1
|
Chu W, Sun X, Yan Y. Study on the regulation of renal tubular cell apoptosis by SIRT1/NF-κB signaling pathway in septic acute kidney injury. Ren Fail 2025; 47:2499904. [PMID: 40329161 PMCID: PMC12057794 DOI: 10.1080/0886022x.2025.2499904] [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: 10/10/2024] [Revised: 03/18/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
SIRT1 regulates gene transcription via various signaling pathways, mitigating oxidative stress damage in renal tubular epithelial cells, reducing renal inflammation, and decreasing apoptosis in tubular cells. This study explores the mechanisms of action of SIRT1 in sepsis-induced acute kidney injury (AKI), offering a theoretical foundation for future treatments. Experiments were carried out in a CLP mouse model and an in vitro model using LPS-stimulated HK-2 cells. Immunoblotting and ELISA were employed to assess the expression levels of inflammatory cytokines (p < 0.01), finding that SIRT1 effectively reduces the inflammatory response in sepsis-induced AKI. Moreover, the detection of cell apoptosis via multiple pathways showed that SIRT1 can reduce the rate of cell apoptosis and effectively decrease oxidative stress in the validation reaction. Transmission electron microscopy observations further supported these findings, demonstrating that SIRT1 expression induces the blockade of cell apoptosis processes. The biochemical experiments concluded that SIRT1 ameliorates sepsis-induced AKI. Consequently, SIRT1 may represent a novel therapeutic target for AKI.
Collapse
Affiliation(s)
- Weiwei Chu
- Cadre Health Center, Shaoxing People’s Hospital, Shaoxing, China
| | - Xuedong Sun
- Intensive Care Unit, Shaoxing People’s Hospital, Shaoxing, China
| | - Yihe Yan
- Intensive Care Unit, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
2
|
Chang H, Wang J, Yang Y, Jiang Z, Chen Y, Lao Z. A mitochondria targeted hydrogen peroxide fluorescent probe for monitoring oxidative stress during influenza virus infection. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 334:125929. [PMID: 40020499 DOI: 10.1016/j.saa.2025.125929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Hydrogen peroxide (H2O2), an important marker of oxidative stress, plays a significant role in infectious diseases. Oxidative stress induced by influenza virus infection is intricately linked to the pathological processes of host cells. In this work, a fluorescent probe QLC1 based on the coumarin was developed for fluorescence imaging of mitochondrial H2O2 level in host cells during influenza virus infection. The self-immolative reaction of QLC1 triggered by H2O2 will lead to a 250-fold fluorescence enhance and the limit of detection was determined to be 0.176 μM. Using this probe, we monitored oxidative stress during influenza infection and identified a link between redox status and influenza virus replication.
Collapse
Affiliation(s)
- Hao Chang
- Department of Laboratory Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Jianing Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Yang Yang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin 300457, China
| | - Zhenyou Jiang
- Department of Microbiology and Immunology,Basic Medicine College,Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China.
| | - Yue Chen
- Department of Laboratory Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China.
| | - Zhiqi Lao
- Department of Laboratory Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China; Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
3
|
Guan Y, Li L, Yang R, Lu Y, Tang J. Targeting mitochondria with natural polyphenols for treating Neurodegenerative Diseases: a comprehensive scoping review from oxidative stress perspective. J Transl Med 2025; 23:572. [PMID: 40410831 PMCID: PMC12100838 DOI: 10.1186/s12967-025-06605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
Neurodegenerative diseases are a class of conditions with widespread detrimental impacts, currently lacking effective therapeutic drugs. Recent studies have identified mitochondrial dysfunction and the resultant oxidative stress as crucial contributors to the pathogenesis of neurodegenerative diseases. Polyphenols, naturally occurring compounds with inherent antioxidant properties, have demonstrated the potential to target mitochondria and mitigate oxidative stress. This therapeutic potential has garnered significant attention in recent years. Investigating the mitochondrial targeting capacity of polyphenols, their role in functional regulation, and their ability to modulate oxidative stress, along with exploring novel technologies and strategies for modifying polyphenol compounds and their formulations, holds promise for providing new avenues for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yueyue Guan
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Lei Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Rui Yang
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Yun Lu
- Department of Emergency Medicine, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Jun Tang
- Department of Encephalopathy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China.
| |
Collapse
|
4
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
5
|
Wang H, Wang Z, Zhang X, Shan L, Liu L, Hu Y, Chen J. Arctigenin derivative TZOA restores mitochondrial homeostasis to combat rhabdovirus infections. Eur J Med Chem 2025; 289:117439. [PMID: 40015160 DOI: 10.1016/j.ejmech.2025.117439] [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: 10/20/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Rhabdoviruses are diverse pathogens known for their broad host range and significant economic and health impacts. Infectious hematopoietic necrosis virus (IHNV), a member of the Novirhabdovirus genus, poses a major threat to aquaculture, particularly affecting rainbow trout. In this study, we further optimize the antiviral properties of arctigenin derivatives based on our previous structure-activity relationship (SAR) research, leading to the synthesis of TZOA. TZOA was synthesized with a 45 % yield and demonstrated no cytotoxicity up to 25 μM in EPC cells. Treatment with TZOA markedly inhibited IHNV replication dose-dependently, achieving over 90 % suppression of viral N, G, and M genes at 25 μM. Notably, TZOA effectively reduced viral titers compared to controls, demonstrating its potent antiviral activity in vitro. Mechanistically, TZOA preserved mitochondrial integrity, mitigated virus-induced mitochondrial fragmentation, and maintained membrane potential in infected cells. Furthermore, TZOA facilitated mitochondrial fusion and mitophagy, clearing damaged mitochondria, which restored MAVS-mediated interferon expression, thus enhancing the host's innate antiviral response. In vivo studies in juvenile rainbow trout revealed a significant 44 % increase in survival rates with TZOA treatment, accompanied by reduced IHNV-induced mortality and viral gene expression in spleen and kidney tissues. Importantly, TZOA also inhibited IHNV horizontal transmission, highlighting its potential application in controlling viral spread. These findings emphasize TZOA as a promising therapeutic candidate, not only for IHNV but also for broader rhabdovirus infections, offering valuable insights for antiviral drug development in aquaculture and beyond.
Collapse
Affiliation(s)
- Huan Wang
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China
| | - Zixuan Wang
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China
| | - Xu Zhang
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China
| | - Lipeng Shan
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China
| | - Lei Liu
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China
| | - Yang Hu
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, 315832, China.
| |
Collapse
|
6
|
Yu T, Li M, Li M, Zhang Q, Zhang H, Jiang Z, Wang S, Mao H, Li D, Fan L, Hu C, Xu X. Zebrafish TDP43 positively regulates p65-mediated apoptotic pathway. Int J Biol Macromol 2025; 308:142599. [PMID: 40157684 DOI: 10.1016/j.ijbiomac.2025.142599] [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: 01/15/2025] [Revised: 03/03/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
TAR DNA-binding protein 43 (TDP43) is a multifunctional RNA/DNA binding protein that serves as a hallmark of neurodegeneration in amyotrophic lateral sclerosis (ALS) and is associated with the inflammatory response related to nuclear factor κB (NF-κB) pathway. However, the relationship between TDP43 and NF-κB is not well known. In this study, zebrafish TDP43 (DrTDP43) can be induced by grass carp reovirus (GCRV) or spring viremia of carp virus (SVCV). DrTDP43 enhances the nuclear factor-kappaB (NF-κB) activity and the expression of p65 and TNFα, as well as promotes the phosphorylation of p65 in response to stimulation of GCRV and SVCV. Further assays indicate that DrTDP43 primarily resides in the nucleus and interacts with p65 via its RRM1. DrTDP43 is required for p65 to induce pro-inflammatory cytokine production (IL-6, IL-10, TNFα, IL-1β). It disrupts mitochondrial membrane potential and exacerbates apoptosis via downregulating Bcl2 and upregulating Bax, caspase3, and eIF2α. Moreover, knockdown of TDP43 decreases the content of reactive oxygen species (ROS) and the number of apoptotic cells in zebrafish larvae, which is attributed to the lower lever of p65 phosphorylation and expression of TNFα, Bax and cleaved-caspase3. In a word, these results establish TDP43 as a critical activator of the NF-κB-mediated apoptotic pathway during antiviral responses, which reveals a previously unrecognized host defense mechanism.
Collapse
Affiliation(s)
- Tingting Yu
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Miaomiao Li
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Meifeng Li
- Institute of Pathogenic Microorganism and College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Quanling Zhang
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Hongying Zhang
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Zeyin Jiang
- College of Food Science&Technology, Nanchang University, Nanchang 330039, China
| | - Shanghong Wang
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Huiling Mao
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Dongming Li
- Fuzhou Medical College, Nanchang University, Fuzhou 344000, Jiangxi, China
| | - Lihua Fan
- School of Life Science, Nanchang University, Nanchang 330031, China
| | - Chengyu Hu
- School of Life Science, Nanchang University, Nanchang 330031, China.
| | - Xiaowen Xu
- School of Life Science, Nanchang University, Nanchang 330031, China; Chongqing Research Institute of Nanchang University, 402660, China.
| |
Collapse
|
7
|
He H, Yuan K, Pan J, Weng S, Li C, Chen Y, He J. Shrimp Virus Regulates ROS Dynamics via the Nrf2 Pathway to Facilitate Viral Replication. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407695. [PMID: 40091388 PMCID: PMC12079347 DOI: 10.1002/advs.202407695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Reactive oxygen species (ROS) of hosts are widely involved in intracellular signaling and against pathogens. Viruses manipulate ROS homeostasis of hosts as a strategy to evade ROS-mediated negative effects of their infection, but the mechanisms remain unclear. The economically important aquaculture shrimp, Litopenaeus vannamei, is selected to investigate the molecular mechanism of how white spot syndrome virus (WSSV) regulates ROS dynamics and enhances viral replication. WSSV protein wsv220 binds to the repressor of shrimp nuclear factor erythroid 2-related factor 2 (LvNrf2), called Kelch-like ECH-associated protein 1 (LvKeap1), disrupting LvNrf2/LvKeap1 complex and facilitating LvNrf2 nuclear translocation. This activation of LvNrf2 causes up-regulation of antioxidant genes, including glucose-6-phosphate dehydrogenase (LvG6PDH), which increases nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH) production, effectively eliminating excessive ROS. Moreover, WSSV exploits LvNrf2 to establish a positive feedback loop by up-regulating viral immediate early gene wsv051, which further enhances wsv220 expression. Knockdown of LvNrf2 or LvG6PDH reduces WSSV replication and increases host ROS levels. Therefore, WSSV hijacks LvNrf2 pathway to maintain ROS homeostasis and establishes a positive feedback loop to facilitate WSSV replication. These findings reveal a novel molecular mechanism of viral manipulation of host ROS dynamics and suggest potential antiviral strategies targeting LvNrf2 pathway.
Collapse
Affiliation(s)
- Honghui He
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Kai Yuan
- School of Life SciencesHuizhou UniversityHuizhouGuangdong516007China
| | - Junming Pan
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Shaoping Weng
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Chaozheng Li
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| | - Yihong Chen
- Institute of Modern Aquaculture Science and EngineeringCollege of Life SciencesSouth China Normal UniversityGuangzhouGuangdong510631China
| | - Jianguo He
- State Key Laboratory of BiocontrolSchool of Marine SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510275China
- China‐ASEAN Belt and Road Joint Laboratory on Mariculture TechnologySouthern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai)ZhuhaiGuangdong519000China
| |
Collapse
|
8
|
Sun J, Cao X, Li Y, Yu K, Cong Y, Pan Q, Yin Y, Wang J. Oxidative stress in the liver of chicken during fowl adenovirus serotype 4 infection. Poult Sci 2025; 104:105054. [PMID: 40120244 PMCID: PMC11987656 DOI: 10.1016/j.psj.2025.105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
Hepatitis is a significant pathological manifestation of fowl adenovirus serotype-4 (FAdV-4) infection, which is a crucial factor contributing to the mortality of chickens. The pathophysiology of liver disease is rooted in oxidative stress. The present study aims to investigate the presence of oxidative stress during the liver lesion process in FAdV-4 infection. Specifically, one-day-old specific pathogen-free (SPF) chickens were allocated into three groups, the control group, the infection group, and the quercetin group. The quercetin group received daily oral administration of quercetin. At the age of 12 days, the chickens belonging to both the infection and quercetin groups were subjected to intramuscular injection of FAdV-4 (0.3 mL103TCID50/mL). Samples were collected from each group at 2, 4, and 6 days post-infection (dpi), and sera were collected to measure the levels of ALT and AST. A portion of liver tissue was fixed to examine the histological changes, cell apoptosis, and mitochondrial morphology, while another portion was homogenized and mitochondria were isolated. The levels of MDA, SOD, H2O2, and GSH-Px in the homogenate supernatants of livers and isolated mitochondria were measured, and the viral load in the liver was studied. And Cyt C levels in the mitochondria and cytosolic supernatant were recorded. The results showed that AST and ALT in the serum of chicken in the infection group were significantly higher than those in the control and quercetin group at 6 dpi. Obvious swelling, steatosis, necrosis, and inflammatory cell infiltration were observed in the liver of the infection group. Administered with quercetin can significantly decrease the viral load in the liver at 4 and 6 dpi. H2O2 in the liver, and MDA, H2O2, GSH and SOD levels in mitochondria in the hepatocyte of the infection group were significantly higher than those in the control and quercetin groups. Cyt C in the mitochondria of the hepatocyte of infection and quercetin groups were significantly lower than those in the control group at 2 dpi. Cyt C in the cytoplasm of the liver in chicken in the quercetin group was significantly higher than those in the control and infection groups. It was found that the outer mitochondrial membrane in hepatocytes was fractured in the infection group. The proportion of apoptotic cells in the liver in the infection groups was significantly higher than those in the control and quercetin group at 4 dpi, and that in the control group was significantly lower than in the infection and quercetin group. The results suggested that during liver injury induced by FAdV-4 infection, oxidative damage occurred obviously in the liver and mitochondria, and hepatocyte apoptosis was observed. Quercetin, as an antioxidant, can inhibit virus replication to some extent, and alleviate oxidative damage, liver damage, and the mortality caused by FAdV-4 infection.
Collapse
Affiliation(s)
- Jiayu Sun
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Xu Cao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yufeng Li
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250023, PR China
| | - Kexiang Yu
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250023, PR China
| | - Yanfang Cong
- Qingdao VL and Biotech Inc, Qingdao 266000, PR China
| | - Qing Pan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yanbo Yin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jianlin Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
9
|
Tang J, Gao Y, Fu Y, Han Z, Xu P, Li X, Wang S, Wang X. Astragaloside IV mitigates influenza-induced inflammatory responses by suppressing the Wnt/β-catenin signalling pathway in alveolar macrophages. Vet Res 2025; 56:95. [PMID: 40307893 PMCID: PMC12042467 DOI: 10.1186/s13567-025-01529-5] [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: 01/28/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
Respiratory viruses, including the influenza virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pose significant health threats. As tissue-resident macrophages, alveolar macrophages (AM) are crucial for defending against respiratory viral infection by producing cytokines, engulfing virus-infected cells, and promoting wound healing. However, excessive inflammatory responses can lead to tissue injury. Growing evidence indicates that astragaloside IV (AST IV) regulates innate immune responses. Specifically, AST IV balances the inflammatory response to mitigate tissue damage and promote tissue repair. However, whether AST IV directly targets AM to alleviate lung damage induced by respiratory viral infection remains unclear. Our results demonstrate that AST IV treatment significantly reduces morbidity and mortality in mice during IAV infection. AST IV markedly decreases proinflammatory cytokine levels, mitigates lung injury and promotes lung recovery through enhancing the repair capacity mediated by alveolar type II cells. Mechanistically, AST IV suppresses the Wnt/β-catenin signalling pathway, which is critical for driving inflammatory responses in AM while maintaining mitochondrial fitness. Thus, our findings suggest that AST IV effectively targets AM to alleviate inflammation and lung damage caused by respiratory viral infections, highlighting its potential as a therapeutic agent for managing viral pneumonia.
Collapse
Affiliation(s)
- Jianli Tang
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China.
- Carter Immunology Center, University of Virginia, Charlottesville, VA, USA.
| | - Yu Gao
- College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
| | - Yuchen Fu
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Zhaoqing Han
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Ping Xu
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Xin Li
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China
| | - Shuaiyong Wang
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China.
| | - Xin Wang
- College of Agriculture and Forestry Science, Linyi University, Linyi, 276000, Shandong, China.
| |
Collapse
|
10
|
Tian Y, Pan P, Luo X, Sun Y, Yang X, Gao H, Yang Y. Palmitic acid-induced insulin resistance triggers granulosa cell senescence by disruption of the UPR mt/mitophagy/lysosome axis. Chem Biol Interact 2025; 411:111450. [PMID: 40023272 DOI: 10.1016/j.cbi.2025.111450] [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: 11/08/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Insulin resistance (IR) is the main pathological feature of polycystic ovary syndrome (PCOS), but the adverse impacts of IR on ovary and granulosa cells (GCs) are unknown. Therefore, the role of palmitic acid (PA) induced IR in GCs, and a mitochondrial proteostasis and mitochondrial homeostasis control system, the mitochondrial unfolded protein response (UPRmt)/mitophagy/lysosome axis were investigated to uncover the side effect and the mechanism of IR on GCs. Our results revealed that IR in GC was successfully constructed by 100 μM PA treatment accompanied with cell senescence. In addition, mitochondrial function was impaired by IR-induced GC senescence accompanied by significantly increased reactive oxygen species (ROS) and decreased mitochondrial membrane potential, and mitochondrial proteostasis was impaired by a dysfunctional UPRmt and increased protein aggregation, leading to more unfolded and misfolded proteins accumulating in mitochondria. Mitochondrial homeostasis was maintained by the mitophagy/lysosome degradation system, although mitophagy was significantly increased, lysosomes were damaged; hence, malfunctional mitochondria were not cleared by the mitophagy/lysosome degradation system, more ROS were produced by malfunctional mitochondria. Therefore, accelerated GC senescence was triggered by excessive ROS, and reversed by the mitophagy inhibitor cyclosporin A (CsA) accompanied with reduced IR. Additionally, the mice were administered with PA, and results revealed that the accelerated ovarian aging was caused by PA, which might be attributed to GC senescence. In conclusion, GC senescence was triggered in PA-induced IR by disruption of the UPRmt/mitophagy/lysosome axis, and IR induced GC senescence was reversed by the CsA.
Collapse
Affiliation(s)
- Yuan Tian
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Pengge Pan
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Xiaoqiang Luo
- Department of Clinical Laboratory, Ningxia Women and Children's Hospital, Beijing University Hospital, Yinchuan, Ningxia, PR China
| | - Yaqi Sun
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Xintong Yang
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Hui Gao
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Yanzhou Yang
- General Hospital, Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, PR China; Emergency Department, The First People's Hospital of Yinchuan, The Second Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, PR China.
| |
Collapse
|
11
|
Sun Y, Wang L, Ma K, Shen M, Liu J, Zhang Y, Sun L. Antiviral Activity of 1-Deoxynojirimycin Extracts of Mulberry Leaves Against Porcine Epidemic Diarrhea Virus. Animals (Basel) 2025; 15:1207. [PMID: 40362022 PMCID: PMC12071020 DOI: 10.3390/ani15091207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a highly infectious alphacoronavirus, has resulted in substantial economic losses within the global swine industry. Existing vaccines and therapeutic agents have proven inadequate in effectively preventing and controlling PEDV. Natural compounds offer distinct advantages in antiviral research due to their abundant availability, diverse biological activities, and low toxicity. In this study, the antiviral properties of the naturally occurring alkaloid 1-deoxynojirimycin (DNJ) against PEDV were examined. The CC50 of DNJ was determined to be 912.5 μM through experimental analysis on Vero-E6 cells. DNJ demonstrated an inhibitory effect on PEDV activity, with a 50% inhibitory concentration (IC50) of 57.76 μM. The compound primarily inhibited PEDV proliferation during the viral life cycle stages of attachment and replication. Moreover, DNJ mitigated the production of reactive oxygen species (ROS) and inflammation associated with PEDV infection. Computational docking predictions suggest that the viral non-structural proteins include Nsp12, Nsp14, and Nsp16 may serve as potential targets for DNJ. Consequently, DNJ represents a promising candidate for the development of novel therapeutic agents against PEDV.
Collapse
Affiliation(s)
- Yiwei Sun
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Liyan Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Keke Ma
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Manman Shen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jiying Liu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yujuan Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Liumei Sun
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (Y.S.); (L.W.); (K.M.); (M.S.); (J.L.); (Y.Z.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
12
|
Ricchi F, Caramaschi S, Sala A, Franceschini L, Fabbiani L, Ardizzoni A, Blasi E, Cermelli C. Candida albicans as a Trailblazer for Herpes Simplex Virus-2 Infection Against an In Vitro Reconstituted Human Vaginal Epithelium. Microorganisms 2025; 13:905. [PMID: 40284741 PMCID: PMC12029243 DOI: 10.3390/microorganisms13040905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
Little is known about the complex events driving host-pathogen and pathogen-pathogen interplay in polymicrobial infections. Using an in vitro model of a reconstituted vaginal epithelium (RVE) employing the A-431 cell line supplemented with synthetic vaginal fluid (SVF), we studied the consequences of single versus dual infections with Candida albicans and/or Herpes Simplex Virus-2 (HSV-2). Our data show (a) a relevant, SVF-enhanced expression of the differentiation marker cytokeratin 5/6 in the RVE; (b) the ability of Candida albicans to enhance HSV-2 in the dual infection model, with the virus titer almost doubling in the presence of SVF; (c) RVE damage (>20%), mostly attributable to Candida albicans and related to oxidative stress whether SVF is present; (d) the dysregulation of mucin-1, the production of which is enhanced (from 13 to 21 ng/mL) or impaired (from 21 to 10 ng/mL) in response to either SVF or infection, respectively; and (e) a partial-to-negligible cytokine response from the RVE, depending upon SVF presence. In conclusion, using an in vitro RVE model upgraded through the addition of synthetic vaginal fluid, we provide details on epithelial cell-pathogen-pathogen interaction, contributing to a better comprehension of the pathogenesis of polymicrobial infections at a mucosal level.
Collapse
Affiliation(s)
- Francesco Ricchi
- Clinical and Experimental Medicine PhD Program, Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.R.); (S.C.)
| | - Stefania Caramaschi
- Clinical and Experimental Medicine PhD Program, Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.R.); (S.C.)
- Pathology Unit, Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Arianna Sala
- Molecular Microbiology and Virology Unit, University Hospital Policlinico, 41124 Modena, Italy;
| | - Laura Franceschini
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Luca Fabbiani
- Pathology Unit, Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Andrea Ardizzoni
- Department of Surgery, Dentistry, Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.B.)
| | - Elisabetta Blasi
- Department of Surgery, Dentistry, Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.B.)
| | - Claudio Cermelli
- Department of Surgery, Dentistry, Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy; (A.A.); (E.B.)
| |
Collapse
|
13
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Effects of oxidative stress on viral infections: an overview. NPJ VIRUSES 2025; 3:27. [PMID: 40295852 PMCID: PMC11993764 DOI: 10.1038/s44298-025-00110-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025]
Abstract
Viral infections can trigger increased reactive oxygen species (ROS) production and a reduced antioxidant response in the host, leading to redox stress, inflammation, apoptosis, and ultimately, cell and tissue damage, which contribute to disease development. A better understanding of how ROS contributes to viral pathogenesis is critical for the development of novel therapeutic interventions. In this review, we discuss the current knowledge on ROS production and its effects across various viral infections, including severe acute respiratory syndrome-coronavirus-2, influenza A virus, dengue virus, Zika virus, hepatitis B virus, hepatitis C virus, and human immunodeficiency virus infections, to improve future therapeutic and preventive strategies for these infections.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, 8210, Bangladesh.
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, 890-0065, Japan.
| |
Collapse
|
14
|
Zhao X, Dai X, Wang F, Li C, Song X, Han Y, Zhang C, Wang L, He Z, Zhang R, Ye L. Artemisia annua L. leaf extracts suppress influenza virus infection by targeting the viral nucleoprotein and blocking mitochondria-mediated apoptosis. Virol Sin 2025; 40:247-259. [PMID: 40043849 PMCID: PMC12131016 DOI: 10.1016/j.virs.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/26/2025] [Indexed: 04/01/2025] Open
Abstract
Artemisia annua L. is a medicinal herb with multiple therapeutic applications, whereas its anti-influenza A virus (IAV) efficiency and mechanism of action are still unclear. Here, we investigated the inhibition activity and mechanism of A. annua leaf methanol extracts (AALME) against IAV in vitro and in vivo. Our results revealed that AALME exhibits potent anti-IAV activity by interacting with IAV particles. Mechanistically, AALME directly targets the IAV nucleoprotein (NP) protein and abolishes the nuclear import of IAV NP. AALME profoundly suppresses IAV-induced mitochondrial apoptosis via suppressing ROS-mediated AIF-dependent pathways. More importantly, we found that AALME plays a crucial role in protecting mice from IAV infection and mitigating IAV pathogenicity. This current work provides mechanistic insight into the mechanism by which AALME controls IAV infection in vitro and in vivo, potentially contributing to the development of antiviral treatments for IAV infection.
Collapse
Affiliation(s)
- Xiwen Zhao
- College of Chinese Medicine, School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China; Department of Immunology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xuan Dai
- Department of Immunology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518055, China
| | - Fuyi Wang
- Department of Immunology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518055, China; College of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Chenyang Li
- College of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xun Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Yingying Han
- College of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Chaowei Zhang
- College of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Lu Wang
- Hyperbaric Oxygen Department, Shenzhen University General Hospital, Shenzhen 518055, China.
| | - Zhendan He
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Rongping Zhang
- College of Chinese Medicine, School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Liang Ye
- Department of Immunology, International Cancer Center, Shenzhen University Medical School, Shenzhen 518055, China.
| |
Collapse
|
15
|
de Almeida ALT, da Costa IPS, Garcia MDDN, da Silva MAN, Lazzaro YG, de Filippis AMB, Nogueira FDB, Barreto-Vieira DF. Oropouche Virus: Isolation and Ultrastructural Characterization from a Human Case Sample from Rio de Janeiro, Brazil, Using an In Vitro System. Viruses 2025; 17:373. [PMID: 40143301 PMCID: PMC11946457 DOI: 10.3390/v17030373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
The Oropouche virus (OROV) is a segmented negative-sense RNA arbovirus member of the Peribunyaviridae family, associated with recurring epidemics of Oropouche fever in Central and South America. Since its identification in 1955, OROV has been responsible for outbreaks in both rural and urban areas, with transmission involving sylvatic and urban cycles. This study focuses on the characterization of an OROV isolate from a human clinical sample collected in the state of Rio de Janeiro, a non-endemic region in Brazil, highlighting ultrastructural and morphological aspects of the viral replicative cycle in Vero cells. OROV was isolated in Vero cell monolayers which, following viral inoculation, exhibited marked cytopathic effects (CPEs), mainly represented by changes in cell morphology, including membrane protrusions and vacuolization, as well as cell death. Studies by transmission electron microscopy (TEM) revealed significant ultrastructural changes, such as apoptosis, intense remodeling of membrane-bound organelles and signs of rough endoplasmic reticulum and mitochondrial stress. Additionally, the formation of specialized cytoplasmic vacuoles and intra- and extracellular vesicles emphasized trafficking and intercellular communication as essential mechanisms in OROV infection. RT-qPCR studies confirmed the production of viral progeny in high titers, corroborating the efficiency of this experimental model. These findings contribute to a better understanding of the cytopathogenic mechanisms of OROV infection and the contribution of cellular alterations in OROV morphogenesis.
Collapse
Affiliation(s)
- Ana Luisa Teixeira de Almeida
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| | - Igor Pinto Silva da Costa
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| | - Maycon Douglas do Nascimento Garcia
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| | - Marcos Alexandre Nunes da Silva
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| | - Yasmim Gonçalves Lazzaro
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| | - Ana Maria Bispo de Filippis
- Laboratory of Arboviruses and Hemorrhagic Viruses, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (A.M.B.d.F.); (F.d.B.N.)
| | - Fernanda de Bruycker Nogueira
- Laboratory of Arboviruses and Hemorrhagic Viruses, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (A.M.B.d.F.); (F.d.B.N.)
| | - Debora Ferreira Barreto-Vieira
- Laboratory of Viral Morphology and Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation—Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.D.d.N.G.); (M.A.N.d.S.); (Y.G.L.)
| |
Collapse
|
16
|
Rosales JJ, Brunner MB, Rodríguez M, Marin M, Maldonado EN, Pérez S. Reactive oxygen species favors Varicellovirus bovinealpha 5 (BoAHV-5) replication in neural cells. Mitochondrion 2025; 81:102005. [PMID: 39778729 DOI: 10.1016/j.mito.2025.102005] [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: 10/09/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Varicellovirus bovinealpha (BoAHV) 1 and 5 are closely related neurotropic alphaherpesviruses with distinct neuropathogenic potential. BoAHV-5 causes meningoencephalitis in calves whereas encephalitis by BoAHV-1 infection is sporadic. the mechanisms underlying the differences in tropism and clinical outcomes of the infections are not yet completely understood. Here, we used neuroblastoma SH-SY5Y cells as non-differentiated in comparison with the SH-SY5Y neuronal-like cells obtained after exposing SH-SY5Y undifferentiated cells to trans-retinoic acid. We aimed to establish whether there was a relationship between the production of reactive oxygen species (ROS) and the kinetics of virus replication. We demonstrated that ROS production after BoAHV infection was higher in differentiated cells. Generation of ROS was also dependent on the infecting BoAHV strain. Higher ROS levels were produced during BoAHV-5 infection concomitantly with enhanced viral replication. We propose that increased ROS production mechanistically contributes to the tissue damage and neuroinflammation induced by BoAHV-5 infection. Future studies will determine specific targets of ROS that mediate the effects on viral replication.
Collapse
Affiliation(s)
- Juan José Rosales
- Laboratorio de Virología, Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil, Buenos Aires, Argentina; Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias, Campus Universitario, Tandil, Buenos Aires, Argentina
| | - María Belén Brunner
- Laboratorio de Virología, Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil, Buenos Aires, Argentina; Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias, Campus Universitario, Tandil, Buenos Aires, Argentina
| | - Marcelo Rodríguez
- Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias, Campus Universitario, Tandil, Buenos Aires, Argentina
| | - Maia Marin
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Eduardo Néstor Maldonado
- Department of Drug Discovery & Pharmaceutical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Sandra Pérez
- Laboratorio de Virología, Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil, Buenos Aires, Argentina; Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), Facultad de Ciencias Veterinarias, Campus Universitario, Tandil, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Cao Z, Yang Y, Zhang S, Zhang T, Lü P, Chen K. Liquid-liquid phase separation in viral infection: From the occurrence and function to treatment potentials. Colloids Surf B Biointerfaces 2025; 246:114385. [PMID: 39561518 DOI: 10.1016/j.colsurfb.2024.114385] [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: 10/02/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Liquid-liquid phase separation (LLPS) of biomacromolecules, as a widespread cellular functional mechanism, is closely related to life processes, and is also commonly present in the lifecycle of viruses. Viral infection often leads to the recombination and redistribution of intracellular components to form biomacromolecule condensates assembled from viral replication-related proteins and intracellular components, which plays an important role in the process of viral infection. In this review, the key and influencing factors of LLPS are generalized, which mainly depend on various molecular interactions and environmental conditions in solution. Meanwhile, some examples of viruses utilizing LLPS are summarized, which are conducive to further understanding the subtle and complex biological regulatory processes between phase condensation and viruses. Finally, some representative antiviral drugs targeting phase separation that have been discovered are also outlined. In conclusion, in-depth study of the role of LLPS in viral infection is helpful to understand the mechanisms of virus-related diseases from a new perspective, and also provide a new therapeutic strategy for future treatments.
Collapse
Affiliation(s)
- Zhaoxiao Cao
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| | - Simeng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Tiancheng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
18
|
Li Y, Wu J, You Y, Miao M, Yu W. Overexpression of Acetylation-Defective Heat Shock Protein 60 Inhibits the Proliferation of Nucleopolyhedrovirus in Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70038. [PMID: 39948826 DOI: 10.1002/arch.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 05/09/2025]
Abstract
Heat shock protein 60 (Hsp60), abundantly presents in mitochondria, is a highly conserved chaperone that maintains the stability and functionality of mitochondrial proteins, while also participating in the regulation of various cellular processes. As a member of the heat shock family, Hsp60 significantly influences viral proliferation. However, limited research is available on its role in the proliferation of entomopathogenic baculoviruses, particularly Bombyx mori nucleopolyhedrovirus (BmNPV). Our previous proteomics results showed a significant decrease of Hsp60 acetylation levels after BmNPV infection. To investigate the impact of Hsp60 deacetylation on viral proliferation, site-direct mutagenesis was performed to generate a deacetylated (K/R) mimic of Hsp60. We found that the acetylation level of lysine 362 (K362) decreased after BmNPV challenge. Furthermore, overexpression of deacetylation-mimicking Hsp60 reduced the chaperone activity of Hsp60, leading to impaired mitochondrial function, including increased reactive oxygen species (ROS) levels, decreased mitochondrial membrane potential, and reduced substrate protein Manganese-containing superoxide dismutase (Mn-SOD) activities, ultimately leading to inhibition of viral proliferation. This study establishes lysine 362 acetylation of Hsp60 as a model for Posttranslational modifications induced by host-virus interactions, providing new insights into potential antiviral strategies.
Collapse
Affiliation(s)
- Yao Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory, Silkworm Bioreactor and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Jiannan Wu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory, Silkworm Bioreactor and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Yi You
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory, Silkworm Bioreactor and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Meng Miao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory, Silkworm Bioreactor and Biomedicine, Hangzhou, Zhejiang Province, China
| | - Wei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory, Silkworm Bioreactor and Biomedicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
19
|
Yu H, Wu C, He J, Zhang Y, Cao Q, Lan H, Li H, Xu C, Chen C, Li R, Zheng B. Metabolic reprogramming induced by PSMA4 overexpression facilitates bortezomib resistance in multiple myeloma. Ann Hematol 2025; 104:1023-1037. [PMID: 39755751 PMCID: PMC11971155 DOI: 10.1007/s00277-024-06163-3] [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: 09/18/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
Multiple myeloma(MM) remains incurable with high relapse and chemoresistance rates. Differentially expressed genes(DEGs) between newly diagnosed myeloma and secondary plasma cell leukemia(sPCL) were subjected to a weighted gene co-expression network analysis(WGCNA). Drug resistant myeloma cell lines were established. Seahorse XF analyzer was applied to detect the metabolism reprogramming associated with the hub gene. The metabolic relevance and the underlying mechanism of the hub gene in myeloma resistance were explored via in vitro experiments. A total of 1310 DEGs were used to construct five co-expression modules. Gene function enrichment analysis demonstrated that candidate hub genes were closely related to oxidative phosphorylation. We performed prognostic analysis and identified PSMA4 as the key hub gene related to the extramedullary invasion of myeloma. The in vitro experiments demonstrated bortezomib resistant myeloma cell lines exhibited high PSMA4 expression, improved oxidative phosphorylation activity with increased ROS level. PSMA4 knockdown re-sensitize resistant myeloma cells via suppressing oxidative phosphorylation activity. Further investigation revealed that PSMA4 induced a hypoxia state which activated the HIF-1α signaling pathway. PSMA4 induces metabolic reprogramming by improving oxidative phosphorylation activity which accounts for the hypoxia state in myeloma cell. The activated HIF-1α signaling pathway causes bortezomib resistance via promoting anti-apoptotic activity in myeloma.
Collapse
Affiliation(s)
- Han Yu
- Department of Oncology, Navy No.905 Hospital of PLA, Naval Medical University, Shanghai, 200052, China
| | - Chengli Wu
- Department of Oncology, Navy No.905 Hospital of PLA, Naval Medical University, Shanghai, 200052, China
| | - Jie He
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China
| | - Yajun Zhang
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China
| | - Qiqi Cao
- Department of Oncology, 971 Hospital of PLA Navy, Qingdao, 266071, China
| | - Hongyan Lan
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China
| | - Hongshan Li
- Department of Oncology, Navy No.905 Hospital of PLA, Naval Medical University, Shanghai, 200052, China
| | - Chengyang Xu
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China
| | - Chen Chen
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China
| | - Rong Li
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China.
| | - Bo Zheng
- Department of Hematology, Navy Medical Center of PLA, Naval Medical University, No. 338 West Huaihai Road, Changning District, Shanghai, 200052, China.
| |
Collapse
|
20
|
Hashemi Sheikhshabani S, Ghafouri-Fard S, Amini-Farsani Z, Modarres P, Khazaei Feyzabad S, Amini-Farsani Z, Shaygan N, Omrani MD. In Silico Prediction of Functional SNPs Interrupting Antioxidant Defense Genes in Relation to COVID-19 Progression. Biochem Genet 2025; 63:499-525. [PMID: 38460087 DOI: 10.1007/s10528-024-10705-9] [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: 09/28/2023] [Accepted: 01/16/2024] [Indexed: 03/11/2024]
Abstract
The excessive production of reactive oxygen species and weakening of antioxidant defense system play a pivotal role in the pathogenesis of different diseases. Extensive differences observed among individuals in terms of affliction with cancer, cardiovascular disorders, diabetes, bacterial, and viral infections, as well as response to treatments can be partly due to their genomic variations. In this work, we attempted to predict the effect of SNPs of the key genes of antioxidant defense system on their structure, function, and expression in relation to COVID-19 pathogenesis using in silico tools. In addition, the effect of SNPs on the target site binding efficiency of SNPs was investigated as a factor with potential to change drug response or susceptibility to COVID-19. According to the predicted results, only six missense SNPs with minor allele frequency (MAF) ≥ 0.1 in the coding region of genes GPX7, GPX8, TXNRD2, GLRX5, and GLRX were able to strongly affect their structure and function. Our results predicted that 39 SNPs with MAF ≥ 0.1 led to the generation or destruction of miRNA-binding sites on target antioxidant genes from GPX, PRDX, GLRX, TXN, and SOD families. The results obtained from comparing the expression profiles of mild vs. severe COVID-19 patients using GEO2R demonstrated a significant change in the expression of approximately 250 miRNAs. The binding efficiency of 21 of these miRNAs was changed due to the elimination or generation of target sites in these genes. Altogether, this study reveals the fundamental role of the SNPs of antioxidant defense genes in COVID-19 progression and susceptibility of individuals to this virus. In addition, different responses of COVID-19 patients to antioxidant defense system enhancement drugs may be due to presence of these SNPs in different individuals.
Collapse
Affiliation(s)
- Somayeh Hashemi Sheikhshabani
- Student Research Committee, Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Student Research Committee, Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Amini-Farsani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parastoo Modarres
- Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| | - Sharareh Khazaei Feyzabad
- Department of Laboratory Sciences, School of Paramedical Sciences, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Zahra Amini-Farsani
- Bayesian Imaging and Spatial Statistics Group, Institute of Statistics, Ludwig-Maximilian-Universität München, Ludwigstraße 33, 80539, Munich, Germany
| | - Nasibeh Shaygan
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Saller BS, Wöhrle S, Fischer L, Dufossez C, Ingerl IL, Kessler S, Mateo-Tortola M, Gorka O, Lange F, Cheng Y, Neuwirt E, Marada A, Koentges C, Urban C, Aktories P, Reuther P, Giese S, Kirschnek S, Mayer C, Pilic J, Falquez-Medina H, Oelgeklaus A, Deepagan VG, Shojaee F, Zimmermann JA, Weber D, Tai YH, Crois A, Ciminski K, Peyronnet R, Brandenburg KS, Wu G, Baumeister R, Heimbucher T, Rizzi M, Riedel D, Helmstädter M, Buescher J, Neumann K, Misgeld T, Kerschensteiner M, Walentek P, Kreutz C, Maurer U, Rambold AS, Vince JE, Edlich F, Malli R, Häcker G, Kierdorf K, Meisinger C, Köttgen A, Jakobs S, Weber ANR, Schwemmle M, Groß CJ, Groß O. Acute suppression of mitochondrial ATP production prevents apoptosis and provides an essential signal for NLRP3 inflammasome activation. Immunity 2025; 58:90-107.e11. [PMID: 39571574 DOI: 10.1016/j.immuni.2024.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/15/2024] [Accepted: 10/24/2024] [Indexed: 01/18/2025]
Abstract
How mitochondria reconcile roles in functionally divergent cell death pathways of apoptosis and NLRP3 inflammasome-mediated pyroptosis remains elusive, as is their precise role in NLRP3 activation and the evolutionarily conserved physiological function of NLRP3. Here, we have shown that when cells were challenged simultaneously, apoptosis was inhibited and NLRP3 activation prevailed. Apoptosis inhibition by structurally diverse NLRP3 activators, including nigericin, imiquimod, extracellular ATP, particles, and viruses, was not a consequence of inflammasome activation but rather of their effects on mitochondria. NLRP3 activators turned out as oxidative phosphorylation (OXPHOS) inhibitors, which we found to disrupt mitochondrial cristae architecture, leading to trapping of cytochrome c. Although this effect was alone not sufficient for NLRP3 activation, OXPHOS inhibitors became triggers of NLRP3 when combined with resiquimod or Yoda-1, suggesting that NLRP3 activation requires two simultaneous cellular signals, one of mitochondrial origin. Therefore, OXPHOS and apoptosis inhibition by NLRP3 activators provide stringency in cell death decisions.
Collapse
Affiliation(s)
- Benedikt S Saller
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Svenja Wöhrle
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Larissa Fischer
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Clara Dufossez
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Isabella L Ingerl
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kessler
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Maria Mateo-Tortola
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Felix Lange
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Emilia Neuwirt
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Adinarayana Marada
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Koentges
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Chiara Urban
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp Aktories
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Peter Reuther
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sebastian Giese
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Kirschnek
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Carolin Mayer
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Johannes Pilic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Hugo Falquez-Medina
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Aline Oelgeklaus
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Veerasikku Gopal Deepagan
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Farzaneh Shojaee
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Julia A Zimmermann
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Damian Weber
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Yi-Heng Tai
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Anna Crois
- Faculty of Biology, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kevin Ciminski
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Remi Peyronnet
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center Freiburg - Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Katharina S Brandenburg
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Gang Wu
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ralf Baumeister
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Heimbucher
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Department of Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Riedel
- Laboratory for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Martin Helmstädter
- EMcore, Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Joerg Buescher
- Metabolomics and FACS Core Facilities, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Konstantin Neumann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians Universität München, Munich, Germany; Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians Universität München, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Peter Walentek
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Internal Medicine IV, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Medical Biometry and Statistics, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ulrich Maurer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute for Molecular Medicine and Cell Research, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angelika S Rambold
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, The Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Frank Edlich
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Veterinary Physiological Chemical Institute, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Roland Malli
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Chris Meisinger
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany; Institute of Genetic Epidemiology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Stefan Jakobs
- Research Group Mitochondrial Structure and Dynamics, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany; Clinic for Neurology, University Medical Center of Göttingen, Göttingen, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Translational Neuroinflammation and Automated Microscopy TNM, Göttingen, Germany
| | - Alexander N R Weber
- Department of Innate Immunity, Institute of Immunology, University of Tübingen, Tübingen, Germany; Clusters of Excellence EXC-2180 (iFIT) and -2124 (CMFI), University of Tübingen, Tübingen, Germany
| | - Martin Schwemmle
- Institute of Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Christina J Groß
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
22
|
Zheng H, Yang X, Zhong H, Song C, Wu Z, Yang H. HDAC6 Facilitates PRV and VSV Infection by Inhibiting Type I Interferon Production. Viruses 2025; 17:90. [PMID: 39861880 PMCID: PMC11768819 DOI: 10.3390/v17010090] [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: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
HDAC6 modulates viral infection through diverse mechanisms. Here, we investigated the role of HDAC6 in influencing viral infection in pig cells with the aim of exploiting the potential antiviral gene targets in pigs. Using gene knockout and overexpression strategies, we found that HDAC6 knockout greatly reduced PRV and VSV infectivity, whereas HDAC6 overexpression increased their infectivity in PK15 cells. Mechanistic studies identified HDAC6 as a DNA damage inhibitor in PK15 cells. HDAC6 overexpression attenuated DNA damage levels, which can further reduce type I IFN production to promote viral infection. Conversely, HDAC6 deficiency can limit viral infection by increasing DNA damage-mediated type I IFN production. This work demonstrates that HDAC6 affects the infection process of multiple viruses by modulating type I IFN production, highlighting a regulatory role of HDAC6 linking host immune response and viral infection levels in pig cells.
Collapse
Affiliation(s)
- Hu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Xiaohui Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Haiwen Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Changxu Song
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| | - Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| |
Collapse
|
23
|
Kwon EB, Kim B, Kim YE, Na SJ, Han SM, Woo SO, Choi HM, Moon S, Kim YS, Choi JG. Hovenia dulcis Thunb. Honey Exerts Antiviral Effect Against Influenza A Virus Infection Through Mitochondrial Stress-Mediated Enhancement of Innate Immunity. Antioxidants (Basel) 2025; 14:71. [PMID: 39857405 PMCID: PMC11761272 DOI: 10.3390/antiox14010071] [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: 11/05/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
To combat influenza A virus (IAV) infection, it is vital to develop effective therapeutic strategies, including immunomodulators. In this study, we examined the antiviral effects of Hovenia dulcis Thunb. honey (HDH) against IAV using RAW 264.7 cells. HDH treatment significantly reduced IAV infection and viral protein expression. Moreover, it enhanced the production of interferon (IFN)-β, activated the innate immune response through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, and upregulated IFN signaling through signal transducer and activator of transcription (STAT)1/2 phosphorylation and interferon-stimulated gene (ISG) expression. In addition, HDH decreased IAV-induced intracellular and mitochondrial reactive oxygen species (ROS) production by upregulating the expression of antioxidant proteins, such as Sirt3 and SOD2. The results suggest that HDH is a potential therapeutic agent inhibiting viral replication and boosting host antiviral immunity.
Collapse
Affiliation(s)
- Eun-Bin Kwon
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-B.K.); (B.K.); (Y.-E.K.)
| | - Buyun Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-B.K.); (B.K.); (Y.-E.K.)
| | - Young-Eun Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-B.K.); (B.K.); (Y.-E.K.)
| | - Sung-Joon Na
- Special Forest Resources Division, National Institute of Forest Science, Suwon 16631, Republic of Korea;
| | - Sang Mi Han
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, Wanju 566-851, Republic of Korea; (S.M.H.); (S.O.W.); (H.M.C.); (S.M.)
| | - Soon Ok Woo
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, Wanju 566-851, Republic of Korea; (S.M.H.); (S.O.W.); (H.M.C.); (S.M.)
| | - Hong Min Choi
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, Wanju 566-851, Republic of Korea; (S.M.H.); (S.O.W.); (H.M.C.); (S.M.)
| | - Siwon Moon
- Department of Agricultural Biology, National Academy of Agricultural Science, Rural Development Administration, Wanju 566-851, Republic of Korea; (S.M.H.); (S.O.W.); (H.M.C.); (S.M.)
| | - Young Soo Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-B.K.); (B.K.); (Y.-E.K.)
| | - Jang-Gi Choi
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-B.K.); (B.K.); (Y.-E.K.)
| |
Collapse
|
24
|
Zandi M, Mousavi FS, Hashemnia SMR. Mpox-Induced Metabolic Alterations. J Cell Mol Med 2025; 29:e70341. [PMID: 39779460 PMCID: PMC11710930 DOI: 10.1111/jcmm.70341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
The resurgence of mpox as a global health threat highlights the need to understand its interaction with host cell metabolism. Unlike other well-studied viruses, research on mpox is limited, particularly regarding its impact on cellular processes. In this article, we explore how mpox might manipulate metabolic pathways-such as glycolysis, lipid synthesis and mitochondrial dynamics-to enhance its replication and evade immune responses. By drawing parallels with related poxviruses, we underscore the potential for targeting these metabolic shifts as novel therapeutic strategies. Understanding these interactions is crucial for developing effective treatments against mpox.
Collapse
Affiliation(s)
- Milad Zandi
- Iranian Society for VirologyDeputy for Education, Ministry of Health and Medical EducationTehranIran
| | - Fatemeh Sadat Mousavi
- Department of Microbiology and Immunology, Faculty of Veterinary MedicineUniversity of TehranTehranIran
| | | |
Collapse
|
25
|
Chang J, Yao Y, Sun X, Wang W, Qian H, Liu Y, Xue C, Ye W, Jiang F. JAG1 mediates apoptosis in herpes simplex keratitis by suppressing autophagy via ROS/JAG1/NOTCH1/pULK1 signaling pathway. Cell Biol Toxicol 2024; 41:1. [PMID: 39704867 PMCID: PMC11662045 DOI: 10.1007/s10565-024-09968-0] [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: 09/03/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Herpes simplex keratitis (HSK), an ocular disease resulted from herpes simplex virus type 1 (HSV-1) infection, leads to the majority of infectious corneal blindness worldwide. The apoptosis of corneal epithelial cells (CECs) resulted from HSV-1 disrupts the epithelial barrier and exacerbates the infection; however, there is no definitive cure for HSK. Jagged1 (JAG1), one of the primary functional ligands for NOTCH receptors, plays a crucial role in regulating apoptosis and autophagy; however, its role in HSK is unclear. Our transcriptome analysis showed JAG1 was significantly upregulated in HSV-1-infected human CECs. We aimed to explore JAG1's role in regulating apoptosis in HSV-1-infected human CECs and in HSK mice. HSV-1 infection induced apoptosis and reactive oxygen species (ROS) generation in CECs. HSV-1 also activated the JAG1/NOTCH1 signaling pathway. The ROS scavenger N-acetylcysteine significantly mitigated these effects. Additionally, inhibiting the JAG1/NOTCH1 pathway with short hairpin RNA against JAG1 or a NOTCH1 inhibitor (N-[N-{3,5-difuorophenacetyl}-1-alanyl]-S-phenylglycine t-butyl ester [DAPT]) alleviated HSV-1-induced CEC apoptosis. Transmission electron microscopy and western blotting revealed that HSV-1 infection suppressed ULK1-mediated autophagy in CECs, while DAPT treatment enhanced autophagy by suppressing ULK1 phosphorylation. The activation of autophagy by rapamycin treatment markedly reduced ROS levels and apoptosis in HSV-1-infected CECs, revealing a synergistic effect between the suppressed autophagy and increased ROS levels, ultimately leading to apoptosis. Thus, HSV-1 induces CEC apoptosis by suppressing autophagy through ROS/JAG1/NOTCH1/pULK1 signaling pathway in vitro and in vivo, providing potential therapeutic targets for HSK.
Collapse
Affiliation(s)
- Jingyao Chang
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yao Yao
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Xinghong Sun
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China
| | - Wenzhe Wang
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Haochen Qian
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Yumeilan Liu
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China
| | - Chunyan Xue
- School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Ophthalmology, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, Hainan, China.
| | - Wei Ye
- Department of Ophthalmology, Jinling Hospital, Nanjing, 210002, Jiangsu, China.
| | - Feng Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
26
|
Yu J, An N, Zhu J, Zhu B, Zhang G, Chen K, Zhou Y, Ye T, Li G. AVL-armed oncolytic vaccinia virus promotes viral replication and boosts antitumor immunity via increasing ROS levels in pancreatic cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200878. [PMID: 39431173 PMCID: PMC11488421 DOI: 10.1016/j.omton.2024.200878] [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: 05/03/2024] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024]
Abstract
Pancreatic malignant neoplasm is an extremely deadly malignancy well known for its resistance to traditional therapeutic approaches. Enhanced treatments are imperative for individuals diagnosed with pancreatic cancer (PC). Recent investigations have shed light on the wide-ranging anticancer properties of genetic therapy facilitated by oncolytic vaccinia virus. To illuminate the precise impacts of Aphrocallistes vastus lectin-armed oncolytic vaccinia virus (oncoVV-AVL) on PC, AsPC-1 and PANC-1 cells underwent treatment with oncoVV-AVL. Our findings revealed that oncoVV-AVL possesses the capacity to heighten oncolytic effects on PC cells and incite the production of diverse cytokines like tumor necrosis factor-α, interleukin-6 (IL-6), IL-8, and interferon-I (IFN-I), without triggering antiviral responses. Additionally, oncoVV-AVL can significantly elevate the levels of ROS in PC cells, initiating an oxidative stress response that promotes viral replication, apoptosis, and autophagy. Moreover, in xenograft tumor models, oncoVV-AVL notably restrained PC growth, enhanced IFN-γ levels in the bloodstream, and reprogrammed macrophages. Our investigation indicates that oncoVV-AVL boosts the efficacy of antitumor actions against PC tumors by orchestrating reactive oxygen species-triggered viral replication, fostering M1 polarization, and reshaping the tumor microenvironment to transform cold PC tumors into hot ones. These findings imply that oncoVV-AVL could present a novel therapeutic approach for treating PC tumors.
Collapse
Affiliation(s)
- Jianlei Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Nan An
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jili Zhu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Borong Zhu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Guohui Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Kan Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yanrong Zhou
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Ting Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Gongchu Li
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| |
Collapse
|
27
|
Tworig J, Grafton F, Fisher K, Hörer M, Reid CA, Mandegar MA. Transcriptomics-informed pharmacology identifies epigenetic and cell cycle regulators that enhance AAV production. Mol Ther Methods Clin Dev 2024; 32:101384. [PMID: 39687728 PMCID: PMC11647610 DOI: 10.1016/j.omtm.2024.101384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024]
Abstract
Recombinant adeno-associated virus (rAAV) is a widely used viral vector for gene therapy. However, these vectors have limited availability due to manufacturing challenges with productivity and quality. These challenges can be addressed by better understanding the mechanisms that influence cellular responses during rAAV production. In this study, we aimed to identify targets that may enhance rAAV production using transcriptomic analyses of five cell lines with variable capacities for rAAV production. Using an intersectional approach, we measured the transcriptional responses of these cells during rAAV production and compared transcriptional profiles between high and base producers to identify possible targets for enhancing production. During rAAV production, we found transcriptional differences in cell cycle and nucleosome components contributed to proliferative capacity and DNA replication. We also saw upregulation of several core functions, including transcription, stress response, and Golgi and endoplasmic reticulum organization. Conversely, we saw consistent downregulation of other factors, including inhibitors of DNA-binding proteins and mitochondrial components. With a drug-connectivity analysis, we identified five classes of drugs that were predicted to enhance rAAV production. We also validated the efficacy of histone deacetylase and microtubule inhibitors. Our data uncover novel and previously identified pathways that may enhance rAAV production and quality to expand availability of rAAV for gene therapies.
Collapse
Affiliation(s)
- Joshua Tworig
- Ascend Advanced Therapies CA, Inc, Alameda, CA 94501, USA
| | | | - Kaylin Fisher
- Ascend Advanced Therapies CA, Inc, Alameda, CA 94501, USA
| | - Markus Hörer
- Ascend Advanced Therapies GmbH, 82152 Planegg, Germany
| | | | | |
Collapse
|
28
|
Wang J, Wang D. Mitophagy in gynecological malignancies: roles, advances, and therapeutic potential. Cell Death Discov 2024; 10:488. [PMID: 39639053 PMCID: PMC11621523 DOI: 10.1038/s41420-024-02259-x] [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: 10/10/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Mitophagy is a process in which impaired or dysfunctional mitochondria are selectively eliminated through the autophagy mechanism to maintain mitochondrial quality control and cellular homeostasis. Based on specific target signals, several mitophagy processes have been identified. Defects in mitophagy are associated with various pathological conditions, including neurodegenerative disorders, cardiovascular diseases, metabolic diseases, and cancer. Mitophagy has been shown to play a critical role in the pathogenesis of gynecological malignancies and the development of drug resistance. In this review, we have summarized and discussed the role and recent advances in understanding the therapeutic potential of mitophagy in the development of gynecological malignancies. Therefore, the valuable insights provided in this review may serve as a basis for further studies that contribute to the development of novel treatment strategies and improved patient outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Dandan Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
29
|
Papp H, Tóth E, Bóvári-Biri J, Bánfai K, Juhász P, Mahdi M, Russo LC, Bajusz D, Sipos A, Petri L, Szalai TV, Kemény Á, Madai M, Kuczmog A, Batta G, Mózner O, Vaskó D, Hirsch E, Bohus P, Méhes G, Tőzsér J, Curtin NJ, Helyes Z, Tóth A, Hoch NC, Jakab F, Keserű GM, Pongrácz JE, Bai P. The PARP inhibitor rucaparib blocks SARS-CoV-2 virus binding to cells and the immune reaction in models of COVID-19. Br J Pharmacol 2024; 181:4782-4803. [PMID: 39191429 DOI: 10.1111/bph.17305] [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: 09/15/2023] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND AND PURPOSE To date, there are limited options for severe Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2 virus. As ADP-ribosylation events are involved in regulating the life cycle of coronaviruses and the inflammatory reactions of the host; we have, here, assessed the repurposing of registered PARP inhibitors for the treatment of COVID-19. EXPERIMENTAL APPROACH The effects of PARP inhibitors on virus uptake were assessed in cell-based experiments using multiple variants of SARS-CoV-2. The binding of rucaparib to spike protein was tested by molecular modelling and microcalorimetry. The anti-inflammatory properties of rucaparib were demonstrated in cell-based models upon challenging with recombinant spike protein or SARS-CoV-2 RNA vaccine. KEY RESULTS We detected high levels of oxidative stress and strong PARylation in all cell types in the lungs of COVID-19 patients, both of which negatively correlated with lymphocytopaenia. Interestingly, rucaparib, unlike other tested PARP inhibitors, reduced the SARS-CoV-2 infection rate through binding to the conserved 493-498 amino acid region located in the spike-ACE2 interface in the spike protein and prevented viruses from binding to ACE2. In addition, the spike protein and viral RNA-induced overexpression of cytokines was down-regulated by the inhibition of PARP1 by rucaparib at pharmacologically relevant concentrations. CONCLUSION AND IMPLICATIONS These results point towards repurposing rucaparib for treating inflammatory responses in COVID-19.
Collapse
Affiliation(s)
- Henrietta Papp
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Emese Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
| | - Judit Bóvári-Biri
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Krisztina Bánfai
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Péter Juhász
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mohamed Mahdi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Lilian Cristina Russo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group, Debrecen, Hungary
| | - László Petri
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
| | - Tibor Viktor Szalai
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Ágnes Kemény
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, Pécs, Hungary
- Department of Medical Biology, Medical School, Pécs, Hungary
| | - Mónika Madai
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Anett Kuczmog
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Gyula Batta
- Department of Organic Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Orsolya Mózner
- Doctoral School of Molecular Medicine, Semmelweis University, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Dorottya Vaskó
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Edit Hirsch
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Tőzsér
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nicola J Curtin
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Centre for Neuroscience, University of Pécs, Pécs, Hungary
- Hungarian Research Network, Chronic Pain Research Group, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Attila Tóth
- Section of Clinical Physiology, Department of Cardiology, University of Debrecen, Debrecen, Hungary
| | - Nicolas C Hoch
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Ferenc Jakab
- National Laboratory of Virology, University of Pécs, Pécs, Hungary
- Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Budapest, Hungary
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Judit E Pongrácz
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Péter Bai
- Szentagothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
30
|
Chooi WH, Winanto, Zeng Y, Lee CYP, Lim ZQ, Gautam P, Chu JJH, Loh YH, Alonso S, Ng SY. Enterovirus-A71 preferentially infects and replicates in human motor neurons, inducing neurodegeneration by ferroptosis. Emerg Microbes Infect 2024; 13:2382235. [PMID: 39017655 DOI: 10.1080/22221751.2024.2382235] [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: 02/19/2024] [Revised: 06/15/2024] [Accepted: 07/16/2024] [Indexed: 07/18/2024]
Abstract
Enterovirus A71 (EV-A71) causes Hand, Foot, and Mouth Disease and has been clinically associated with neurological complications. However, there is a lack of relevant models to elucidate the neuropathology of EV-A71 and its mechanism, as the current models mainly utilize animal models or immortalized cell lines. In this study, we established a human motor neuron model for EV-A71 infection. Single cell transcriptomics of a mixed neuronal population reveal higher viral RNA load in motor neurons, suggesting higher infectivity and replication of EV-A71 in motor neurons. The elevated RNA load in motor neurons correlates with the downregulation of ferritin-encoding genes. Subsequent analysis confirms that neurons infected with EV-A71 undergo ferroptosis, as evidenced by increased levels of labile Fe2+ and peroxidated lipids. Notably, the Fe2+ chelator Deferoxamine improves mitochondrial function and promotes survival of motor neurons by 40% after EV-A71 infection. These findings deepen understanding of the molecular pathogenesis of EV-A71 infection, providing insights which suggest that improving mitochondrial respiration and inhibition of ferroptosis can mitigate the impact of EV-A71 infection in the central nervous system.
Collapse
Affiliation(s)
- Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Winanto
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- National University of Singapore, Faculty of Science (Department of Biological Science), Singapore
| | - Yingying Zeng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Cheryl Yi-Pin Lee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ze Qin Lim
- Infectious Diseases Translational Research Programme (IDTRP); Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore
| | - Pradeep Gautam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Justin Jang Hann Chu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Infectious Diseases Translational Research Programme (IDTRP); Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Sylvie Alonso
- Infectious Diseases Translational Research Programme (IDTRP); Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Science Institute, National University of Singapore, Singapore
| | - Shi-Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National Neuroscience Institute, Singapore
| |
Collapse
|
31
|
Sundstrom J, Vanderleeden E, Barton NJ, Redick SD, Dawes P, Murray LF, Olson MN, Tran K, Chigas SM, Orszulak AR, Church GM, Readhead B, Oh HS, Harlan DM, Knipe DM, Wang JP, Chan Y, Lim ET. Herpes Simplex Virus 1 Infection of Human Brain Organoids and Pancreatic Stem Cell-Islets Drives Organoid-Specific Transcripts Associated with Alzheimer's Disease and Autoimmune Diseases. Cells 2024; 13:1978. [PMID: 39682726 PMCID: PMC11640215 DOI: 10.3390/cells13231978] [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: 10/16/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Viral infections leading to inflammation have been implicated in several common diseases, such as Alzheimer's disease (AD) and type 1 diabetes (T1D). Of note, herpes simplex virus 1 (HSV-1) has been reported to be associated with AD. We sought to identify the transcriptomic changes due to HSV-1 infection and anti-viral drug (acyclovir, ACV) treatment of HSV-1 infection in dissociated cells from human cerebral organoids (dcOrgs) versus stem cell-derived pancreatic islets (sc-islets) to gain potential biological insights into the relevance of HSV-1-induced inflammation in AD and T1D. We observed that differentially expressed genes (DEGs) in HSV-1-infected sc-islets were enriched for genes associated with several autoimmune diseases, most significantly, T1D, but also rheumatoid arthritis, psoriasis, Crohn's disease, and multiple sclerosis, whereas DEGs in HSV-1-infected dcOrgs were exclusively enriched for genes associated with AD. The ACV treatment of sc-islets was not as effective in rescuing transcript perturbations of autoimmune disease-associated genes. Finally, we identified gene ontology categories that were enriched for DEGs that were in common across, or unique to, viral treatment of dcOrgs and sc-islets, such as categories involved in the transferase complex, mitochondrial, and autophagy function. In addition, we compared transcriptomic signatures from HSV-1-infected sc-islets with sc-islets that were infected with the coxsackie B virus (CVB) that had been associated with T1D pathogenesis. Collectively, this study provides tissue-specific insights into the molecular effects of inflammation in AD and T1D.
Collapse
Affiliation(s)
- Jonathan Sundstrom
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Emma Vanderleeden
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nathaniel J. Barton
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sambra D. Redick
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Pepper Dawes
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Liam F. Murray
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Meagan N. Olson
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Khanh Tran
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Graduate Program in Biochemistry & Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Samantha M. Chigas
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Graduate Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Adrian R. Orszulak
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Graduate Program in Immunology and Microbiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - George M. Church
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Benjamin Readhead
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85281, USA
| | - Hyung Suk Oh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - David M. Harlan
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - David M. Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jennifer P. Wang
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Yingleong Chan
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Elaine T. Lim
- Department of Medicine, Division of Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
32
|
Chen S, Xu H, Li W, Nie Y, Xie Q, Chen W. Deciphering Immune Modulation in Chickens Co-Infected with ALV-J and CIAV: A Transcriptomic Approach. Microorganisms 2024; 12:2453. [PMID: 39770656 PMCID: PMC11676111 DOI: 10.3390/microorganisms12122453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Viral co-infections pose significant challenges, causing substantial economic losses worldwide in the poultry industry. Among these, avian lLeukosis virus subgroup J (ALV-J) and chicken infectious anemia virus (CIAV) are particularly concerning, as they frequently lead to co-infections in chickens, further compromising their immune defenses, increasing susceptibility to secondary infections and diminishing vaccine efficacy. While our previous studies have examined the pathogenicity and immunosuppressive effects of these co-infections in vitro and in vivo, the key genes and molecular pathways involved remain largely unexplored. This study investigates the synergistic effects of co-infection with ALV-J and CIAV through comprehensive transcriptome analysis using high-throughput sequencing. We identified 1007 differentially expressed mRNAs (DEmRNAs) and 62 differentially expressed miRNAs (DEmiRNAs) associated with the synergistic activation effects of co-infection, along with 331 DEmRNAs and 62 DEmiRNAs linked to specific activation processes. Notably, the immune suppression observed in co-infected chickens may be influenced by the enhanced utilization of reactive oxygen species (ROS) and oxidative stress pathways, which impact host immune responses. Furthermore, co-infection appears to employ distinct immune evasion strategies through the modulation of rRNA metabolism, differing from single infections. These insights provide a deeper understanding of the molecular mechanisms underlying immune suppression during viral co-infections and help develop targeted therapies and improve disease control in poultry, reducing economic losses.
Collapse
Affiliation(s)
- Sheng Chen
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
- Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Huijuan Xu
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
| | - Wenxue Li
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
| | - Yu Nie
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
| | - Qingmei Xie
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
- Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| | - Weiguo Chen
- State Key Laboratory of Swine and Poultry Breeding Industry & Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (S.C.); (H.X.); (W.L.); (Y.N.); (Q.X.)
- Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
| |
Collapse
|
33
|
Wang J, Chen Y, Cong J, Wang W. Metformin regulates cellulase production in Trichoderma reesei via calcium signaling and mitochondrial function. Microb Cell Fact 2024; 23:314. [PMID: 39574147 PMCID: PMC11580550 DOI: 10.1186/s12934-024-02593-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Trichoderma reesei is renowned for its cellulase-producing ability and is used for biofuel production from lignocellulose. In plants and fungi, cellulase production is induced by cellulose and suppressed by glucose; however, whether metformin can enhance cellulase production and mitochondrial function in T. reesei remains unclear. Metformin reduces blood glucose levels by inhibiting hepatic gluconeogenesis; therefore, it is worth investigating whether metformin transmission modulates cellulase biosynthesis in T. reesei. RESULTS Metformin increased cellulase production and the transcription of cellulase-related genes. It also enhanced the concentrations of Ca2+ in the cytosol and mitochondria and regulated the transcription levels of cellulase-related genes by modulating calcium homeostasis in T. reesei QM6a. In addition, metformin was identified as an antioxidant that can enhance cellulase activity by reducing reactive oxygen species (ROS). Our results demonstrated that metformin influences the state of mitochondria by enhancing mitochondrial activity and membrane potential to promote cellulase production. CONCLUSION Collectively, these results indicate that metformin enhances cellulase production, scavenges ROS, and protects mitochondrial activity in T. reesei.
Collapse
Affiliation(s)
- Jiajia Wang
- The State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yumeng Chen
- The State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Jiawei Cong
- The State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Wei Wang
- The State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
34
|
Wang H, Tang J, Yan S, Li C, Li Z, Xiong Z, Li Z, Tu C. Liquid-liquid Phase Separation in Aging: Novel Insights in the Pathogenesis and Therapeutics. Ageing Res Rev 2024; 102:102583. [PMID: 39566743 DOI: 10.1016/j.arr.2024.102583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/14/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
The intricate organization of distinct cellular compartments is paramount for the maintenance of normal biological functions and the orchestration of complex biochemical reactions. These compartments, whether membrane-bound organelles or membraneless structures like Cajal bodies and RNA transport granules, play crucial roles in cellular function. Liquid-liquid phase separation (LLPS) serves as a reversible process that elucidates the genesis of membranelles structures through the self-assembly of biomolecules. LLPS has been implicated in a myriad of physiological and pathological processes, encompassing immune response and tumor genesis. But the association between LLPS and aging has not been clearly clarified. A recent advancement in the realm of aging research involves the introduction of a new edition outlining the twelve hallmarks of aging, categorized into three distinct groups. By delving into the role and mechanism of LLPS in the formation of membraneless structures at a molecular level, this review encapsulates an exploration of the interaction between LLPS and these aging hallmarks, aiming to offer novel perspectives of the intricate mechanisms underlying the aging process and deeper insights into aging therapeutics.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Shuxiang Yan
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, Institute of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zhaoqi Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zijian Xiong
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China; Hunan Key Laboratory of Tumor Models and Individualized Medicine, Engineering Research Center of Artificial Intelligence-Driven Medical Device, The Second Xiangya Hospital of Central South University Changsha 410011, China, Changsha 410011, China; Shenzhen Research Institute of Central South University, Shenzhen 518063, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University Changsha 410011, China; Changsha Medical University, Changsha 410219, China
| |
Collapse
|
35
|
Zhang Y, Samuelson AV. Antiviral defense in aged Caenorhabditis elegans declines due to loss of DRH-1/RIG-I deSUMOylation via ULP-4/SENP7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623310. [PMID: 39605404 PMCID: PMC11601531 DOI: 10.1101/2024.11.12.623310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Innate host defense mechanisms require posttranslational modifications (PTM) to protect against viral infection. Age-associated immunosenescence results in increased pathogenesis and mortality in the elderly, but the contribution of altered PTM regulation to immunosenescence is unknown. SUMOylation is a rapid and reversible post-translational modification that has been implicated in age-associated disease and plays conflicting roles in viral replication and antiviral defenses in mammals. We have discovered in Caenorhabditis elegans that induction of antiviral defense is regulated through SUMOylation of DRH-1, the ortholog of the DEAD/H-box helicase and cytosolic pattern recognition receptor RIG-I, and that this regulation breaks down during aging. We find the SUMO isopeptidase ULP-4 is essential for deSUMOylation of DRH-1 and activation of the intracellular pathogen response (IPR) after exposure to Orsay virus (OV), a natural enteric C. elegans pathogen. ULP-4 promotes stabilization of DRH-1, which translocates to the mitochondria to activate the IPR in young animals exposed to virus. Loss of either drh-1 or ulp-4 compromises antiviral defense resulting in a failure to clear the virus and signs of intestinal pathogenesis. During aging, expression of ulp-4 decreases, which results in increased proteosomal degradation of DRH-1 and loss of the IPR. Mutating the DRH-1 SUMOylated lysines resulted in the constitutive activation of the IPR in young animals and partially rescued the age-associated lost inducibility of the IPR. Our work establishes that aging results in dysregulated SUMOylation and loss of DRH-1, which compromises antiviral defense and creates a physiological shift to favor chronic pathological infection in older animals.
Collapse
|
36
|
Zhao R, Jiang C, Yuan Y, Zhang S, Ghonaim AH, Che C, Li X, Jin M, Jin E, Zeng X, Li S, Ren M. Isoleucine Enhanced the Function of the Small Intestinal Mucosal Barrier in Weaned Piglets to Alleviate Rotavirus Infection. Animals (Basel) 2024; 14:3146. [PMID: 39518871 PMCID: PMC11545378 DOI: 10.3390/ani14213146] [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: 09/12/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Rotavirus (RV) is a major cause of diarrhea in young children and animals, especially piglets, leading to substantial economic losses in the global pig industry. Isoleucine (Ile), a branched-chain amino acid, plays an important role in regulating nutrient metabolism and has been shown to improve diarrhea. This study aimed to evaluate the effects of Ile supplementation on the mucosal immune barrier of the small intestine in RV-infected weaned piglets. METHODS Forty-eight 21-day-old weaned piglets were randomly divided into three dietary treatments (each treatment was subdivided into two groups, eight replicates per group), with 0%, 0.5%, or 1% Ile added for 15 days, and then, one group from each treatment was challenged with RV. RESULTS The results showed that 1% Ile added to the diet promoted the healthy development of the intestinal mucosa. Ile could restore the reduced villus height in the ileum and the goblet cell number in the duodenum and ileum to normal levels, improving the intestinal epithelial tight junctions in RV-infected piglets. Additionally, Ile increased the activity of lipase, amylase, and sucrase, as well as superoxide dismutase (SOD) and glutathione (GSH), along with the expression of SIgA, DEFβ1, and DEFβ2 in parts of the small intestine. CONCLUSIONS The addition of Ile to the diet mitigated the effects of RV infection on intestinal morphology and mucosal barrier function, as well as the physiological functions of weaned piglets, and improved the antioxidant and immune functions of the piglets to some extent. These findings offer valuable insights, contributing to a deeper understanding of the role of Ile in supporting intestinal health.
Collapse
Affiliation(s)
- Rongkun Zhao
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Changsheng Jiang
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Yuchen Yuan
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Shen Zhang
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Ahmed H. Ghonaim
- National Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
- Desert Research Center, Cairo 11435, Egypt
| | - Chuanyan Che
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Xiaojin Li
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Mengmeng Jin
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Erhui Jin
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China;
| | - Shenghe Li
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| | - Man Ren
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China; (R.Z.); (C.J.); (Y.Y.); (S.Z.); (C.C.); (X.L.); (M.J.); (E.J.)
| |
Collapse
|
37
|
Zhao G, Zhao P, Wang Y, Zhang H, Zhu Y, Zhong J, You W, Shen G, Luo C, Mei O, Wu X, Li J, Shu Y, Wang H, Wagstaff W, Luu HH, Bi Y, Shi LL, Reid RR, He TC, Jiang L, Tang W, Fan J, Tang Z. GAPDH suppresses adenovirus-induced oxidative stress and enables a superfast production of recombinant adenovirus. Genes Dis 2024; 11:101344. [PMID: 39188753 PMCID: PMC11345542 DOI: 10.1016/j.gendis.2024.101344] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/15/2024] [Indexed: 08/28/2024] Open
Abstract
Recombinant adenovirus (rAdV) is a commonly used vector system for gene transfer. Efficient initial packaging and subsequent production of rAdV remains time-consuming and labor-intensive, possibly attributable to rAdV infection-associated oxidative stress and reactive oxygen species (ROS) production. Here, we show that exogenous GAPDH expression mitigates adenovirus-induced ROS-associated apoptosis in HEK293 cells, and expedites adenovirus production. By stably overexpressing GAPDH in HEK293 (293G) and 293pTP (293GP) cells, respectively, we demonstrated that rAdV-induced ROS production and cell apoptosis were significantly suppressed in 293G and 293GP cells. Transfection of 293G cells with adenoviral plasmid pAd-G2Luc yielded much higher titers of Ad-G2Luc at day 7 than that in HEK293 cells. Similarly, Ad-G2Luc was amplified more efficiently in 293G than in HEK293 cells. We further showed that transfection of 293GP cells with pAd-G2Luc produced much higher titers of Ad-G2Luc at day 5 than that of 293pTP cells. 293GP cells amplified the Ad-G2Luc much more efficiently than 293pTP cells, indicating that exogenous GAPDH can further augment pTP-enhanced adenovirus production. These results demonstrate that exogenous GAPDH can effectively suppress adenovirus-induced ROS and thus accelerate adenovirus production. Therefore, the engineered 293GP cells represent a superfast rAdV production system for adenovirus-based gene transfer and gene therapy.
Collapse
Affiliation(s)
- Guozhi Zhao
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Piao Zhao
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai 200000, China
| | - Hui Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 4000430, China
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Beijing Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jiamin Zhong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Wulin You
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu 214071, China
| | - Guowei Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Benq Medical Center, The Affiliated Benq Hospital of Nanjing Medical University, Nanjing, Jiangsu 210019, China
| | - Changqi Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Yibin Second People's Hospital, Affiliated with West China School of Medicine, Yibin, Sichuan 644000, China
| | - Ou Mei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedics, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Xingye Wu
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jingjing Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Oncology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261053, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, The National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hongwei Wang
- Division of Research and Development, Decoding Therapeutics, Inc., Mt Prospect, IL 60056, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yang Bi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, The National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Li Jiang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Tang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ziwei Tang
- Departments of Urology, Endocrinology, Orthopedic Surgery, and Gastroenterological Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
38
|
Jarosz-Griffiths H, Caley L, Lara-Reyna S, Savic S, Clifton I, McDermott M, Peckham D. Heightened mitochondrial respiration in CF cells is normalised by triple CFTR modulator therapy through mechanisms involving calcium. Heliyon 2024; 10:e39244. [PMID: 39498005 PMCID: PMC11532250 DOI: 10.1016/j.heliyon.2024.e39244] [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: 02/06/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Background Cystic fibrosis (CF) is associated with increased resting energy expenditure. However, the introduction of elexacaftor/tezacaftor/ivacaftor (ETI) has resulted in a paradigm shift in nutritional status for many people with CF, with increase body mass index and reduction in the need for nutritional support. While these changes are likely to reflect improved clinical status and an associated downregulation of energy expenditure, they may also reflect drug-induced alterations in metabolic perturbations within CF cells. We hypothesise that some of these changes relate to normalisation of mitochondrial respiration in CF. Methods Using wild-type (WT) and F508del/F508del CFTR human bronchial epithelial cell lines (HBE cell lines) and baby hamster kidney (BHK) cells we examined the impact of ETI on cellular metabolism. We monitored mitochondrial respiration, using Seahorse extracellular flux assays and monitored mitochondrial reactive oxygen species (mROS) and intracellular calcium levels by flow cytometry. Results Increased mitochondrial respiration was found in HBE cell lines and BHK cells expressing CFTR F508del/F508del when assessing basal, maximal, spare respiratory capacities and ATP production, as well as increased mitochondrial ROS generated via forward electron transport. ETI significantly decreased basal, maximal, spare respiratory capacity and ATP production to WT levels or below. Calcium blocker, BAPTA-AM normalised mitochondrial respiration, suggesting a calcium-mediated mechanism. ETI decreased intracellular calcium levels in CF cells to the same extent as BAPTA-AM, highlighting the importance of calcium and chloride in mitochondrial respiration in CF. Conclusions CF cell lines exhibit increased mitochondrial respiration, which can be downregulated by ETI therapy through mechanisms involving calcium.
Collapse
Affiliation(s)
| | - L.R. Caley
- Leeds Institute of Medical Research, University of Leeds, United Kingdom
| | - S. Lara-Reyna
- Institute of Microbiology and Infection, University of Birmingham, United Kingdom
| | - S. Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, United Kingdom
- Department of Clinical Immunology and Allergy, St James's University Hospital, United Kingdom
| | - I.J. Clifton
- Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - M.F. McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, United Kingdom
| | - D.G. Peckham
- Leeds Institute of Medical Research, University of Leeds, United Kingdom
- Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
39
|
Pérez SE, Gooz M, Maldonado EN. Mitochondrial Dysfunction and Metabolic Disturbances Induced by Viral Infections. Cells 2024; 13:1789. [PMID: 39513896 PMCID: PMC11545457 DOI: 10.3390/cells13211789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Viruses are intracellular parasites that utilize organelles, signaling pathways, and the bioenergetics machinery of the cell to replicate the genome and synthesize proteins to build up new viral particles. Mitochondria are key to supporting the virus life cycle by sustaining energy production, metabolism, and synthesis of macromolecules. Mitochondria also contribute to the antiviral innate immune response. Here, we describe the different mechanisms involved in virus-mitochondria interactions. We analyze the effects of viral infections on the metabolism of glucose in the Warburg phenotype, glutamine, and fatty acids. We also describe how viruses directly regulate mitochondrial function through modulation of the activity of the electron transport chain, the generation of reactive oxygen species, the balance between fission and fusion, and the regulation of voltage-dependent anion channels. In addition, we discuss the evasion strategies used to avoid mitochondrial-associated mechanisms that inhibit viral replication. Overall, this review aims to provide a comprehensive view of how viruses modulate mitochondrial function to maintain their replicative capabilities.
Collapse
Affiliation(s)
- Sandra E. Pérez
- Centro de Investigación Veterinaria de Tandil (CIVETAN), UNCPBA-CICPBA-CONICET, Campus Universitario, Tandil CC7000, Buenos Aires, Argentina;
| | - Monika Gooz
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
| | - Eduardo N. Maldonado
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD 506 Drug Discovery Building, 70 President Street, MSC 139, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
40
|
Sánchez-García S, Castrillo A, Boscá L, Prieto P. Potential Beneficial Role of Nitric Oxide in SARS-CoV-2 Infection: Beyond Spike-Binding Inhibition. Antioxidants (Basel) 2024; 13:1301. [PMID: 39594443 PMCID: PMC11591382 DOI: 10.3390/antiox13111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
SARS-CoV-2, the causative virus for the COVID-19 disease, uses its spike glycoprotein to bind to human ACE2 as a first step for viral entry into the cell. For this reason, great efforts have been made to find mechanisms that disrupt this interaction, avoiding the infection. Nitric oxide (NO) is a soluble endogenous gas with known antiviral and immunomodulatory properties. In this study, we aimed to test whether NO could inhibit the binding of the viral spike to ACE2 in human cells and its effects on ACE2 enzymatic activity. Our results show that ACE2 activity was decreased by the NO donors DETA-NONOate and GSNO and by the NO byproduct peroxynitrite. Furthermore, we found that DETA-NONOate could break the spike-ACE2 interaction using the spike from two different variants (Alpha and Gamma) and in two different human cell types. Moreover, the same result was obtained when using NO-producing murine macrophages, while no significant changes were observed in ACE2 expression or distribution within the cell. These results support that it is worth considering NO as a therapeutic agent for COVID-19, as previous reports have suggested.
Collapse
Affiliation(s)
- Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Unidad de Biomedicina (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
| | - Patricia Prieto
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain
| |
Collapse
|
41
|
Lin S, Liu Q, Xie Y, Zhang Q. Accelerated Wound Healing of Tetrahedral-Framework Nucleic Acid Nanozymes with High Penetration and Antioxidant Capacity. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1693. [PMID: 39513773 PMCID: PMC11547624 DOI: 10.3390/nano14211693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024]
Abstract
The wound repair process usually leads to a non-functioning mass of fibrotic tissue because of the oxidative imbalance of deep tissue layers. However, how to improve the penetration of active ingredients into deeper layers and regulate oxidative imbalances to create a regenerative microenvironment still remains a challenge. In this study, we designed a novel tetrahedral-framework nucleic acid (tFNA) nanozyme that could penetrate the skin/mucosa barrier as deep as 450 μm within 24 h. We also demonstrated the protective role of tFNAs on the mitochondrial structural and functional integrity and inhibition of reactive oxygen species production to repair oxidative imbalances through ERK1/2-Nrf2-HO-1 during repair processes. It was found that the proliferative state and the migration ability of postburn cells in vitro were accelerated, and the early closure of wounds in vivo was significantly promoted. This study therefore provides a promising strategy to efficiently regulate the oxidative imbalances in the deep layers of the skin during wound healing.
Collapse
Affiliation(s)
- Shiyu Lin
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Qian Liu
- Department of Stomatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200070, China;
| | - Yu Xie
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China;
| | - Qi Zhang
- Department of Oral and Maxillofacial Implantology, Shanghai PerioImplant Innovation Center, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China;
| |
Collapse
|
42
|
Jadaun P, Harshithkumar R, Seniya C, Gaikwad SY, Bhoite SP, Chandane-Tak M, Borse S, Chavan-Gautam P, Tillu G, Mukherjee A. Mitochondrial resilience and antioxidant defence against HIV-1: unveiling the power of Asparagus racemosus extracts and Shatavarin IV. Front Microbiol 2024; 15:1475457. [PMID: 39507335 PMCID: PMC11537936 DOI: 10.3389/fmicb.2024.1475457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Asparagus racemosus (AR), an Ayurvedic botanical, possesses various biological characteristics, yet its impact on HIV-1 replication remains to be elucidated. This study aimed to investigate the inhibitory effects of AR root extracts and its principal bioactive molecule, Shatavarin IV (Shatavarin), on HIV-1 replication and their role in mitigating mitochondrial dysfunction during HIV-1 infection, utilizing both in vitro and in silico methodologies. The cytotoxicity of the extracts was evaluated using MTT and ATPlite assays. In vitro anti-HIV-1 activity was assessed in TZM-bl cells against X4 and R5 subtypes, and confirmed in peripheral blood mononuclear cells using HIV-1 p24 antigen capture ELISA and viral copy number assessment. Mechanistic insights were obtained through enzymatic assays targeting HIV-1 Integrase, Protease and Reverse Transcriptase. Shatavarin's activity was also validated via viral copy number and p24 antigen capture assays, along with molecular interaction studies against key HIV-1 replication enzymes. HIV-1 induced mitochondrial dysfunction was evaluated by detecting mitochondrial reactive oxygen species (ROS), calcium accumulation, mitochondrial potential, and caspase activity within the infected cells. Non-cytotoxic concentrations of both aqueous and hydroalcoholic extracts derived from Asparagus racemosus roots displayed dose-dependent inhibition of HIV-1 replication. Notably, the hydroalcoholic extract exhibited superior Reverse Transcriptase activity, complemented by moderate activity observed in the Protease assay. Molecular interaction studies revealed that Shatavarin IV, the key bioactive constituent of AR, formed hydrogen bonds within the active binding pocket site residues crucial for HIV replication enzyme catalysis, suggesting its potential in attenuating HIV-1 infection. Mitochondrial dysfunction induced by HIV-1 infection, marked by increased oxidative stress, mitochondrial calcium overload, loss of mitochondrial membrane potential, and elevated caspase activity, was effectively mitigated by treatment with AR extracts and Shatavarin IV. These findings underscore the potential of AR extracts and Shatavarin IV as antiviral agents, while enhancing mitochondrial function during HIV-1 infection. In conclusion, Asparagus racemosus extracts, particularly Shatavarin IV, demonstrate promising inhibitory effects against HIV-1 replication while concurrently ameliorating mitochondrial dysfunction induced by the virus. These findings suggest the therapeutic potential of AR extracts and Shatavarin in combating HIV-1 infection and improving mitochondrial health.
Collapse
Affiliation(s)
- Pratiksha Jadaun
- Division of Virology, ICMR – National Institute of Translational Virology and AIDS Research, Pune, India
| | - R. Harshithkumar
- Division of Virology, ICMR – National Institute of Translational Virology and AIDS Research, Pune, India
| | - Chandrabhan Seniya
- School of Biosciences, Engineering and Technology, VIT Bhopal University, Bhopal, India
| | - Shraddha Y. Gaikwad
- Division of Virology, ICMR – National Institute of Translational Virology and AIDS Research, Pune, India
| | | | - Madhuri Chandane-Tak
- Division of Virology, ICMR – National Institute of Translational Virology and AIDS Research, Pune, India
| | - Swapnil Borse
- AYUSH-Center of Excellence, CCIH-Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Preeti Chavan-Gautam
- AYUSH-Center of Excellence, CCIH-Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Girish Tillu
- AYUSH-Center of Excellence, CCIH-Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Anupam Mukherjee
- Division of Virology, ICMR – National Institute of Translational Virology and AIDS Research, Pune, India
| |
Collapse
|
43
|
Stefano GB, Weissenberger S, Ptacek R, Anders M, Raboch J, Büttiker P. Viruses and Mitochondrial Dysfunction in Neurodegeneration and Cognition: An Evolutionary Perspective. Cell Mol Neurobiol 2024; 44:68. [PMID: 39417916 PMCID: PMC11486811 DOI: 10.1007/s10571-024-01503-3] [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: 08/29/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Mitochondria, the cellular powerhouses with bacterial evolutionary origins, play a pivotal role in maintaining neuronal function and cognitive health. Several viruses have developed sophisticated mechanisms to target and disrupt mitochondrial function which contribute to cognitive decline and neurodegeneration. The interplay between viruses and mitochondria might be traced to their co-evolutionary history with bacteria and may reflect ancient interactions that have shaped modern mitochondrial biology.
Collapse
Affiliation(s)
- George B Stefano
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00, Prague, Czech Republic.
| | - Simon Weissenberger
- Department of Psychology, University of New York in Prague, Prague, Czech Republic
| | - Radek Ptacek
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00, Prague, Czech Republic
| | - Martin Anders
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00, Prague, Czech Republic
| | - Jiri Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00, Prague, Czech Republic
| | - Pascal Büttiker
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00, Prague, Czech Republic
| |
Collapse
|
44
|
Leclerc S. [Mitochondria play a critical role in the replication of Herpes simplex virus of type 1]. Med Sci (Paris) 2024; 40:716-718. [PMID: 39450954 DOI: 10.1051/medsci/2024120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Affiliation(s)
- Simon Leclerc
- Department of biological and environmental science and nanoscience center, Université de Jyväskylä Jyväskylä Finlande
| |
Collapse
|
45
|
Liu Q, He Q, Tao X, Yu P, Liu S, Xie Y, Zhu W. Resveratrol inhibits rabies virus infection in N2a cells by activating the SIRT1/Nrf2/HO-1 pathway. Heliyon 2024; 10:e36494. [PMID: 39281556 PMCID: PMC11399676 DOI: 10.1016/j.heliyon.2024.e36494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/12/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024] Open
Abstract
Rabies is a highly lethal infectious disease with no existing treatment available, thus investigating effective antiviral compounds to control rabies virus (RABV) infection is of utmost importance. Resveratrol is a natural phenolic compound that, as a phytoalexin, exhibits several biological activities, including antiviral activity. In this study, we evaluated the inhibitory effect of resveratrol on RABV infection and investigated its molecular antiviral mechanism. We found that resveratrol significantly inhibited RABV infection, including the phases of adsorption, replication, and release, and also directly inactivated RABV and inhibited its infectivity. However, resveratrol had no significant effect on RABV internalization. Resveratrol also reduced RABV-induced oxidative stress, specifically reactive oxygen species and malondialdehyde levels. Western blotting analysis revealed that resveratrol enhanced antioxidant signaling via the SIRT1/Nrf2/HO-1 pathway and inhibited viral replication. Viral infection was enhanced after SIRT1 knockdown, which inhibited the SIRT1/Nrf2/HO-1 antioxidant signaling pathway, suggesting that this pathway plays an important role in RABV replication. Overall, resveratrol prevented the adsorption, replication, and release of RABV and directly inactivated RABV, but failed to inhibit RABV internalization. Furthermore, resveratrol activated the SIRT1/Nrf2/HO-1 pathway to inhibit RABV replication and suppressed RABV-induced oxidative stress. These findings highlight the therapeutic potential of resveratrol for fighting RABV infections.
Collapse
Affiliation(s)
- Qian Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Qing He
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Xiaoyan Tao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Pengcheng Yu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Shuqing Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yuan Xie
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Wuyang Zhu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| |
Collapse
|
46
|
Mehta SK, Pradhan RB. Phytochemicals in antiviral drug development against human respiratory viruses. Drug Discov Today 2024; 29:104107. [PMID: 39032810 DOI: 10.1016/j.drudis.2024.104107] [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: 12/25/2023] [Revised: 05/30/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
This review explores the potential antiviral properties of various plant-based compounds, including polyphenols, phytochemicals, and terpenoids. It emphasizes the diverse functionalities of compounds such as epigallocatechin-3-gallate (EGCG), quercetin, griffithsin (GRFT,) resveratrol, linalool, and carvacrol in the context of respiratory virus infections, including SARS-CoV-2. Emphasizing their effectiveness in modulating immune responses, disrupting viral envelopes, and influencing cellular signaling pathways, the review underlines the imperative for thorough research to establish safety and efficacy. Additionally, the review underscores the necessity of well-designed clinical trials to evaluate the efficacy and safety of these compounds as potential antiviral agents. This approach would establish a robust framework for future drug development efforts focused on bolstering host defense mechanisms against human respiratory viral infections.
Collapse
Affiliation(s)
- Surya Kant Mehta
- Laboratory of Algal Biology, Department of Botany, School of Life Sciences, Mizoram University, Aizawl, PIN 796004, Mizoram, India.
| | - Ran Bahadur Pradhan
- Laboratory of Algal Biology, Department of Botany, School of Life Sciences, Mizoram University, Aizawl, PIN 796004, Mizoram, India
| |
Collapse
|
47
|
Sharma R, Pandey S, Prasad M, Prasad A. Catalase regulation during plant-virus-vector interaction. PHYSIOLOGIA PLANTARUM 2024; 176:e14580. [PMID: 39403868 DOI: 10.1111/ppl.14580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024]
Abstract
Plant-virus-host interaction is a complex process involving several players. A constant arms race between the hosts and viruses has led to their co-evolution. Reactive oxygen species (ROS) are important signaling molecules that regulate plant growth, development, and stress responses. Barley yellow dwarf virus (BYDV) has a wide host range and infects several plant species such as barley, rice, oats, wheat, etc. A recent study by Tian et al. (2024) has highlighted that the movement protein (MP) of BYDV is involved in manipulation of the host ROS pathway to promote viral multiplication as well as transmission. The findings display the multifaceted role of a viral protein that is otherwise involved in movement. The limited coding ability of viruses is compensated by their proteins having multiple roles in the modulation of several different host molecular pathways. This is one of the key reasons for viruses being successful pathogens despite their limited coding ability.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Botany, Kurukshetra University, Kurukshetra, India
| | - Saurabh Pandey
- Department of Agriculture, Guru Nanak Dev University, Amritsar, India
| | - Manoj Prasad
- Department of Genetics, University of Delhi South Campus, New Delhi, India
- National Institute of Plant Genome Research, New Delhi, India
| | - Ashish Prasad
- Department of Botany, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
48
|
Slama Schwok A, Henri J. Long Neuro-COVID-19: Current Mechanistic Views and Therapeutic Perspectives. Biomolecules 2024; 14:1081. [PMID: 39334847 PMCID: PMC11429791 DOI: 10.3390/biom14091081] [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: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 09/30/2024] Open
Abstract
Long-lasting COVID-19 (long COVID) diseases constitute a real life-changing burden for many patients around the globe and, overall, can be considered societal and economic issues. They include a variety of symptoms, such as fatigue, loss of smell (anosmia), and neurological-cognitive sequelae, such as memory loss, anxiety, brain fog, acute encephalitis, and stroke, collectively called long neuro-COVID-19 (long neuro-COVID). They also include cardiopulmonary sequelae, such as myocardial infarction, pulmonary damage, fibrosis, gastrointestinal dysregulation, renal failure, and vascular endothelial dysregulation, and the onset of new diabetes, with each symptom usually being treated individually. The main unmet challenge is to understand the mechanisms of the pathophysiologic sequelae, in particular the neurological symptoms. This mini-review presents the main mechanistic hypotheses considered to explain the multiple long neuro-COVID symptoms, namely immune dysregulation and prolonged inflammation, persistent viral reservoirs, vascular and endothelial dysfunction, and the disruption of the neurotransmitter signaling along various paths. We suggest that the nucleoprotein N of SARS-CoV-2 constitutes a "hub" between the virus and the host inflammation, immunity, and neurotransmission.
Collapse
Affiliation(s)
- Anny Slama Schwok
- Sorbonne Université, INSERM U938, Biology and Cancer Therapeutics, Centre de Recherche Saint Antoine, Saint Antoine Hospital, 75231 Paris, France
| | - Julien Henri
- Sorbonne Université, CNRS UMR 7238, Laboratoire de Biologie Computationnelle et Quantitative, Institut de Biologie Paris-Seine, 75005 Paris, France
| |
Collapse
|
49
|
Frasson I, Diamante L, Zangrossi M, Carbognin E, Pietà AD, Penna A, Rosato A, Verin R, Torrigiani F, Salata C, Dizanzo MP, Vaccaro L, Cacchiarelli D, Richter SN, Montagner M, Martello G. Identification of druggable host dependency factors shared by multiple SARS-CoV-2 variants of concern. J Mol Cell Biol 2024; 16:mjae004. [PMID: 38305139 PMCID: PMC11411213 DOI: 10.1093/jmcb/mjae004] [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: 02/03/2023] [Revised: 06/23/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024] Open
Abstract
The high mutation rate of SARS-CoV-2 leads to the emergence of multiple variants, some of which are resistant to vaccines and drugs targeting viral elements. Targeting host dependency factors, e.g. cellular proteins required for viral replication, would help prevent the development of resistance. However, it remains unclear whether different SARS-CoV-2 variants induce conserved cellular responses and exploit the same core host factors. To this end, we compared three variants of concern and found that the host transcriptional response was conserved, differing only in kinetics and magnitude. Clustered regularly interspaced short palindromic repeats screening identified host genes required for each variant during infection. Most of the genes were shared by multiple variants. We validated our hits with small molecules and repurposed the US Food and Drug Administration-approved drugs. All the drugs were highly active against all the tested variants, including new variants that emerged during the study (Delta and Omicron). Mechanistically, we identified reactive oxygen species production as a key step in early viral replication. Antioxidants such as N-acetyl cysteine (NAC) were effective against all the variants in both human lung cells and a humanized mouse model. Our study supports the use of available antioxidant drugs, such as NAC, as a general and effective anti-COVID-19 approach.
Collapse
Affiliation(s)
- Ilaria Frasson
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Linda Diamante
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Manuela Zangrossi
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Elena Carbognin
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Alessandro Penna
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua 35128, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | | | - Lorenzo Vaccaro
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples Federico II, Naples 80138, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Microbiology and Virology Unit, Padua University Hospital, Padua 35128, Italy
| | - Marco Montagner
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Graziano Martello
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| |
Collapse
|
50
|
Amratia PS, Kerr-Jones LE, Chapman L, Marsden M, Clement M, Stanton RJ, Humphreys IR. Cytomegalovirus-induced peroxynitrite promotes virus entry and contributes to pathogenesis in a murine model of infection. mBio 2024; 15:e0315223. [PMID: 38953361 PMCID: PMC11323495 DOI: 10.1128/mbio.03152-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
There are no licensed vaccines for human cytomegalovirus (HCMV), and current antiviral drugs that target viral proteins are toxic and prone to resistance. Targeting host pathways essential for virus replication provides an alternate strategy that may reduce opportunities for drug resistance to occur. Oxidative stress is triggered by numerous viruses including HCMV. Peroxynitrite is a reactive nitrogen species that is formed during oxidative stress. Herein, we identified that HCMV rapidly induces the generation of intracellular peroxynitrite upon infection in a manner partially dependent upon xanthine oxidase generation. Peroxynitrite promoted HCMV infection in both cell-free and cell-associated infection systems in multiple cell types. Inhibiting peroxynitrite within the first 24 hours of infection prevented HCMV replication and peroxynitrite promoted cell entry and pp65 translocation into the host cell nuclei. Furthermore, using the murine cytomegalovirus model, we demonstrated that antagonizing peroxynitrite significantly reduces cytomegalovirus replication and pathogenesis in vivo. Overall, our study highlights a proviral role for peroxynitrite in CMV infection and implies that RNS and/or the mechanisms that induce their production could be targeted as a novel strategy to inhibit HCMV infection. IMPORTANCE Human cytomegalovirus (HCMV) causes significant disease in individuals with impaired or immature immune systems, such as transplant patients and after congenital infection. Antiviral drugs that target the virus directly are toxic and are susceptible to antiviral drug resistance due to virus mutations. An alternate strategy is to target processes within host cells that are required by the virus for replication. Herein, we show that HCMV infection triggers a highly reactive molecule, peroxynitrite, during the initial stages of infection. Peroxynitrite was required for the initial entry of the virus into the cell and promotes virus replication in multiple cell types, suggesting a broad pro-viral function. Importantly, targeting peroxynitrite dramatically inhibited cytomegalovirus replication in cells in the laboratory and in mice, suggesting that therapeutic targeting of this molecule and/or the cellular functions it regulates could represent a novel strategy to inhibit HCMV infection.
Collapse
Affiliation(s)
- Pragati S. Amratia
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Lauren E. Kerr-Jones
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Lucy Chapman
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Morgan Marsden
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Mathew Clement
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Richard J. Stanton
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Ian R. Humphreys
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|