.
Collapse
Affiliation(s)
| | | | | | | | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (J.Z.H.); (D.L.); (L.H.); (R.C.-C.)
| |
Collapse
|
14
|
Zhou Y, Xu C, Gu S, Xiao Y, Wu S, Wang H, Bao W. Integrated Metabolomic and transcriptomic analyses reveal deoxycholic acid promotes transmissible gastroenteritis virus infection by inhibiting phosphorylation of NF-κB and STAT3. BMC Genomics 2024; 25:239. [PMID: 38438836 PMCID: PMC10913532 DOI: 10.1186/s12864-024-10167-8] [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: 07/06/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Acute diarrhea, dehydration and death in piglets are all symptoms of transmissible gastroenteritis virus (TGEV), which results in significant financial losses in the pig industry. It is important to understand the pathogenesis and identify new antiviral targets by revealing the metabolic interactions between TGEV and host cells. RESULTS We performed metabolomic and transcriptomic analyses of swine testicular cells infected with TGEV. A total of 1339 differential metabolites and 206 differentially expressed genes were detected post TEGV infection. The differentially expressed genes were significantly enriched in the HIF-1 signaling pathway and PI3K-Akt signaling. Integrated analysis of differentially expressed genes and differential metabolites indicated that they were significantly enriched in the metabolic processes such as nucleotide metabolism, biosynthesis of cofactors and purine metabolism. In addition, the results showed that most of the detected metabolites involved in the bile secretion was downregulated during TGEV infection. Furthermore, exogenous addition of key metabolite deoxycholic acid (DCA) significantly enhanced TGEV replication by NF-κB and STAT3 signal pathways. CONCLUSIONS We identified a significant metabolite, DCA, related to TGEV replication. It added TGEV replication in host cells by inhibiting phosphorylation of NF-κB and STAT3. This study provided novel insights into the metabolomic and transcriptomic alterations related to TGEV infection and revealed potential molecular and metabolic targets for the regulation of TGEV infection.
Collapse
Affiliation(s)
- Yajing Zhou
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China
| | - Chao Xu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China
| | - Shanshen Gu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China
| | - Yeyi Xiao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China
| | - Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China.
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design, College of Animal Science and Technology, Yangzhou University, 225009, Yangzhou, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, 225009, Yangzhou, China.
| |
Collapse
|
15
|
Wan Q, Tavakoli L, Wang TY, Tucker AJ, Zhou R, Liu Q, Feng S, Choi D, He Z, Gack MU, Zhao J. Hijacking of nucleotide biosynthesis and deamidation-mediated glycolysis by an oncogenic herpesvirus. Nat Commun 2024; 15:1442. [PMID: 38365882 PMCID: PMC10873312 DOI: 10.1038/s41467-024-45852-5] [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: 06/21/2023] [Accepted: 02/05/2024] [Indexed: 02/18/2024] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS) and multiple types of B cell malignancies. Emerging evidence demonstrates that KSHV reprograms host-cell central carbon metabolic pathways, which contributes to viral persistence and tumorigenesis. However, the mechanisms underlying KSHV-mediated metabolic reprogramming remain poorly understood. Carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotase (CAD) is a key enzyme of the de novo pyrimidine synthesis, and was recently identified to deamidate the NF-κB subunit RelA to promote aerobic glycolysis and cell proliferation. Here we report that KSHV infection exploits CAD for nucleotide synthesis and glycolysis. Mechanistically, KSHV vCyclin binds to and hijacks cyclin-dependent kinase CDK6 to phosphorylate Ser-1900 on CAD, thereby activating CAD-mediated pyrimidine synthesis and RelA-deamidation-mediated glycolytic reprogramming. Correspondingly, genetic depletion or pharmacological inhibition of CDK6 and CAD potently impeded KSHV lytic replication and thwarted tumorigenesis of primary effusion lymphoma (PEL) cells in vitro and in vivo. Altogether, our work defines a viral metabolic reprogramming mechanism underpinning KSHV oncogenesis, which may spur the development of new strategies to treat KSHV-associated malignancies and other diseases.
Collapse
Affiliation(s)
- Quanyuan Wan
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Leah Tavakoli
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Ting-Yu Wang
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Andrew J Tucker
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Ruiting Zhou
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Qizhi Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- State Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, Hunan, China
| | - Shu Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Dongwon Choi
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhiheng He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Jun Zhao
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA.
| |
Collapse
|
16
|
Peng C, Xiao P, Li N. Does oncolytic viruses-mediated metabolic reprogramming benefit or harm the immune microenvironment? FASEB J 2024; 38:e23450. [PMID: 38294796 DOI: 10.1096/fj.202301947rr] [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: 09/22/2023] [Revised: 12/11/2023] [Accepted: 01/11/2024] [Indexed: 02/01/2024]
Abstract
Oncolytic virus immunotherapy as a new tumor therapy has made remarkable achievements in clinical practice. And metabolic reprogramming mediated by oncolytic virus has a significant impact on the immune microenvironment. This review summarized the reprogramming of host cell glucose metabolism, lipid metabolism, oxidative phosphorylation, and glutamine metabolism by oncolytic virus and illustrated the effects of metabolic reprogramming on the immune microenvironment. It was found that oncolytic virus-induced reprogramming of glucose metabolism in tumor cells has both beneficial and detrimental effects on the immune microenvironment. In addition, oncolytic virus can promote fatty acid synthesis in tumor cells, inhibit oxidative phosphorylation, and promote glutamine catabolism, which facilitates the anti-tumor immune function of immune cells. Therefore, targeted metabolic reprogramming is a new direction to improve the efficacy of oncolytic virus immunotherapy.
Collapse
Affiliation(s)
- Chengcheng Peng
- Institute of Virology, Wenzhou University, Wenzhou, China
- Key Laboratory of Virology and Immunology of Wenzhou, Wenzhou University, Wenzhou, China
| | - Pengpeng Xiao
- Institute of Virology, Wenzhou University, Wenzhou, China
- Key Laboratory of Virology and Immunology of Wenzhou, Wenzhou University, Wenzhou, China
| | - Nan Li
- Institute of Virology, Wenzhou University, Wenzhou, China
- Key Laboratory of Virology and Immunology of Wenzhou, Wenzhou University, Wenzhou, China
| |
Collapse
|
17
|
Goyal P, Rajala MS. Reprogramming of glucose metabolism in virus infected cells. Mol Cell Biochem 2023; 478:2409-2418. [PMID: 36709223 PMCID: PMC9884135 DOI: 10.1007/s11010-023-04669-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 01/16/2023] [Indexed: 01/30/2023]
Abstract
Viral infection is a kind of cellular stress that leads to the changes in cellular metabolism. Many metabolic pathways in a host cell such as glycolysis, amino acid and nucleotide synthesis are altered following virus infection. Both oncogenic and non-oncogenic viruses depend on host cell glycolysis for their survival and pathogenesis. Recent studies have shown that the rate of glycolysis plays an important role in oncolysis as well by oncolytic therapeutic viruses. During infection, viral proteins interact with various cellular glycolytic enzymes, and this interaction enhances the catalytic framework of the enzymes subsequently the glycolytic rate of the cell. Increased activity of glycolytic enzymes following their interaction with viral proteins is vital for replication and to counteract the inhibition of glycolysis caused by immune response. In this review, the importance of host cell glycolysis and the modulation of glycolysis by various viruses such as oncogenic, non-oncogenic and oncolytic viruses are presented.
Collapse
Affiliation(s)
- Priya Goyal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Maitreyi S Rajala
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
18
|
Privatt SR, Braga CP, Johnson A, Lidenge SJ, Berry L, Ngowi JR, Ngalamika O, Chapple AG, Mwaiselage J, Wood C, West JT, Adamec J. Comparative polar and lipid plasma metabolomics differentiate KSHV infection and disease states. Cancer Metab 2023; 11:13. [PMID: 37653396 PMCID: PMC10470137 DOI: 10.1186/s40170-023-00316-0] [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: 04/12/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Kaposi sarcoma (KS) is a neoplastic disease etiologically associated with infection by the Kaposi sarcoma-associated herpesvirus (KSHV). KS manifests primarily as cutaneous lesions in individuals due to either age (classical KS), HIV infection (epidemic KS), or tissue rejection preventatives in transplantation (iatrogenic KS) but can also occur in individuals, predominantly in sub-Saharan Africa (SSA), lacking any obvious immune suppression (endemic KS). The high endemicity of KSHV and human immunodeficiency virus-1 (HIV) co-infection in Africa results in KS being one of the top 5 cancers there. As with most viral cancers, infection with KSHV alone is insufficient to induce tumorigenesis. Indeed, KSHV infection of primary human endothelial cell cultures, even at high levels, is rarely associated with long-term culture, transformation, or growth deregulation, yet infection in vivo is sustained for life. Investigations of immune mediators that distinguish KSHV infection, KSHV/HIV co-infection, and symptomatic KS disease have yet to reveal consistent correlates of protection against or progression to KS. In addition to viral infection, it is plausible that pathogenesis also requires an immunological and metabolic environment permissive to the abnormal endothelial cell growth evident in KS tumors. In this study, we explored whether plasma metabolomes could differentiate asymptomatic KSHV-infected individuals with or without HIV co-infection and symptomatic KS from each other. METHODS To investigate how metabolic changes may correlate with co-infections and tumorigenesis, plasma samples derived from KSHV seropositive sub-Saharan African subjects in three groups, (A) asymptomatic (lacking neoplastic disease) with KSHV infection only, (B) asymptomatic co-infected with KSHV and HIV, and (C) symptomatic with clinically diagnosed KS, were subjected to analysis of lipid and polar metabolite profiles RESULTS: Polar and nonpolar plasma metabolic differentials were evident in both comparisons. Integration of the metabolic findings with our previously reported KS transcriptomics data suggests dysregulation of amino acid/urea cycle and purine metabolic pathways, in concert with viral infection in KS disease progression. CONCLUSIONS This study is, to our knowledge, the first to report human plasma metabolic differentials between in vivo KSHV infection and co-infection with HIV, as well as differentials between co-infection and epidemic KS.
Collapse
Affiliation(s)
- Sara R Privatt
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | - Alicia Johnson
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Salum J Lidenge
- Ocean Road Cancer Institute, Dar Es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Luke Berry
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - John R Ngowi
- Ocean Road Cancer Institute, Dar Es Salaam, Tanzania
| | - Owen Ngalamika
- Dermatology and Venereology Section, Adult Hospital of the University Teaching Hospitals, University of Zambia School of Medicine, Lusaka, Zambia
| | - Andrew G Chapple
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Julius Mwaiselage
- Ocean Road Cancer Institute, Dar Es Salaam, Tanzania
- Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Charles Wood
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - John T West
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | - Jiri Adamec
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
19
|
Li J, Wang Y, Deng H, Li S, Qiu HJ. Cellular metabolism hijacked by viruses for immunoevasion: potential antiviral targets. Front Immunol 2023; 14:1228811. [PMID: 37559723 PMCID: PMC10409484 DOI: 10.3389/fimmu.2023.1228811] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023] Open
Abstract
Cellular metabolism plays a central role in the regulation of both innate and adaptive immunity. Immune cells utilize metabolic pathways to modulate the cellular differentiation or death. The intricate interplay between metabolism and immune response is critical for maintaining homeostasis and effective antiviral activities. In recent years, immunometabolism induced by viral infections has been extensively investigated, and accumulating evidence has indicated that cellular metabolism can be hijacked to facilitate viral replication. Generally, virus-induced changes in cellular metabolism lead to the reprogramming of metabolites and metabolic enzymes in different pathways (glucose, lipid, and amino acid metabolism). Metabolic reprogramming affects the function of immune cells, regulates the expression of immune molecules and determines cell fate. Therefore, it is important to explore the effector molecules with immunomodulatory properties, including metabolites, metabolic enzymes, and other immunometabolism-related molecules as the antivirals. This review summarizes the relevant advances in the field of metabolic reprogramming induced by viral infections, providing novel insights for the development of antivirals.
Collapse
Affiliation(s)
| | | | | | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
20
|
Clark SA, Vazquez A, Furiya K, Splattstoesser MK, Bashmail AK, Schwartz H, Russell M, Bhark SJ, Moreno OK, McGovern M, Owsley ER, Nelson TA, Sanchez EL, Delgado T. Rewiring of the Host Cell Metabolome and Lipidome during Lytic Gammaherpesvirus Infection Is Essential for Infectious-Virus Production. J Virol 2023; 97:e0050623. [PMID: 37191529 PMCID: PMC10308918 DOI: 10.1128/jvi.00506-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Oncogenic virus infections are estimated to cause ~15% of all cancers. Two prevalent human oncogenic viruses are members of the gammaherpesvirus family: Epstein-Barr virus (EBV) and Kaposi's sarcoma herpesvirus (KSHV). We use murine herpesvirus 68 (MHV-68), which shares significant homology with KSHV and EBV, as a model system to study gammaherpesvirus lytic replication. Viruses implement distinct metabolic programs to support their life cycle, such as increasing the supply of lipids, amino acids, and nucleotide materials necessary to replicate. Our data define the global changes in the host cell metabolome and lipidome during gammaherpesvirus lytic replication. Our metabolomics analysis found that MHV-68 lytic infection induces glycolysis, glutaminolysis, lipid metabolism, and nucleotide metabolism. We additionally observed an increase in glutamine consumption and glutamine dehydrogenase protein expression. While both glucose and glutamine starvation of host cells decreased viral titers, glutamine starvation led to a greater loss in virion production. Our lipidomics analysis revealed a peak in triacylglycerides early during infection and an increase in free fatty acids and diacylglyceride later in the viral life cycle. Furthermore, we observed an increase in the protein expression of multiple lipogenic enzymes during infection. Interestingly, pharmacological inhibitors of glycolysis or lipogenesis resulted in decreased infectious virus production. Taken together, these results illustrate the global alterations in host cell metabolism during lytic gammaherpesvirus infection, establish essential pathways for viral production, and recommend targeted mechanisms to block viral spread and treat viral induced tumors. IMPORTANCE Viruses are intracellular parasites which lack their own metabolism, so they must hijack host cell metabolic machinery in order to increase the production of energy, proteins, fats, and genetic material necessary to replicate. Using murine herpesvirus 68 (MHV-68) as a model system to understand how similar human gammaherpesviruses cause cancer, we profiled the metabolic changes that occur during lytic MHV-68 infection and replication. We found that MHV-68 infection of host cells increases glucose, glutamine, lipid, and nucleotide metabolic pathways. We also showed inhibition or starvation of glucose, glutamine, or lipid metabolic pathways results in an inhibition of virus production. Ultimately, targeting changes in host cell metabolism due to viral infection can be used to treat gammaherpesvirus-induced cancers and infections in humans.
Collapse
Affiliation(s)
- Sarah A. Clark
- Northwest University, Department of Biology, Kirkland, Washington, USA
| | - Angie Vazquez
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Kelsey Furiya
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | | | | | - Haleigh Schwartz
- Northwest University, Department of Biology, Kirkland, Washington, USA
| | - Makaiya Russell
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Shun-Je Bhark
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Osvaldo K. Moreno
- San Francisco State University, Department of Biology, San Francisco, California, USA
| | - Morgan McGovern
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Eric R. Owsley
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Timothy A. Nelson
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
| | - Erica L. Sanchez
- San Francisco State University, Department of Biology, San Francisco, California, USA
- University of Texas at Dallas, Department of Biological Sciences, Richardson, Texas, USA
| | - Tracie Delgado
- Seattle Pacific University, Department of Biology, Seattle, Washington, USA
- Northwest University, Department of Biology, Kirkland, Washington, USA
| |
Collapse
|
21
|
Gong Z, Yan Z, Liu W, Luo B. Oncogenic viruses and host lipid metabolism: a new perspective. J Gen Virol 2023; 104. [PMID: 37279154 DOI: 10.1099/jgv.0.001861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
As noncellular organisms, viruses do not have their own metabolism and rely on the metabolism of host cells to provide energy and metabolic substances for their life cycles. Increasing evidence suggests that host cells infected with oncogenic viruses have dramatically altered metabolic requirements and that oncogenic viruses produce substances used for viral replication and virion production by altering host cell metabolism. We focused on the processes by which oncogenic viruses manipulate host lipid metabolism and the lipid metabolism disorders that occur in oncogenic virus-associated diseases. A deeper understanding of viral infections that cause changes in host lipid metabolism could help with the development of new antiviral agents as well as potential new therapeutic targets.
Collapse
Affiliation(s)
- Zhiyuan Gong
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao 266071, PR China
| | - Zhiyong Yan
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao 266071, PR China
| | - Wen Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao 266071, PR China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao 266071, PR China
| |
Collapse
|
22
|
Tao L, Dryden P, Lowe A, Wang G, Achuthkumar A, Chang T, Reese TA. WY14643 Increases Herpesvirus Replication and Inhibits IFNβ Production Independently of PPARα Expression. Microbiol Spectr 2023; 11:e0233722. [PMID: 36715509 PMCID: PMC10100363 DOI: 10.1128/spectrum.02337-22] [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: 06/28/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023] Open
Abstract
Peroxisome proliferator activated receptor (PPAR) agonists are commonly used to treat metabolic disorders in humans because they regulate fatty acid oxidation and cholesterol metabolism. In addition to their roles in controlling metabolism, PPAR agonists also regulate inflammation and are immunosuppressive in models of autoimmunity. We aimed to test whether activation of PPARα with clinically relevant ligands could impact gammaherpesvirus infection using murine gammaherpesvirus-68 (MHV68, MuHV-4). We found that PPAR agonists WY14643 and fenofibrate increased herpesvirus replication in vitro. In vivo, WY14643 increased viral replication and caused lethality in mice. Unexpectedly, these effects proved independent of PPARα. We found that WY14643 suppressed production of type I interferon after MHV68 infection in vitro and in vivo. Taken together, our data indicate that caution should be employed when using PPARα agonists in immuno-metabolic studies, as they can have off-target effects on viral replication through the inhibition of type I interferon production. IMPORTANCE PPAR agonists are used clinically to treat both metabolic and inflammatory disorders. Because viruses are known to rewire host metabolism to their own benefit, the intersection of immunity, metabolism, and virology is an important research area. Our article is an important contribution to this field for two reasons. First, it shows a role for PPARα agonists in altering virus replication. Second, it shows that PPARα agonists can affect virus replication in a manner independent of their predicted target. This knowledge is valuable for anyone seeking to use PPARα agonists as a research tool.
Collapse
Affiliation(s)
- Lili Tao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Phillip Dryden
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Alexandria Lowe
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Guoxun Wang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amritha Achuthkumar
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tyron Chang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tiffany A. Reese
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
23
|
Huang P, Wang X, Lei M, Ma Y, Chen H, Sun J, Hu Y, Shi J. Metabolomics Profiles Reveal New Insights of Herpes Simplex Virus Type 1 Infection. Int J Mol Sci 2023; 24:ijms24021521. [PMID: 36675052 PMCID: PMC9862159 DOI: 10.3390/ijms24021521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/18/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a ubiquitous human pathogen that can cause significant morbidity, primarily facial cold sores and herpes simplex encephalitis. Previous studies have shown that a variety of viruses can reprogram the metabolic profiles of host cells to facilitate self-replication. In order to further elucidate the metabolic interactions between the host cell and HSV-1, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) to analyze the metabolic profiles in human lung fibroblasts KMB17 infected with HSV-1. The results showed that 654 and 474 differential metabolites were identified in positive and negative ion modes, respectively, and 169 and 114 metabolic pathways that might be altered were screened. These altered metabolites are mainly involved in central carbon metabolism, choline metabolism, amino acid metabolism, purine and pyrimidine metabolism, cholesterol metabolism, bile secretion, and prolactin signaling pathway. Further, we confirmed that the addition of tryptophan metabolite kynurenine promotes HSV-1 replication, and the addition of 25-Hydroxycholesterol inhibits viral replication. Significantly, HSV-1 replication was obviously enhanced in the ChOKα (a choline metabolic rate-limiting enzyme) deficient mouse macrophages. These results indicated that HSV-1 induces the metabolic reprogramming of host cells to promote or resist viral replication. Taken together, these observations highlighted the significance of host cell metabolism in HSV-1 replication, which would help to clarify the pathogenesis of HSV-1 and identify new anti-HSV-1 therapeutic targets.
Collapse
Affiliation(s)
- Pu Huang
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Xu Wang
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Mengyue Lei
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Ying Ma
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Hongli Chen
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Institute of Medical Biology, Kunming Medical University, Kunming 650032, China
| | - Jing Sun
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Correspondence: (J.S.); (J.S.); Tel.: +86-871-68335334 (Jiandong Shi); Fax: +86-871-68175829 (Jiandong Shi)
| | - Yunzhang Hu
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jiandong Shi
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
- Correspondence: (J.S.); (J.S.); Tel.: +86-871-68335334 (Jiandong Shi); Fax: +86-871-68175829 (Jiandong Shi)
| |
Collapse
|
24
|
An Update on the Metabolic Landscape of Oncogenic Viruses. Cancers (Basel) 2022; 14:cancers14235742. [PMID: 36497226 PMCID: PMC9738352 DOI: 10.3390/cancers14235742] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
Viruses play an important role in cancer development as about 12% of cancer types are linked to viral infections. Viruses that induce cellular transformation are known as oncoviruses. Although the mechanisms of viral oncogenesis differ between viruses, all oncogenic viruses share the ability to establish persistent chronic infections with no obvious symptoms for years. During these prolonged infections, oncogenic viruses manipulate cell signaling pathways that control cell cycle progression, apoptosis, inflammation, and metabolism. Importantly, it seems that most oncoviruses depend on these changes for their persistence and amplification. Metabolic changes induced by oncoviruses share many common features with cancer metabolism. Indeed, viruses, like proliferating cancer cells, require increased biosynthetic precursors for virion production, need to balance cellular redox homeostasis, and need to ensure host cell survival in a given tissue microenvironment. Thus, like for cancer cells, viral replication and persistence of infected cells frequently depend on metabolic changes. Here, we draw parallels between metabolic changes observed in cancers or induced by oncoviruses, with a focus on pathways involved in the regulation of glucose, lipid, and amino acids. We describe whether and how oncoviruses depend on metabolic changes, with the perspective of targeting them for antiviral and onco-therapeutic approaches in the context of viral infections.
Collapse
|
25
|
Zandi M, Shokri S, Mahmoudvand S, Hosseinzadeh Adli A, Mohammadi R, Haddadi A. Interplay between cellular metabolism and DNA viruses. J Med Virol 2022; 94:5163-5173. [PMID: 35869415 DOI: 10.1002/jmv.28018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022]
Abstract
Viruses as intracellular pathogens take over the host metabolism and reprogram to facilitate optimal virus production. DNA viruses can cause alterations in several metabolic pathways, including aerobic glycolysis also known as the Warburg effect, pentose phosphate pathway activation, and amino acid catabolism such as glutaminolysis, nucleotide biosynthesis, lipid metabolism, and amino acid biosynthesis. The available energy for productive infection can be increased in infected cells via modification of different carbon source utilization. This review discusses the metabolic alterations of the DNA viruses that will be the basis for future novel therapeutic approaches.
Collapse
Affiliation(s)
- Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Shokri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahab Mahmoudvand
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ahmad Hosseinzadeh Adli
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Mohammadi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Azita Haddadi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
26
|
Qi X, Yan Q, Shang Y, Zhao R, Ding X, Gao SJ, Li W, Lu C. A viral interferon regulatory factor degrades RNA-binding protein hnRNP Q1 to enhance aerobic glycolysis via recruiting E3 ubiquitin ligase KLHL3 and decaying GDPD1 mRNA. Cell Death Differ 2022; 29:2233-2246. [PMID: 35538151 PMCID: PMC9613757 DOI: 10.1038/s41418-022-01011-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/09/2022] Open
Abstract
Reprogramming of host metabolism is a common strategy of viral evasion of host cells, and is essential for successful viral infection and induction of cancer in the context cancer viruses. Kaposi's sarcoma (KS) is the most common AIDS-associated cancer caused by KS-associated herpesvirus (KSHV) infection. KSHV-encoded viral interferon regulatory factor 1 (vIRF1) regulates multiple signaling pathways and plays an important role in KSHV infection and oncogenesis. However, the role of vIRF1 in KSHV-induced metabolic reprogramming remains elusive. Here we show that vIRF1 increases glucose uptake, ATP production and lactate secretion by downregulating heterogeneous nuclear ribonuclear protein Q1 (hnRNP Q1). Mechanistically, vIRF1 upregulates and recruits E3 ubiquitin ligase Kelch-like 3 (KLHL3) to degrade hnRNP Q1 through a ubiquitin-proteasome pathway. Furthermore, hnRNP Q1 binds to and stabilizes the mRNA of glycerophosphodiester phosphodiesterase domain containing 1 (GDPD1). However, vIRF1 targets hnRNP Q1 for degradation, which destabilizes GDPD1 mRNA, resulting in induction of aerobic glycolysis. These results reveal a novel role of vIRF1 in KSHV metabolic reprogramming, and identifying a potential therapeutic target for KSHV infection and KSHV-induced cancers.
Collapse
Affiliation(s)
- Xiaoyu Qi
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qin Yan
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuancui Shang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Runran Zhao
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xiangya Ding
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China
| | - Shou-Jiang Gao
- Tumor Virology Program, UPMC Hillman Cancer Center, and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Wan Li
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China.
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China.
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China.
| | - Chun Lu
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China.
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China.
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China.
| |
Collapse
|
27
|
Hernández-Velázquez IM, Zamora-Briseño JA, Hernández-Bolio GI, Hernández-Nuñez E, Lozano-Álvarez E, Briones-Fourzán P, Rodríguez-Canul R. Metabolic changes in antennal glands of Caribbean spiny lobsters Panulirus argus infected by Panulirus argus virus 1 (PaV1). DISEASES OF AQUATIC ORGANISMS 2022; 151:11-22. [PMID: 36047670 DOI: 10.3354/dao03682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Panulirus argus virus 1 (PaV1) (Family Mininucleoviridae) causes chronic and systemic infection in wild juvenile spiny lobsters Panulirus argus (Latreille, 1804), ending in death by starvation and metabolic wasting. In marine decapods, the antennal gland is involved in osmoregulation and excretion. In this compact organ, fluid is filtered from the hemolymph, and ions are reabsorbed to produce a hypotonic urine. Although PaV1 is released with the urine in infected individuals, little is known regarding the metabolic effect of PaV1 in the antennal gland. The objective of this study was to perform a comparative evaluation of the metabolic profile of the antennal gland of clinically PaV1-infected lobsters versus those with no clinical signs of infection, using proton nuclear magnetic resonance analysis. Overall, 48 compounds were identified, and the most represented metabolites were those involved in carbohydrate, amino acid, energy, and nucleotide metabolism. Most of the metabolites that were down-regulated in the infected group were essential and non-essential amino acids. Some metabolites involved in the urea cycle and carbohydrate metabolism were also altered. This study represents a first approach to the metabolic evaluation of the antennal gland. We broadly discuss alterations in the content of several proteinogenic and non-proteinogenic amino acids and other key metabolites involved in energetic and nucleotide metabolism.
Collapse
Affiliation(s)
- Ioreni Margarita Hernández-Velázquez
- Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional. Carr. Mérida-Progreso, CP 97310 Mérida, Yucatán, México
| | | | | | | | | | | | | |
Collapse
|
28
|
Choi UY, Lee JJ, Park A, Jung KL, Lee SA, Choi YJ, Lee HR, Lai CJ, Eoh H, Jung JU. Herpesvirus-induced spermidine synthesis and eIF5A hypusination for viral episomal maintenance. Cell Rep 2022; 40:111234. [PMID: 35977517 DOI: 10.1016/j.celrep.2022.111234] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/16/2022] [Accepted: 07/28/2022] [Indexed: 11/03/2022] Open
Abstract
Spermidine is essential for cellular growth and acts as a prerequisite of hypusination, a post-translational modification of eukaryotic initiation factor 5A (eIF5A), allowing the translation of polyproline-containing proteins. Here, we show that oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) increases spermidine synthesis and eIF5A hypusination to enhance expression of polyproline-containing latency-associated nuclear antigen (LANA) for viral episomal maintenance. KSHV upregulates intracellular spermidine levels by dysregulating polyamine metabolic pathways in three-dimensional (3D) culture and 2D de novo infection conditions. Increased intracellular spermidine leads to increased eIF5A hypusination, ultimately enhancing LANA expression. In contrast, inhibition of spermidine synthesis or eIF5A hypusination alleviates LANA expression, decreasing viral episomal maintenance and KSHV-infected cell proliferation in vitro and in vivo, which is reversed by spermidine supplement. This demonstrates that KSHV hijacks spermidine synthesis and eIF5A hypusination pathways to enhance LANA expression for viral episomal maintenance, suggesting polyamine metabolism and eIF5A hypusination as therapeutic targets for KSHV-induced tumorigenesis.
Collapse
Affiliation(s)
- Un Yung Choi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Jae Jin Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Angela Park
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kyle L Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shin-Ae Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Youn Jung Choi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hye-Ra Lee
- Department of Biotechnology and Bioinformatics, College of Science and Technology, Korea University, Sejong 30019, Republic of Korea
| | - Chih-Jen Lai
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hyungjin Eoh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jae U Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
29
|
Ren G, Xu L, Zhao J, Shao Y, Chen X, Lu T, Zhang Q. Supplementation of Dietary Crude Lentinan Improves the Intestinal Microbiota and Immune Barrier in Rainbow Trout (Oncorhynchus mykiss) Infected by Infectious Hematopoietic Necrosis Virus. Front Immunol 2022; 13:920065. [PMID: 35812417 PMCID: PMC9258421 DOI: 10.3389/fimmu.2022.920065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
The effects of crude lentinan (CLNT) on the intestinal microbiota and the immune barrier were evaluated in rainbow trout (Oncorhynchus mykiss) infected by infectious hematopoietic necrosis virus (IHNV). The results showed that supplementary CLNT declined the rainbow trout mortality caused by IHNV, which suggested that CLNT has preventive effects on IHNV infection. IHNV destroyed intestinal integrity, as well as caused the intestinal oxidative and damage in rainbow trout. Supplementary CLNT significantly strengthened the intestinal immune barrier by declining intestinal permeability, as well as enhancing intestinal antioxidant and anti-inflammatory abilities in IHNV-infected rainbow trout (P<0.05). In addition, CLNT modified the aberrant changes of intestinal microbiota induced by IHNV, mainly represented by promoting the growths of Carnobacterium and Deefgea and inhibiting Mycobacterium and Nannocystis. Especially, supplementing with CLNT significantly promoted the growth of short-chain fatty acid–producing bacteria (P<0.05) and consequently increased the production of acetic acid, butanoic acid, and hexanoic acid in the intestine of IHNV-infected rainbow trout. Furthermore, it was speculated that CLNT could regulate the self-serving metabolic pathways of intestinal microbiota induced by IHNV, such as fatty acid metabolism and amino acid metabolism. Together, CLNT played the antiviral effects on IHNV infection through strengthening the intestinal immune barrier, as well as regulating intestinal microbiota and SCFA metabolism in rainbow trout. The present data revealed that CLNT exerted a promising prebiotic role in preventing the rainbow trout from IHNV infection.
Collapse
Affiliation(s)
- Guangming Ren
- Department of Aquatic Animal Diseases and Control, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Liming Xu
- Department of Aquatic Animal Diseases and Control, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin, China
| | - Jingzhuang Zhao
- Department of Aquatic Animal Diseases and Control, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin, China
| | - Yizhi Shao
- Department of Aquatic Animal Diseases and Control, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin, China
| | - Xiaoyu Chen
- Technology Center of Wuhan Customs, Wuhan, China
| | - Tongyan Lu
- Department of Aquatic Animal Diseases and Control, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Harbin, China
- *Correspondence: Tongyan Lu, ; Qiya Zhang,
| | - Qiya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Tongyan Lu, ; Qiya Zhang,
| |
Collapse
|
30
|
The Isolation and Full-Length Transcriptome Sequencing of a Novel Nidovirus and Response of Its Infection in Japanese Flounder (Paralichthys olivaceus). Viruses 2022; 14:v14061216. [PMID: 35746687 PMCID: PMC9230003 DOI: 10.3390/v14061216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
A novel nidovirus, CSBV Bces-Po19, was isolated from the marine fish, Japanese flounder (Paralichthys olivaceus). The viral genome was 26,597 nucleotides long and shared 98.62% nucleotide identity with CSBV WHQSR4345. PacBio Sequel and Illumina sequencing were used to perform full-length transcriptome sequencing on CSBV Bces-Po19-sensitive (S) and -resistant (R) Japanese flounder. The results of negative staining revealed bacilliform and spherical virions. There were in total 1444 different genes between CSBV Bces-Po19 S and R groups, with 935 being up-regulated and 513 being down-regulated. Metabolism-, immune-, and RNA-related pathways were significantly enriched. Furthermore, CSBV Bces-Po19 infection induced alternative splicing (AS) events in Japanese flounder; the S group had a higher numbers of AS events (12,352) than the R group (11,452). The number of long non-coding RNA (lncRNA) in the S group, on the other hand, was significantly lower than in the R group. In addition to providing valuable information that sheds more light on CSBV Bces-Po19 infection, these research findings provide further clues for CSBV Bces-Po19 prevention and treatment.
Collapse
|
31
|
Indari O, Jakhmola S, Pathak DK, Tanwar M, Kandpal M, Mishra A, Kumar R, Jha HC. Comparative Account of Biomolecular Changes Post Epstein Barr Virus Infection of the Neuronal and Glial Cells Using Raman Microspectroscopy. ACS Chem Neurosci 2022; 13:1627-1637. [PMID: 35561419 DOI: 10.1021/acschemneuro.2c00081] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Raman microspectroscopy is a vibrational spectroscopy technique used for investigating molecular fingerprints of a wide range of liquid or solid samples. The technique can be efficiently utilized to understand the virus-mediated cellular changes and could provide valuable insights into specific biomolecular alterations. The Epstein Barr virus (EBV) has been associated with various types of cancers as well as neurodegenerative diseases. However, EBV-mediated neurological ailments are yet underexplored in terms of biomolecular changes in neuronal and glial cells (astrocytes and microglia). In continuation of our earlier exploration of EBV-influenced glial cells, we tried to decipher biomolecular changes in EBV-infected neuronal cells using Raman microspectroscopy. Additionally, we compared the consecutive biomolecular changes observed in neuronal cells with both the glial cells. We observed that EBV infection gets differentially regulated in the neuronal cells, astrocytes, and microglia. The viral entry and initiation of infection-mediated cellular modulation could start as soon as 2 h post infection but may regulate a distinct biomolecular milieu in different time intervals. Similar to the early timespan, the 24-36 h interval could also be important for EBV to manipulate neuronal as well as glial cells as depicted from elevated biomolecular activities. At these time intervals, some common biomolecules such as proline, glucose, lactic acid, nucleotides, or cholesterol were observed in the cells. However, at these time intervals, some distinct biomolecules were also observed in each cell, such as collagen, lipid, and protein stretches in the neuronal nucleus (2-4 h); tyrosine and RNA in the astrocyte nucleus (2-4 h nucleus); and fatty acids in the microglia nucleus (24-36 h). The observed biomolecular entities could ultimately play pivotal roles in the viral usurpation of cells. We also provided insights into whether these biomolecular changes can be correlated to each other and mediate virus-associated manifestations which can be linked to neurological complications. Our study aids in the understanding of EBV-mediated biomolecular changes in the various compartments of the central nervous system.
Collapse
Affiliation(s)
- Omkar Indari
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Shweta Jakhmola
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Devesh K. Pathak
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
- Department of Chemical Engineering, University of Seoul, Seoul 02504, Republic of Korea
| | - Manushree Tanwar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur 342011, India
| | - Rajesh Kumar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore 453552, India
| |
Collapse
|
32
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
33
|
Mullen PJ, Christofk HR. The Metabolic Relationship Between Viral Infection and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2022. [DOI: 10.1146/annurev-cancerbio-070120-090423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viruses are fundamental tools in cancer research. They were used to discover the first oncogenes in the 1970s, and they are now being modified for use as antitumor therapeutics. Key to both of these oncogenic and oncolytic properties is the ability of viruses to rewire host cell metabolism. In this review, we describe how viral oncogenes alter metabolism to increase the synthesis of macromolecules necessary for both viral replication and tumor growth. We then describe how understanding the specific metabolic requirements of virus-infected cells can help guide strategies to improve the efficacy of oncolytic viruses, and we highlight immunometabolism and tumor microenvironment research that could also increase the therapeutic benefits of oncolytic viruses. We also describe how studies describing the therapeutic effects of dietary nutrient restriction in cancer can suggest new avenues for research into antiviral therapeutics.
Collapse
Affiliation(s)
- Peter J. Mullen
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Heather R. Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center and Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, California, USA
| |
Collapse
|
34
|
Aliyari SR, Ghaffari AA, Pernet O, Parvatiyar K, Wang Y, Gerami H, Tong AJ, Vergnes L, Takallou A, Zhang A, Wei X, Chilin LD, Wu Y, Semenkovich CF, Reue K, Smale ST, Lee B, Cheng G. Suppressing fatty acid synthase by type I interferon and chemical inhibitors as a broad spectrum anti-viral strategy against SARS-CoV-2. Acta Pharm Sin B 2022; 12:1624-1635. [PMID: 35251918 PMCID: PMC8883762 DOI: 10.1016/j.apsb.2022.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/27/2022] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is an emerging viral pathogen and a major global public health challenge since December of 2019, with limited effective treatments throughout the pandemic. As part of the innate immune response to viral infection, type I interferons (IFN-I) trigger a signaling cascade that culminates in the activation of hundreds of genes, known as interferon stimulated genes (ISGs), that collectively foster an antiviral state. We report here the identification of a group of type I interferon suppressed genes, including fatty acid synthase (FASN), which are involved in lipid metabolism. Overexpression of FASN or the addition of its downstream product, palmitate, increased viral infection while knockout or knockdown of FASN reduced infection. More importantly, pharmacological inhibitors of FASN effectively blocked infections with a broad range of viruses, including SARS-CoV-2 and its variants of concern. Thus, our studies not only suggest that downregulation of metabolic genes may present an antiviral strategy by type I interferon, but they also introduce the potential for FASN inhibitors to have a therapeutic application in combating emerging infectious diseases such as COVID-19.
Collapse
Affiliation(s)
- Saba R. Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Amir Ali Ghaffari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Olivier Pernet
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
- EnViro International Laboratories, Los Angeles, CA 90077, USA
| | - Kislay Parvatiyar
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Yao Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Hoda Gerami
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Ann-Jay Tong
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Armin Takallou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Adel Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Xiaochao Wei
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Linda D. Chilin
- Center for Infectious Disease Research, School of Systems Biology, George Mason University Manassas, VA 20110, USA
| | - Yuntao Wu
- Center for Infectious Disease Research, School of Systems Biology, George Mason University Manassas, VA 20110, USA
| | - Clay F. Semenkovich
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Diabetic Cardiovascular Disease Center, Washington, University School of Medicine, St. Louis, MO 63110, USA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stephen T. Smale
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Benhur Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
- Corresponding author. Tel.:+1 310 825 8896; fax: +1 310 206 5553.
| |
Collapse
|
35
|
Xu L, Wang Z, Chen Z, Cui L, Liu Z, Liang Y, Li X, Zhang Y, Liu S, Li H. PFT-α Inhibits Gallid Alpha Herpesvirus 1 Replication by Repressing Host Nucleotide Metabolism and ATP Synthesis. Vet Microbiol 2022; 269:109435. [DOI: 10.1016/j.vetmic.2022.109435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/06/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022]
|
36
|
Guo X, Zheng Q, Pan Z, Huang Y, Huang X, Qin Q. Singapore Grouper Iridovirus Induces Glucose Metabolism in Infected Cells by Activation of Mammalian Target of Rapamycin Signaling. Front Microbiol 2022; 13:827818. [PMID: 35432224 PMCID: PMC9006996 DOI: 10.3389/fmicb.2022.827818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Singapore grouper iridovirus (SGIV), a member of the Iridoviridae family, is an important marine cultured fish pathogen worldwide. Our previous studies have demonstrated that lipid metabolism was essential for SGIV entry and replication, but the roles of glucose metabolism during SGIV infection still remains largely unknown. In this study, we found that the transcription levels of key enzymes involved in glycolysis were regulated in varying degrees during SGIV infection based on the transcriptomic analysis. Quantitative PCR and western blot analysis also indicated that the expression of both glucose transporters (GLUT1 and GLUT2) and the enzymes of glucose metabolism (hexokinase 2, HK2 and pyruvate dehydrogenase complex, PDHX) were upregulated during SGIV infection in vivo or in vitro, suggesting that glycolysis might be involved in SGIV infection. Exogenous glucose supplementation promoted the expression of viral genes and infectious virion production, while glutamine had no effect on SGIV infection, indicating that glucose was required for SGIV replication. Consistently, pharmacological inhibition of glycolysis dramatically reduced the protein synthesis of SGIV major capsid protein (MCP) and infectious virion production, and promotion of glycolysis significantly increased SGIV infection. Furthermore, knockdown of HK2, PDHX, or GLUT1 by siRNA decreased the transcription and protein synthesis of SGIV MCP and suppressed viral replication, indicating that those enzymes exerted essential roles in SGIV replication. In addition, inhibition of mTOR activity in SGIV-infected cells effectively reduced the expression of glycolysis key enzymes, including HK2, PDHX, GLUT1, and GLUT2, and finally inhibited SGIV replication, suggesting that mTOR was involved in SGIV-induced glycolysis. Thus, our results not only provided new insights into the mechanism of how SGIV infection affects host cell glycolysis, but also contributed to further understanding of the iridovirus pathogenesis.
Collapse
Affiliation(s)
- Xixi Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Qi Zheng
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Zanbin Pan
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Youhua Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Xiaohong Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Qiwei Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
37
|
Allen CNS, Arjona SP, Santerre M, Sawaya BE. Hallmarks of Metabolic Reprogramming and Their Role in Viral Pathogenesis. Viruses 2022; 14:602. [PMID: 35337009 PMCID: PMC8955778 DOI: 10.3390/v14030602] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer and has proven to be critical in viral infections. Metabolic reprogramming provides the cell with energy and biomass for large-scale biosynthesis. Based on studies of the cellular changes that contribute to metabolic reprogramming, seven main hallmarks can be identified: (1) increased glycolysis and lactic acid, (2) increased glutaminolysis, (3) increased pentose phosphate pathway, (4) mitochondrial changes, (5) increased lipid metabolism, (6) changes in amino acid metabolism, and (7) changes in other biosynthetic and bioenergetic pathways. Viruses depend on metabolic reprogramming to increase biomass to fuel viral genome replication and production of new virions. Viruses take advantage of the non-metabolic effects of metabolic reprogramming, creating an anti-apoptotic environment and evading the immune system. Other non-metabolic effects can negatively affect cellular function. Understanding the role metabolic reprogramming plays in viral pathogenesis may provide better therapeutic targets for antivirals.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
- Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
38
|
Liu F, Ni B, Wei R. Senecavirus A- and Non-Infected Cells at Early Stage of Infection: Comparative Metabolomic Profiles. Front Cell Infect Microbiol 2022; 11:736506. [PMID: 35071028 PMCID: PMC8776658 DOI: 10.3389/fcimb.2021.736506] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Senecavirus A (SVA), classified into the genus Senecavirus in the family Picornaviridae, causes an infectious disease in pigs. This virus can efficiently replicate in some non-pig-derived cells, such as the BHK cell line and its derivative (BSR-T7/5 cell line). We had recovered a wild-type SVA from its cDNA clone previously, and then uncovered the proteomic profile of SVA-infected BSR-T7/5 cells at 12 h post inoculation (hpi). In order to explore the cellular metabolomics further, the SVA-inoculated BSR-T7/5 cell monolayer was collected at 12 hpi for assay via liquid chromatography-tandem mass spectrometry (LC-MS/MS). The resultant data set was comprehensively analyzed using bioinformatics tools. A total of 451 metabolites were identified using in-house and public databases. Out of these metabolites, sixty-one showed significantly differential values (p value < 0.05). The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to analyze metabolic pathways of the significantly differential metabolites. There were eighty-one identified KEGG pathways, out of which twenty-seven showed their p values < 0.05. The pyrimidine metabolism revealed the minimum p value and the maximum number of significantly differential metabolites, implying the pyrimidine played a key role in cellular metabolism after SVA infection. SVA replication must rely on the cellular metabolism. The present study on metabolomics would shed light on impacts of SVA-induced multiple interactions among metabolites on cells or even on natural hosts.
Collapse
Affiliation(s)
- Fuxiao Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Bo Ni
- Surveillance Laboratory of Livestock Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Rong Wei
- Surveillance Laboratory of Livestock Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| |
Collapse
|
39
|
Miller L, Berber E, Sumbria D, Rouse BT. Controlling the Burden of COVID-19 by Manipulating Host Metabolism. Viral Immunol 2022; 35:24-32. [PMID: 34905407 PMCID: PMC8863913 DOI: 10.1089/vim.2021.0150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the coronavirus-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to cause global health problems, but its impact would be minimized if the many effective vaccines that have been developed were available and in widespread use by all societies. This ideal situation is not occurring so other means of controlling COVID-19 are needed. In this short review, we make the case that manipulating host metabolic pathways could be a therapeutic approach worth exploring. The rationale for such an approach comes from the fact that viruses cause metabolic changes in cells they infect, effective host defense mechanisms against viruses requires the activity of one or more metabolic pathways, and that hosts with metabolic defects such as diabetes are more susceptible to severe consequences after COVID-19. We describe the types of approaches that could be used to redirect various aspects of host metabolism and the success that some of these maneuvers have had at controlling other virus infections. Manipulating metabolic activities to control the outcome of COVID-19 has to date received minimal attention. Manipulating host metabolism will never replace vaccines to control COVID-19 but could be used as an adjunct therapy to the extent of ongoing infection.
Collapse
Affiliation(s)
- Logan Miller
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Engin Berber
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Deepak Sumbria
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Barry T. Rouse
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
40
|
Awad K, Maghraby AS, Abd-Elshafy DN, Bahgat MM. Carbohydrates Metabolic Signatures in Immune Cells: Response to Infection. Front Immunol 2022; 13:912899. [PMID: 35983037 PMCID: PMC9380592 DOI: 10.3389/fimmu.2022.912899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Metabolic reprogramming in immune cells is diverse and distinctive in terms of complexity and flexibility in response to heterogeneous pathogenic stimuli. We studied the carbohydrate metabolic changes in immune cells in different types of infectious diseases. This could help build reasonable strategies when understanding the diagnostics, prognostics, and biological relevance of immune cells under alternative metabolic burdens. METHODS Search and analysis were conducted on published peer-reviewed papers on immune cell metabolism of a single pathogen infection from the four known types (bacteria, fungi, parasites, and viruses). Out of the 131 selected papers based on the PIC algorithm (pathogen type/immune cell/carbohydrate metabolism), 30 explored immune cell metabolic changes in well-studied bacterial infections, 17 were on fungal infections of known medical importance, and 12 and 57 were on parasitic and viral infections, respectively. RESULTS AND DISCUSSION While carbohydrate metabolism in immune cells is signaled by glycolytic shift during a bacterial or viral infection, it is widely evident that effector surface proteins are expressed on the surface of parasites and fungi to modulate metabolism in these cells. CONCLUSIONS Carbohydrate metabolism in immune cells can be categorized according to the pathogen or the disease type. Accordingly, this classification can be used to adopt new strategies in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Kareem Awad
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- *Correspondence: Kareem Awad, ; Mahmoud Mohamed Bahgat, ,
| | - Amany Sayed Maghraby
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
| | - Dina Nadeem Abd-Elshafy
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
- Department of Water Pollution Research, Institute of Environmental Research, National Research Center, Cairo, Egypt
| | - Mahmoud Mohamed Bahgat
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
- *Correspondence: Kareem Awad, ; Mahmoud Mohamed Bahgat, ,
| |
Collapse
|
41
|
Havranek KE, Reyes Ballista JM, Hines KM, Brindley MA. Untargeted Lipidomics of Vesicular Stomatitis Virus-Infected Cells and Viral Particles. Viruses 2021; 14:v14010003. [PMID: 35062207 PMCID: PMC8778780 DOI: 10.3390/v14010003] [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: 10/28/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 11/16/2022] Open
Abstract
The viral lifecycle is critically dependent upon host lipids. Enveloped viral entry requires fusion between viral and cellular membranes. Once an infection has occurred, viruses may rely on host lipids for replication and egress. Upon exit, enveloped viruses derive their lipid bilayer from host membranes during the budding process. Furthermore, host lipid metabolism and signaling are often hijacked to facilitate viral replication. We employed an untargeted HILIC-IM-MS lipidomics approach and identified host lipid species that were significantly altered during vesicular stomatitis virus (VSV) infection. Many glycerophospholipid and sphingolipid species were modified, and ontological enrichment analysis suggested that the alterations to the lipid profile change host membrane properties. Lysophosphatidylcholine (LPC), which can contribute to membrane curvature and serve as a signaling molecule, was depleted during infection, while several ceramide sphingolipids were augmented during infection. Ceramide and sphingomyelin lipids were also enriched in viral particles, indicating that sphingolipid metabolism is important during VSV infection.
Collapse
Affiliation(s)
- Katherine E. Havranek
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (K.E.H.); (J.M.R.B.)
| | - Judith Mary Reyes Ballista
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (K.E.H.); (J.M.R.B.)
| | - Kelly Marie Hines
- Department of Chemistry, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602, USA
- Correspondence: (K.M.H.); (M.A.B.); Tel.: +1-706-542-1966 (K.M.H.); +1-706-542-5796 (M.A.B.)
| | - Melinda Ann Brindley
- Department of Infectious Diseases, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
- Correspondence: (K.M.H.); (M.A.B.); Tel.: +1-706-542-1966 (K.M.H.); +1-706-542-5796 (M.A.B.)
| |
Collapse
|
42
|
Li T, Gao SJ. Metabolic reprogramming and metabolic sensors in KSHV-induced cancers and KSHV infection. Cell Biosci 2021; 11:176. [PMID: 34579773 PMCID: PMC8475840 DOI: 10.1186/s13578-021-00688-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/10/2021] [Indexed: 12/05/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus associated with several human cancers. KSHV infection and KSHV-induced anabolic cell proliferation and cellular transformation depend on reprogramming of cellular metabolic pathways, which provide the building blocks and energy for the growth of both the virus and the infected cells. Furthermore, KSHV dysregulates numerous metabolic sensors including mTOR, AMPK, CASTOR1 and sirtuins to maintain cellular energetic homeostasis during infection and in KSHV-induced cancers. In this review, we summarize the recent advances in the understanding of KSHV hijacking of metabolic pathways and sensors, providing insights into the molecular basis of KSHV infection and KSHV-induced oncogenesis. In addition, we highlight the critical metabolic targets and sensors for developing potential new therapies against KSHV infection and KSHV-induced cancers.
Collapse
Affiliation(s)
- Tingting Li
- Cancer Virology Program, UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center, Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
43
|
Koufaris C, Nicolaidou V. Glutamine addiction in virus-infected mammalian cells: A target of the innate immune system? Med Hypotheses 2021; 153:110620. [PMID: 34130112 DOI: 10.1016/j.mehy.2021.110620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/30/2021] [Accepted: 05/31/2021] [Indexed: 12/19/2022]
Abstract
Control of core cell metabolism is a key aspect of the evolutionary conflict between viruses and the host's defence mechanisms. From their side, the invading viruses press the accelerator on their host cell's glycolysis, fatty acid, and glutaminolytic metabolic processes among others. It is also well established that activation of innate immune system responses modulates facets of metabolism such as that of polyamine, cholesterol, tryptophan and many more. But what about glutamine, a proteogenic amino acid that is a crucial nutrient for multiple cellular biosynthetic processes? Although mammalian cells can normally synthesize glutamine de novo, it has been noted that infections with genetically and phylogenetically diverse viruses are followed by the acquisition of a dependency on supplies of exogenous glutamine i.e. "glutamine addiction". Here we present our novel hypothesis that glutamine metabolism is also a target of the innate immune system, possibly through the action of interferons, as part of the evolutionary conserved antiviral metabolic reprogramming.
Collapse
Affiliation(s)
- C Koufaris
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - V Nicolaidou
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus.
| |
Collapse
|
44
|
Dzobo K. The Role of Viruses in Carcinogenesis and Molecular Targeting: From Infection to Being a Component of the Tumor Microenvironment. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:358-371. [PMID: 34037476 DOI: 10.1089/omi.2021.0052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
About a tenth of all cancers are caused by viruses or associated with viral infection. Recent global events including the coronavirus disease-2019 (COVID-19) pandemic means that human encounter with viruses is increased. Cancer development in individuals with viral infection can take many years after infection, demonstrating that the involvement of viruses in cancer development is a long and complex process. This complexity emanates from individual genetic heterogeneity and the many steps involved in cancer development owing to viruses. The process of tumorigenesis is driven by the complex interaction between several viral factors and host factors leading to the creation of a tumor microenvironment (TME) that is ideal and promotes tumor formation. Viruses associated with human cancers ensure their survival and proliferation through activation of several cellular processes including inflammation, migration, and invasion, resistance to apoptosis and growth suppressors. In addition, most human oncoviruses evade immune detection and can activate signaling cascades including the PI3K-Akt-mTOR, Notch and Wnt pathways associated with enhanced proliferation and angiogenesis. This expert review examines and synthesizes the multiple biological factors related to oncoviruses, and the signaling cascades activated by these viruses contributing to viral oncogenesis. In particular, I examine and review the Epstein-Barr virus, human papillomaviruses, and Kaposi's sarcoma herpes virus in a context of cancer pathogenesis. I conclude with a future outlook on therapeutic targeting of the viruses and their associated oncogenic pathways within the TME. These anticancer strategies can be in the form of, but not limited to, antibodies and inhibitors.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
45
|
Liu X, Zhu C, Wang Y, Wei F, Cai Q. KSHV Reprogramming of Host Energy Metabolism for Pathogenesis. Front Cell Infect Microbiol 2021; 11:621156. [PMID: 34055662 PMCID: PMC8153180 DOI: 10.3389/fcimb.2021.621156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 04/27/2021] [Indexed: 12/16/2022] Open
Abstract
Reprogramming of energy metabolism is a key for cancer development. Kaposi’s sarcoma-associated herpesvirus (KSHV), a human oncogenic herpesvirus, is tightly associated with several human malignancies by infecting B-lymphocyte or endothelial cells. Cancer cell energy metabolism is mainly dominated by three pathways of central carbon metabolism, including aerobic glycolysis, glutaminolysis, and fatty acid synthesis. Increasing evidence has shown that KSHV infection can alter central carbon metabolic pathways to produce biomass for viral replication, as well as the survival and proliferation of infected cells. In this review, we summarize recent studies exploring how KSHV manipulates host cell metabolism to promote viral pathogenesis, which provides the potential therapeutic targets and strategies for KSHV-associated cancers.
Collapse
Affiliation(s)
- Xiaoqing Liu
- Ministry of Education (MOE) & National Health Committee (NHC) & Chinese Academy of Medical Science (CAMS), Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Caixia Zhu
- Ministry of Education (MOE) & National Health Committee (NHC) & Chinese Academy of Medical Science (CAMS), Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuyan Wang
- Ministry of Education (MOE) & National Health Committee (NHC) & Chinese Academy of Medical Science (CAMS), Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qiliang Cai
- Ministry of Education (MOE) & National Health Committee (NHC) & Chinese Academy of Medical Science (CAMS), Key Laboratory of Medical Molecular Virology, Department of Medical Microbiology and Parasitology, School of Basic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Magon KL, Parish JL. From infection to cancer: how DNA tumour viruses alter host cell central carbon and lipid metabolism. Open Biol 2021; 11:210004. [PMID: 33653084 PMCID: PMC8061758 DOI: 10.1098/rsob.210004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
Infections cause 13% of all cancers globally, and DNA tumour viruses account for almost 60% of these cancers. All viruses are obligate intracellular parasites and hijack host cell functions to replicate and complete their life cycles to produce progeny virions. While many aspects of viral manipulation of host cells have been studied, how DNA tumour viruses manipulate host cell metabolism and whether metabolic alterations in the virus life cycle contribute to carcinogenesis are not well understood. In this review, we compare the differences in central carbon and fatty acid metabolism in host cells following infection, oncogenic transformation, and virus-driven cancer of DNA tumour viruses including: Epstein-Barr virus, hepatitis B virus, human papillomavirus, Kaposi's sarcoma-associated herpesvirus and Merkel cell polyomavirus.
Collapse
Affiliation(s)
- Kamini L. Magon
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Joanna L. Parish
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
47
|
Epstein-Barr Virus-Encoded Latent Membrane Protein 1 and B-Cell Growth Transformation Induce Lipogenesis through Fatty Acid Synthase. J Virol 2021; 95:JVI.01857-20. [PMID: 33208446 PMCID: PMC7851568 DOI: 10.1128/jvi.01857-20] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022] Open
Abstract
Despite many attempts to develop novel therapies, EBV-specific therapies currently remain largely investigational, and EBV-associated malignancies are often associated with a worse prognosis. Therefore, there is a clear demand for EBV-specific therapies for both prevention and treatment of virus-associated malignancies. Latent membrane protein 1 (LMP1) is the major transforming protein of Epstein-Barr virus (EBV) and is critical for EBV-induced B-cell transformation in vitro. Several B-cell malignancies are associated with latent LMP1-positive EBV infection, including Hodgkin’s and diffuse large B-cell lymphomas. We have previously reported that promotion of B cell proliferation by LMP1 coincided with an induction of aerobic glycolysis. To further examine LMP1-induced metabolic reprogramming in B cells, we ectopically expressed LMP1 in an EBV-negative Burkitt’s lymphoma (BL) cell line preceding a targeted metabolic analysis. This analysis revealed that the most significant LMP1-induced metabolic changes were to fatty acids. Significant changes to fatty acid levels were also found in primary B cells following EBV-mediated B-cell growth transformation. Ectopic expression of LMP1- and EBV-mediated B-cell growth transformation induced fatty acid synthase (FASN) and increased lipid droplet formation. FASN is a crucial lipogenic enzyme responsible for de novo biogenesis of fatty acids in transformed cells. Furthermore, inhibition of lipogenesis caused preferential killing of LMP1-expressing B cells and significantly hindered EBV immortalization of primary B cells. Finally, our investigation also found that USP2a, a ubiquitin-specific protease, is significantly increased in LMP1-positive BL cells and mediates FASN stability. Our findings demonstrate that ectopic expression of LMP1- and EBV-mediated B-cell growth transformation leads to induction of FASN, fatty acids, and lipid droplet formation, possibly pointing to a reliance on lipogenesis. Therefore, the use of lipogenesis inhibitors could be used in the treatment of LMP1+ EBV-associated malignancies by targeting an LMP1-specific dependency on lipogenesis. IMPORTANCE Despite many attempts to develop novel therapies, EBV-specific therapies currently remain largely investigational, and EBV-associated malignancies are often associated with a worse prognosis. Therefore, there is a clear demand for EBV-specific therapies for both prevention and treatment of virus-associated malignancies. Noncancerous cells preferentially obtain fatty acids from dietary sources, whereas cancer cells will often produce fatty acids themselves by de novo lipogenesis, often becoming dependent on the pathway for cell survival and proliferation. LMP1- and EBV-mediated B-cell growth transformation leads to induction of FASN, a key enzyme responsible for the catalysis of endogenous fatty acids. Preferential killing of LMP1-expressing B cells following inhibition of FASN suggests that targeting LMP-induced lipogenesis is an effective strategy in treating LMP1-positive EBV-associated malignancies. Importantly, targeting unique metabolic perturbations induced by EBV could be a way to explicitly target EBV-positive malignancies and distinguish their treatment from EBV-negative counterparts.
Collapse
|
48
|
Kuang J, Xu P, Shi Y, Yang Y, Liu P, Chen S, Zhou C, Li G, Zhuang Y, Hu R, Hu G, Guo X. Nephropathogenic Infectious Bronchitis Virus Infection Altered the Metabolome Profile and Immune Function of the Bursa of Fabricius in Chicken. Front Vet Sci 2021; 7:628270. [PMID: 33553290 PMCID: PMC7858655 DOI: 10.3389/fvets.2020.628270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023] Open
Abstract
Infectious bronchitis is a highly contagious, acute viral respiratory disease of chickens, regardless of the strain, and its infection may lead to considerable economic losses to the poultry industry. New nephropathogenic infectious bronchitis virus (NIBV) strains have increasingly emerged in recent years; hence, evaluating their infection-influenced immune function changes and the alteration of metabolite profiling is important. Initially, chickens were randomly distributed into two groups: the control group (Con) and the disease group (Dis). Here, the partial cytokines were examined, and the metabolome alterations of the bursa of Fabricius (BF) in NIBV infections in chickens were profiled by gas chromatography time-of-flight/mass spectrometry (GC-TOF/MS). The results revealed that the NIBV infection promotes the mRNA expression of inflammatory cytokines. Metabolic profile analysis indicated that clustering differed between the two groups and there were 75 significantly different metabolites detected between the two groups, suggesting that the host metabolism was significantly changed by NIBV infection. Notably, the following 12 metabolites were identified as the potential biomarkers: 3-phenyllactic acid, 2-deoxytetronic acid, aminomalonic acid, malonamide 5, uric acid, arachidonic acid, 2-methylglutaric acid, linoleic acid, ethanolamine, stearic acid, N-alpha-acetyl-l-ornithine, and O-acetylserine. Furthermore, the results of the correlation analysis showed that a strong correlation existed between metabolic biomarkers and inflammatory cytokines. Our results describe an immune and metabolic profile for the BF of chickens when infected with NIBV and provide new biomarkers of NIBV infection as potential targets and indicators of indicating therapeutic efficacy.
Collapse
Affiliation(s)
- Jun Kuang
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Puzhi Xu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yan Shi
- School of Computer and Information Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Yitian Yang
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Ping Liu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Shupeng Chen
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Changming Zhou
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Guyue Li
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Ruiming Hu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
49
|
The Crosstalk of Epigenetics and Metabolism in Herpesvirus Infection. Viruses 2020; 12:v12121377. [PMID: 33271926 PMCID: PMC7760534 DOI: 10.3390/v12121377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/23/2020] [Accepted: 11/29/2020] [Indexed: 01/31/2023] Open
Abstract
Epigenetics is a versatile player in manipulating viral infection and a potential therapeutic target for the treatment of viral-induced diseases. Both epigenetics and metabolism are crucial in establishing a highly specific transcriptional network, which may promote or suppress virus infection. Human herpesvirus infection can induce a broad range of human malignancies and is largely dependent on the status of cellular epigenetics as well as its related metabolism. However, the crosstalk between epigenetics and metabolism during herpesvirus infection has not been fully explored. Here, we describe how epigenetic regulation of cellular metabolism affects herpesvirus infection and induces viral diseases. This further highlights the importance of epigenetics and metabolism during viral infection and provides novel insights into the development of targeted therapies.
Collapse
|
50
|
Dyer A, Frost S, Fisher KD, Seymour LW. The role of cancer metabolism in defining the success of oncolytic viro-immunotherapy. Cytokine Growth Factor Rev 2020; 56:115-123. [PMID: 32921554 DOI: 10.1016/j.cytogfr.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022]
Abstract
Oncolytic viruses infect, replicate in, and kill cancer cells selectively without harming normal cells. The rapidly expanding clinical development of oncolytic virotherapy is an exciting interdisciplinary field that provides insights into virology, oncology, and immunotherapy. Recent years have seen greater focus on rational design of cancer-selective viruses together with strategies to exploit their immunostimulatory capabilities, ultimately to develop powerful oncolytic cancer vaccines. However, despite great interest in the field, many important experiments are still conducted under optimum conditions in vitro, with many nutrients present in excess and with cellular stress kept to a minimum. Whilst this provides a convenient platform for cell culture, it bears little relation to the typical conditions found within a tumour in vivo, where cells are often subject to a range of metabolic and environmental stresses. Viral infection and cancer will both lead to production of metabolites that are also not present in media in vitro. Understanding how oncolytic viruses interact with cells exposed to more representative metabolic conditions in vitro represents an under-explored area of study that could provide valuable insight into the intelligent design of superior oncolytic viruses and help bridge the gap between bench and bedside. This review summarises the major metabolic pathways altered in cancer cells, during viral infection and highlights possible targets for future studies.
Collapse
Affiliation(s)
- Arthur Dyer
- Department of Oncology, University of Oxford, Oxford, UK
| | - Sally Frost
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, Oxford, UK
| | - Len W Seymour
- Department of Oncology, University of Oxford, Oxford, UK; Old Road Campus Research Building, Department of Oncology, University of Oxford, OX37DQ, UK.
| |
Collapse
|