1
|
de Campos Martin Berber G, de Sarges KML, da Cruz TCD, Roma EH, Miyajima F, Almeida JR, de Souza CF, da Silva AA, Vallinoto IMVC, Vallinoto ACR, Queiroz MAF, Baccin TG, de Castro AC, Vieira AT, de Azevedo FKS, da Costa Pereira A, Falcão LFM, Quaresma JAS, Kehdy F, Santos ET, Siqueira MM, Garcia CC, Dos Santos EJM, Slhessarenko RD. Polymorphisms in HLA genes among Brazilian patients hospitalized with COVID-19: Insights from a multicentric study. Microb Pathog 2025; 204:107542. [PMID: 40188974 DOI: 10.1016/j.micpath.2025.107542] [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: 01/17/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Immunogenetic factors such as human leukocyte antigen (HLA) alleles, have yielded contrasting associations with protection or increased chances of hospitalization due to COVID-19 worldwide. This case-control study included 834 patients with confirmed COVID-19 diagnosis from five Brazilian states: Ceará (n = 110), Mato Grosso (n = 192), Pará (n = 209), Rio de Janeiro (n = 211) and Rio Grande do Sul (n = 112). Genotyping was performed using the Axiom™ Human Genotyping SARS-CoV-2 array, targeting single nucleotide polymorphisms in HLA class I and II genes; HLA alleles were imputed for eight loci. Among the 15 preselected candidate alleles, only DQA1∗05:01 (p = 0.015) in the state of Ceará remained significantly associated with hospitalization. The meta-analysis of the most frequent alleles in all states revealed that HLA-DPA1∗01:03 (p = 0.0229, OR = 0.76, 95 % CI = 0.60-0.96) and HLA-DPB1∗04:01 (p = 0.0474, OR = 0.78, 95 % CI = 0.611.00) were associated with protection against hospitalization, whereas HLA-DPA1∗02:01 (p = 0.0259, OR = 1.37, 95 % CI = 1.04-1.80), HLA-DQA1∗05:01 (p = 0.0133, OR = 1.40, 95 % CI = 1.07-1.82), and HLA-DRB1∗03:01 (p = 0.0276, OR: 1.59, 95 % CI: 1.05-2.40) associated with increased risk of hospitalization. HLA evolutionary divergence (HED) scores were significantly higher among the non-hospitalized group for the HLA-A locus, which has been shown to be a protective factor for the most severe forms and consequently hospitalization due to COVID-19.
Collapse
Affiliation(s)
| | - Kevin Matheus Lima de Sarges
- Post-graduation Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém, PA, Brazil
| | - Thais Campos Dias da Cruz
- Post-graduation Program in Health Sciences, Federal University of Mato Grosso School of Medicine, Cuiabá, MT, Brazil
| | - Eric Henrique Roma
- Laboratory of Immunology and Imunogenetics in Infectious Diseases, Evandro Chagas National Infectology Institute, Rio de Janeiro, RJ, Brazil
| | - Fábio Miyajima
- Analytical Competence Molecular Epidemiology Laboratory, Fiocruz Genomic Surveillance Network, Fiocruz Ceará, Eusebio, CE, Brazil
| | - Jorge Reis Almeida
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences, Fluminense Federal University, Niterói, RJ, Brazil
| | - Cíntia Fernandes de Souza
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences, Fluminense Federal University, Niterói, RJ, Brazil; Laboratory of Respiratory, Exanthematic, Enteric Viruses and Viral Emergencies, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Andrea Alice da Silva
- Multiuser Laboratory for Research Support in Nephrology and Medical Sciences, Fluminense Federal University, Niterói, RJ, Brazil
| | - Izaura Maria Vieira Cayres Vallinoto
- Post-graduation Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém, PA, Brazil; Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Antônio Carlos Rosário Vallinoto
- Post-graduation Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém, PA, Brazil; Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | - Maria Alice Freitas Queiroz
- Post-graduation Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém, PA, Brazil; Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém, PA, Brazil
| | | | | | - Angélica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexandre da Costa Pereira
- Laboratory of Molecular Genetics and Cardiology, InCor - Hospital das Clínicas - FMUSP, University of São Paulo, SP, Brazil
| | | | | | - Fernanda Kehdy
- Laboratory of Leprosy, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Eduardo Tarazona Santos
- Laboratory of Human Genetic Diversity, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marilda Mendonça Siqueira
- Laboratory of Respiratory, Exanthematic, Enteric Viruses and Viral Emergencies, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Cristiana Couto Garcia
- Laboratory of Respiratory, Exanthematic, Enteric Viruses and Viral Emergencies, Instituto Oswaldo Cruz, Rio de Janeiro, RJ, Brazil; Integrated Research Group on Biomarkers, René Rachou Institute, Fiocruz Minas, Belo Horizonte, MG, Brazil
| | - Eduardo José Melo Dos Santos
- Post-graduation Program in Biology of Infectious and Parasitic Agents, Federal University of Pará, Belém, PA, Brazil.
| | | |
Collapse
|
2
|
Rajsfus BF, Nascimento-Júnior JX, Chichierchio MS, Todeschini AR, Oliveira IDA, Borges FDS, Barroso SP, Mohana-Borges R, Camacho-Pereira J, Zancan P, Allonso D. Chikungunya virus infection under high glucose environment reverts insulin unresponsiveness and promotes cellular metabolic shift to increase anaplerosis and virion production. Life Sci 2025; 376:123729. [PMID: 40404124 DOI: 10.1016/j.lfs.2025.123729] [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: 03/24/2025] [Revised: 05/08/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
AIMS Chikungunya virus (CHIKV) is a medically significant arbovirus responsible for Chikungunya fever (CHIKF), a debilitating disease marked by persistent joint and muscle pain. The most severe outcomes of CHIKF, including mortality, are frequently observed in individuals with comorbidities diabetes mellitus (DM). This study aimed to investigate how varying glucose concentrations influence CHIKV infection. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVEC) were cultured in normal glucose (NG - 5.5 mM glucose) or high glucose (HG - 25 mM glucose) to mimic normoglycemia and hyperglycemia conditions. Cells were infected with CHIKV and changes in host glucose metabolism were evaluated. Metabolic flux, viral replication, and cellular insulin sensitivity were assessed through biochemical, molecular analyses and metabolomics. KEY FINDINGS CHIKV infection modulates host cell metabolism in a glucose-dependent manner. Under NG conditions, the virus regulates glucose metabolism to support replication and virion production. In contrast, HG environments enhance viral replication, exploiting the altered metabolic landscape. Notably, CHIKV restores insulin sensitivity in HG conditions, leading to increased glucose uptake. It also promotes anaplerotic reactions by diverting tricarboxylic acid (TCA) cycle intermediates toward amino acid synthesis and upregulates glycolytic flux into the hexosamine biosynthesis pathway (HBP). SIGNIFICANCE These findings provide mechanistic insight into how hyperglycemia associated with DM can exacerbate CHIKV pathogenesis. The virus's ability to hijack and redirect host metabolic processes in high glucose environments may underline the worsened disease severity observed in diabetic patients. Understanding these interactions could inform targeted therapeutic strategies for managing CHIKF in individuals with metabolic comorbidities.
Collapse
Affiliation(s)
- Bia Francis Rajsfus
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biotecnologia e Bioengenharia Estrutural, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - José Xavier Nascimento-Júnior
- Laboratório de Oncobiologia Molecular, The metaboliZSm' grouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marina Santos Chichierchio
- Instituto de Bioquímica Médica Leopoldo de Méis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Adriane Regina Todeschini
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Isadora de Araujo Oliveira
- Laboratório de Glicobiologia Estrutural e Funcional, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Fernando de Souza Borges
- Laboratório de Biologia Molecular, Instituto de Pesquisas Biomédicas, Hospital Naval Marcílio Dias Marinha do Brasil, Rio de Janeiro 20725-090, Brazil
| | - Shana Priscila Barroso
- Laboratório de Biologia Molecular, Instituto de Pesquisas Biomédicas, Hospital Naval Marcílio Dias Marinha do Brasil, Rio de Janeiro 20725-090, Brazil
| | - Ronaldo Mohana-Borges
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biotecnologia e Bioengenharia Estrutural, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Juliana Camacho-Pereira
- Instituto de Bioquímica Médica Leopoldo de Méis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Patrícia Zancan
- Laboratório de Oncobiologia Molecular, The metaboliZSm' grouP, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Diego Allonso
- Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biotecnologia e Bioengenharia Estrutural, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
3
|
Gao X, Wu B, Qiu Y, Feng S, Zhang J, Miao J. STING contributes to the inflammation and proliferation of Staphylococcus aureus via mitochondrial reactive oxygen species-hypoxic inducible factor 1α axis in epithelial cells. Infect Immun 2025:e0013825. [PMID: 40387431 DOI: 10.1128/iai.00138-25] [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: 03/11/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025] Open
Abstract
Staphylococcus aureus infection poses a serious threat to the dairy industry and public health safety. The stimulator of interferon gene (STING) signaling pathway has been well established as effective in defending against viral infections. However, the role of STING is controversial during bacterial infections. Herein, we provide an insight into the role of STING during S. aureus infection. Our data revealed that the STING signaling pathway was activated in S. aureus-infected cells. In vitro investigations demonstrated that inhibiting STING reduced inflammation, hypoxia-inducible factor-1 alpha (HIF1α) expression, and mitochondrial reactive oxygen species (mROS) production. Interestingly, blocking HIF1α eliminated the escalation of inflammation associated with STING. Additionally, suppressing mROS production significantly reduced HIF1α expression and inflammation levels, while elevating mROS had the opposite effect. These results indicate that STING promoted inflammation through the mROS-HIF1α pathway. Given that glycolysis is driven by HIF1α, we investigated the role of glycolysis during infection. As expected, STING-elevated inflammation was linked with HIF1α-driven glycolysis. In terms of pathogenesis, STING contributed to S. aureus proliferation within cells and mouse mammary glands. Collectively, our findings demonstrate that STING facilitates infection via the mROS-HIF1α-glycolysis axis, highlighting its potential as a promising anti-inflammatory target.
Collapse
Affiliation(s)
- Xing Gao
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Binfeng Wu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yawei Qiu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shiyuan Feng
- Sanya Research Institute, Nanjing Agricultural University, Sanya, China
| | - Jinqiu Zhang
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, China
| | - Jinfeng Miao
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
4
|
Luo W, Zhao M, Gu M, Huang J, Wu S, Zhao B. The role of Tim-3+T cell subsets in the peripheral blood of patients with COVID-19 and diabetes. iScience 2025; 28:112339. [PMID: 40330891 PMCID: PMC12053769 DOI: 10.1016/j.isci.2025.112339] [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: 07/02/2024] [Revised: 11/03/2024] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Corona Virus Disease 2019 (COVID-19) and diabetes interact to influence disease severity, yet their combined immunological characteristics remain unclear. Here, we analyzed Tim-3+ T cells in patients with COVID-19, Type 1 Diabetes (T1D), or both conditions. COVID-19 reduced peripheral T cell subsets but increased Tim-3+ cells, while T1D and COVID-19 with T1D showed the opposite pattern. Patients with Type 2 Diabetes (T2D) exhibited no significant alterations. In human samples and mouse models, Tim-3+ T cells demonstrated impaired activation and cytokine production. RNA-seq analysis in mice and RT-PCR analysis in human samples together identified the dysregulation of the JAK-STAT pathway in Tim-3+ T cells. These findings highlight Tim-3-mediated JAK-STAT dysregulation in T-cells as a potential mechanism linking COVID-19 and T1D, offering insights for therapeutic targeting.
Collapse
Affiliation(s)
- Wenjun Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Furong Laboratory, Changsha, China
- CSU-Sinocare Research Center for Nutrition and Metabolic Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Mingjiu Zhao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Furong Laboratory, Changsha, China
- CSU-Sinocare Research Center for Nutrition and Metabolic Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Mengyao Gu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jiaqi Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Furong Laboratory, Changsha, China
- CSU-Sinocare Research Center for Nutrition and Metabolic Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Shiyao Wu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Dermatology and Immunology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Furong Laboratory, Changsha, China
- CSU-Sinocare Research Center for Nutrition and Metabolic Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Liu L, Yu J, Liu Y, Xie L, Hu F, Liu H. Hypoxia-driven angiogenesis and metabolic reprogramming in vascular tumors. Front Cell Dev Biol 2025; 13:1572909. [PMID: 40443737 PMCID: PMC12119610 DOI: 10.3389/fcell.2025.1572909] [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: 02/07/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Hypoxia is a hallmark of the tumor microenvironment (TME), and it plays a crucial role in the occurrence and progression in vascular tumors. Under hypoxic conditions, hypoxia-inducible factor 1-alpha (HIF-1α) is stabilized, inducing changes in the expression of various target genes involved in angiogenesis, metabolism, and cell survival. This includes the upregulation of pro-angiogenic factors like VEGF, which promotes the formation of dysfunctional blood vessels, contributing to the worsening of the hypoxic microenvironment. At the same time, hypoxia induces a metabolic shift toward glycolysis, even in the presence of oxygen, supporting tumor cell survival and proliferation by providing necessary energy and biosynthetic precursors. This review discusses the molecular mechanisms by which hypoxia regulates angiogenesis and metabolic reprogramming in vascular tumors, highlighting the intricate link between these processes, and explores potential therapeutic strategies to target these pathways in order to develop effective treatment strategies for patients.
Collapse
Affiliation(s)
- Lu Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayun Yu
- Department of Radiotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fan Hu
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- Department of Pediatric Cardiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defect and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatric Pulmonology and Immunology, WCSUH-Tianfu·Sichuan Provincial Children’s Hospital, Sichuan University, Meishan, China
| |
Collapse
|
6
|
Wang X, Li Y, Wang Q, Xia F, Chen W, You C, Ma L. Glucose concentration predicting mortality in patients with COVID-19: A propensity score-matched study. Am J Med Sci 2025:S0002-9629(25)01035-3. [PMID: 40374008 DOI: 10.1016/j.amjms.2025.05.003] [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: 07/10/2024] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Patients with coronavirus disease-19 (COVID-19) often develop systemic inflammation, which is associated with increased mortality. Elevated blood glucose levels can exacerbate the cytokine storm, further worsening disease severity and accelerating patient death. Therefore, this study aims to investigate the association between glucose levels and mortality in hospitalized patients, providing insights into the importance of optimizing glucose management in hospitalized COVID-19 patients. METHODS A retrospective cohort study was conducted, involving adult COVID-19 patients in a university hospital. The primary outcome was in-hospital mortality. Propensity score matching (PSM) was utilized to match patients' baseline characteristics. Discrimination capacity of different models was assessed using C-statistics, net reclassification improvement (NRI), and integrated discrimination improvement (IDI). Trends in blood glucose over time were detected using the ordinary least squares model. RESULTS Among the 4583 COVID-19 patients during the study period, 2147 (46.8%) exhibited normal glycemia, while 2436 (53.2%) had admission hyperglycemia. After adjusting for confounding factors through multivariate regression analysis, patients with hyperglycemia showed significantly higher odds of in-hospital mortality (adjusted odds ratio [aOR]: 3.10, 95% CI: 2.25 to 4.28; P < 0.001). PSM analysis yielded similar results (aOR: 2.66, 95% CI: 2.09 to 3.41; P < 0.001). The incorporation of admission glucose significantly improved C-statistics (P < 0.001), IDI (P < 0.001), and NRI (P < 0.001) for predicting mortality. CONCLUSION This study concludes that blood glucose levels ≥ 6.1 mmol/L can independently predict all-cause mortality and clinical sequelae in COVID-19 patients. Furthermore, even a mild increase in blood glucose was associated with a significantly higher risk of mortality in these patients. These findings underscore the importance of managing hyperglycemia and monitoring blood glucose in individuals with COVID-19.
Collapse
Affiliation(s)
- Xing Wang
- Traumatic Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Li
- Research Core Facility of West China Hospital, Sichuan University
| | - Qiao Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wuqian Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Wu JS, Xu CY, Mo SM, Wu XM, Du ZB, Che L, Zhang YL, Yang KL, Li TD, Ge SX, Zhang TY, Lin ZN, Lin YC. Palmitoylated COX-2 Cys555 reprogrammed mitochondrial metabolism in pyroptotic inflammatory injury in patients with post-acute COVID-19 syndrome. J Adv Res 2025:S2090-1232(25)00299-1. [PMID: 40349960 DOI: 10.1016/j.jare.2025.05.005] [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: 01/07/2025] [Revised: 05/04/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025] Open
Abstract
INTRODUCTION The complex interplay between protein palmitoylation, mitochondrial dynamics, and inflammatory responses plays a pivotal role in respiratory diseases. One significant feature of post-acute coronavirus disease 2019 (COVID-19) syndrome (PACS) is the occurrence of a storm of inflammatory cytokines related to the NOD-like receptor protein 3 (NLRP3). However, the specific mechanisms via which palmitoylation affects mitochondrial function and its impact on the NLRP3 inflammasome under pathological respiratory conditions remain to be elucidated. OBJECTIVE This study aimed to investigate how protein palmitoylation influences inflammatory responses and mitochondrial dynamics in respiratory diseases, such as those induced by the SARS-CoV-2 spike S protein in PACS, thereby providing a therapeutic target for inflammatory lung injury. METHODS In vivo experiments were conducted using AdV5-pADM-CMV-COVID-19-S (AdV5-S) nasal drip-treated C57BL/6 mice to assess NLRP3 inflammasome activation and inflammatory response. In vitro experiments were performed using pCMV-S-transfected human lung epithelial BEAS-2B cells to analyze the effects of DHHC5-mediated palmitoylation of cyclooxygenase-2 (COX-2) at cysteine 555 (COX-2Cys555) on mitochondrial metabolism and NLRP3 inflammasome activation. RESULTS Palmitoylation of COX-2Cys555 enhanced its interaction with hexokinase 2 (HK2) to regulate mitochondrial metabolic reprogramming, leading to NLRP3 inflammasome activation and pyroptosis. Pharmacological and genetic suppression of palmitoylation diminished the mitochondrial localization of palmitoylated COX-2 and its interaction with HK2, thereby reducing mitochondrial metabolic reprogramming. Furthermore, genetic intervention targeting DHHC5 (shDhhc5) alleviated NLRP3 activation and pyroptosis, mitigating the chronic inflammatory damage associated with PACS. CONCLUSION This study highlights the regulatory role of COX-2Cys555 palmitoylation in mitochondrial metabolism and lung inflammatory injury, and suggests potential therapeutic targets to combat respiratory pathogenesis linked to palmitoylated COX-2.
Collapse
Affiliation(s)
- Jia-Shen Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Chi-Yu Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Su-Min Mo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Xin-Mou Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ze-Bang Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Lin Che
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Yi-Ling Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Kai-Li Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Ting-Dong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Sheng-Xiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Tian-Ying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Zhong-Ning Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Yu-Chun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiang'an Hospital of Xiamen University, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
8
|
Xiong S, Liu Z, Yao J, Huang S, Ding X, Yu H, Lin T, Zhang X, Zhao F. HIF-1α regulated GLUT1-mediated glycolysis enhances Treponema pallidum-induced cytokine responses. Cell Commun Signal 2025; 23:219. [PMID: 40346557 PMCID: PMC12065375 DOI: 10.1186/s12964-025-02211-1] [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: 12/17/2024] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Syphilis, caused by Treponema pallidum (Tp), represents a significant public health challenge. The clinical manifestations of syphilis are attributed to local inflammatory responses induced by Tp, notably monocyte infiltration into local lesions and the secretion of inflammatory cytokines. However, the mechanisms driving cytokine production in response to Tp infection remain largely unknown. Given that increased glycolysis is associated with inflammatory responses, we aimed to investigate the role of glycolysis in Tp-induced secretion of inflammatory cytokines. In this study, we found that Tp promotes the secretion of inflammatory cytokines IL-6, IL-8, and CCL2 from monocytes while enhancing glycolysis through increased GLUT1 plasma membrane expression and glucose uptake. Importantly, inhibiting glycolysis and GLUT1 reduced the Tp-induced secretion of monocyte inflammatory cytokines. Additionally, Tp significantly increased HIF-1α expression and induced its nuclear translocation, thereby promoting glycolysis by upregulating the expression of GLUT1 and LDHA glycolytic enzymes. Knockdown of HIF-1α inhibits Tp-induced monocyte cytokine secretion, highlighting the crucial role of HIF-1α-mediated glycolysis in the cytokine response to Tp. Also, expression of HIF-1α and an increase in glycolysis were confirmed in patients with syphilis. In conclusion, we demonstrated that HIF-1α-regulated GLUT1-mediated glycolysis enhances inflammatory cytokine secretion following Tp infection. Our findings not only elucidate the mechanism of glycolysis in Tp-induced inflammatory responses in monocytes but also contribute to the development of a potential biomarker in syphilis diagnosis and treatment.
Collapse
Affiliation(s)
- Shun Xiong
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
| | - Zhaoping Liu
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Jiangchen Yao
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shaobin Huang
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuan Ding
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
| | - Han Yu
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
| | - Ting Lin
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaohong Zhang
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China.
| | - Feijun Zhao
- MOE Key Lab of Rare Pediatric Diseases & Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China.
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China.
- Department of Clinical Laboratory Medicine, Changsha Central Hospital, Changsha, 410004, P.R. China.
| |
Collapse
|
9
|
Mezouar S, Mege J. Monitoring Macrophage Polarization in Infectious Disease, Lesson From SARS-CoV-2 Infection. Rev Med Virol 2025; 35:e70034. [PMID: 40148134 PMCID: PMC11976041 DOI: 10.1002/ird3.70006] [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: 03/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The concept of macrophage polarization has been largely used in human diseases to define a typology of activation of myeloid cells reminiscent of lymphocyte functional subsets. In COVID-19, several studies have investigated myeloid compartment dysregulation and macrophage polarization as an indicator of disease prognosis and monitoring. SARS-CoV-2 induces an in vitro activation state in monocytes and macrophages that does not match the polarization categories in most studies. In COVID-19 patients, monocytes and macrophages are activated but they do not show a polarization profile. Therefore, the investigation of polarization under basic conditions was not relevant to assess monocyte and macrophage activation. The analysis of monocytes and macrophages with high-throughput methods has allowed the identification of new functional subsets in the context of COVID-19. This approach proposes an innovative stratification of myeloid cell activation. These new functional subsets of myeloid cells would be better biomarkers to assess the risk of complications in COVID-19, reserving the concept of polarization for pharmacological programme evaluation. This review reappraises the polarization of monocytes and macrophages in viral infections, particularly in COVID-19.
Collapse
Affiliation(s)
- Soraya Mezouar
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille UniversityHIPE Human LabMarseilleFrance
| | - Jean‐Louis Mege
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Department of ImmunologyLa Timone HospitalMarseilleFrance
| |
Collapse
|
10
|
da Silva FPG, Matte R, Wiedmer DB, da Silva APG, Menin RM, Barbosa FB, Meneguzzi TAM, Pereira SB, Fausto AT, Klug L, Melim BP, Beltrão CJ. HIF-1α Pathway in COVID-19: A Scoping Review of Its Modulation and Related Treatments. Int J Mol Sci 2025; 26:4202. [PMID: 40362439 PMCID: PMC12071378 DOI: 10.3390/ijms26094202] [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: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The COVID-19 pandemic, driven by SARS-CoV-2, has led to a global health crisis, highlighting the virus's unique molecular mechanisms that distinguish it from other respiratory pathogens. It is known that the Hypoxia-Inducible Factor 1α (HIF-1α) activates a complex network of intracellular signaling pathways regulating cellular energy metabolism, angiogenesis, and cell survival, contributing to the wide range of clinical manifestations of COVID-19, including Post-Acute COVID-19 Syndrome (PACS). Emerging evidence suggests that dysregulation of HIF-1α is a key driver of systemic inflammation, silent hypoxia, and pathological tissue remodeling in both the acute and post-acute phases of the disease. This scoping review was conducted following PRISMA-ScR guidelines and registered in INPLASY. It involved a literature search in Scopus and PubMed, supplemented by manual reference screening, with study selection facilitated by Rayyan software. Our analysis clarifies the dual role of HIF-1α, which may either worsen inflammatory responses and viral persistence or support adaptive mechanisms that reduce cellular damage. The potential for targeting HIF-1α therapeutically in COVID-19 is complex, requiring further investigation to clarify its precise role and translational applications. This review deepens the molecular understanding of SARS-CoV-2-induced cellular and tissue dysfunction in hypoxia, offering insights for improving clinical management strategies and addressing long-term sequelae.
Collapse
Affiliation(s)
- Felipe Paes Gomes da Silva
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Rafael Matte
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - David Batista Wiedmer
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Arthur Paes Gomes da Silva
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Rafaela Makiak Menin
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| | - Fernanda Bressianini Barbosa
- School of Medicine, Faculdade Evangélica Mackenzie do Paraná, R. Padre Anchieta, no. 2770—Bigorrilho, Curitiba 80730-000, PR, Brazil;
| | - Thainá Aymê Mocelin Meneguzzi
- School of Medicine, Universidade Nove de Julho—UNINOVE, Rua Vergueiro, 249—Liberdade, São Paulo 01504-001, SP, Brazil; (T.A.M.M.); (B.P.M.)
| | - Sabrina Barancelli Pereira
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Amanda Terres Fausto
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Larissa Klug
- School of Medicine, Universidade Positivo—UP, R. Professor Pedro Viriato Parigot de Souza, 5300, Curitiba 81280-330, PR, Brazil; (S.B.P.); (A.T.F.); (L.K.)
| | - Bruna Pinheiro Melim
- School of Medicine, Universidade Nove de Julho—UNINOVE, Rua Vergueiro, 249—Liberdade, São Paulo 01504-001, SP, Brazil; (T.A.M.M.); (B.P.M.)
| | - Claudio Jose Beltrão
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná—PUCPR, R. Imaculada Conceição, 1155—Prado Velho, Curitiba 80215-901, PR, Brazil; (F.P.G.d.S.); (R.M.); (D.B.W.); (A.P.G.d.S.); (R.M.M.)
| |
Collapse
|
11
|
Davanzo GG, Castelucci BG, de Souza GF, Muraro SP, Menezes Dos Reis L, de Oliveira IB, Fachi JL, Virgilio-da-Silva JV, Berçot MR, Fernandes MF, de Oliveira S, Araujo NVP, Ribeiro G, de Castro G, Costa WLG, Santoro AL, Rodrigues-Luiz GF, do Carmo HRP, Breder I, Mori MA, Farias AS, Martins-de-Souza D, Guarnieri JW, Wallace DC, Vinolo MAR, Proença-Módena JL, Beheshti A, Sposito AC, Moraes-Vieira PM. Obesity-Induced Metabolic Priming Exacerbates SARS-CoV-2 Inflammation. Immunology 2025. [PMID: 40265287 DOI: 10.1111/imm.13934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
Despite the early recognition that individuals living with obesity are more prone to develop adverse outcomes during COVID-19, the mechanisms underlying these conditions are still unclear. During obesity, an accumulation of free fatty acids (FFAs) in the circulation promotes low-grade inflammation. Here, we show that FFAs induce epigenetic reprogramming of monocytes, exacerbating their inflammatory profile after SARS-CoV-2 infection, a mechanism named metabolic-primed immunity. Monocytes from people with obesity or primed with palmitate, a central component of circulating FFAs, presented elevated viral load and higher gene expression of IL-6. Palmitate-primed monocytes upregulate fatty acid oxidation and FFAs entry into the mitochondria. FFA-derived acetyl-CoA is then converted into citrate, exiting the mitochondria and is used to support H3K18 histone acetylation, which regulates IL-6 accessibility. Ingestion of palm oil by lean and healthy individuals increased circulating FFAs levels and was sufficient to exacerbate the inflammatory profile of monocytes upon SARS-CoV-2 infection. Our findings demonstrate that obesity-derived FFAs induce the metabolic priming of monocytes, which exacerbates the inflammatory response observed in people with severe COVID-19.
Collapse
Affiliation(s)
- Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Bianca Gazieri Castelucci
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Gabriela Fabiano de Souza
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Stéfanie Primon Muraro
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Larissa Menezes Dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | | | - José Luís Fachi
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - João Victor Virgilio-da-Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Marcelo Rodrigues Berçot
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Mariane Font Fernandes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Nathalia Vitoria Pereira Araujo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Guilherme Ribeiro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Gisele de Castro
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Webster Leonardo Guimarães Costa
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Adriana Leandra Santoro
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Gabriela Flavia Rodrigues-Luiz
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Helison Rafael P do Carmo
- Department of Clinical Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil
| | - Ikaro Breder
- Department of Clinical Medicine, School of Medical Sciences, University of Campinas, São Paulo, Brazil
| | - Marcelo A Mori
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Alessandro S Farias
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Autoimmune Research Laboratory, Department of Genetics, Microbiology, and Immunology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Joseph W Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| | - José Luiz Proença-Módena
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| | - Afshin Beheshti
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrei C Sposito
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, São Paulo, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, São Paulo, Brazil
| |
Collapse
|
12
|
Michalak KP, Michalak AZ, Brenk-Krakowska A. Acute COVID-19 and LongCOVID syndrome - molecular implications for therapeutic strategies - review. Front Immunol 2025; 16:1582783. [PMID: 40313948 PMCID: PMC12043656 DOI: 10.3389/fimmu.2025.1582783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been recognized not only for its acute effects but also for its ability to cause LongCOVID Syndrome (LCS), a condition characterized by persistent symptoms affecting multiple organ systems. This review examines the molecular and immunological mechanisms underlying LCS, with a particular focus on autophagy inhibition, chronic inflammation, oxidative, nitrosative and calcium stress, viral persistence and autoimmunology. Potential pathophysiological mechanisms involved in LCS include (1) autoimmune activation, (2) latent viral persistence, where SARS-CoV-2 continues to influence host metabolism, (3) reactivation of latent pathogens such as Epstein-Barr virus (EBV) or cytomegalovirus (CMV), exacerbating immune and metabolic dysregulation, and (4) possible persistent metabolic and inflammatory dysregulation, where the body fails to restore post-infection homeostasis. The manipulation of cellular pathways by SARS-CoV-2 proteins is a critical aspect of the virus' ability to evade immune clearance and establish long-term dysfunction. Viral proteins such as NSP13, ORF3a and ORF8 have been shown to disrupt autophagy, thereby impairing viral clearance and promoting immune evasion. In addition, mitochondrial dysfunction, dysregulated calcium signaling, oxidative stress, chronic HIF-1α activation and Nrf2 inhibition create a self-sustaining inflammatory feedback loop that contributes to tissue damage and persistent symptoms. Therefore understanding the molecular basis of LCS is critical for the development of effective therapeutic strategies. Targeting autophagy and Nrf2 activation, glycolysis inhibition, and restoration calcium homeostasis may provide novel strategies to mitigate the long-term consequences of SARS-CoV-2 infection. Future research should focus on personalized therapeutic interventions based on the dominant molecular perturbations in individual patients.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | | - Alicja Brenk-Krakowska
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| |
Collapse
|
13
|
Gabaev I, Rowland A, Jovanovic E, Gawden-Bone CM, Crozier TWM, Teixeira-Silva A, Greenwood EJD, Gerber PP, Wit N, Nathan JA, Matheson NJ, Lehner PJ. CRISPR-Cas9 genetic screens reveal regulation of TMPRSS2 by the Elongin BC-VHL complex. Sci Rep 2025; 15:11907. [PMID: 40195420 PMCID: PMC11976923 DOI: 10.1038/s41598-025-95644-0] [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: 07/16/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
The TMPRSS2 cell surface protease is used by a broad range of respiratory viruses to facilitate entry into target cells. Together with ACE2, TMPRSS2 represents a key factor for SARS-CoV-2 infection, as TMPRSS2 mediates cleavage of viral spike protein, enabling direct fusion of the viral envelope with the host cell membrane. Since the start of the COVID-19 pandemic, TMPRSS2 has gained attention as a therapeutic target for protease inhibitors which would inhibit SARS-CoV-2 infection, but little is known about TMPRSS2 regulation, particularly in cell types physiologically relevant for SARS-CoV-2 infection. Here, we performed an unbiased genome-wide CRISPR-Cas9 library screen, together with a library targeted at epigenetic modifiers and transcriptional regulators, to identify cellular factors that modulate cell surface expression of TMPRSS2 in human colon epithelial cells. We find that endogenous TMPRSS2 is regulated by the Elongin BC-VHL complex and HIF transcription factors. Depletion of Elongin B or treatment of cells with PHD inhibitors resulted in downregulation of TMPRSS2 and inhibition of SARS-CoV-2 infection. We show that TMPRSS2 is still utilised by SARS-CoV-2 Omicron variants for entry into colonic epithelial cells. Our study enhances our understanding of the regulation of endogenous surface TMPRSS2 in cells physiologically relevant to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Ildar Gabaev
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Alexandra Rowland
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Emilija Jovanovic
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Christian M Gawden-Bone
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Thomas W M Crozier
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Ana Teixeira-Silva
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Edward J D Greenwood
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Pehuén Pereyra Gerber
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Niek Wit
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - James A Nathan
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Nicholas J Matheson
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Paul J Lehner
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, UK.
| |
Collapse
|
14
|
Joshi G, Verma G, Kaur S, Ramasamy E, Chauhan N, Singh S, Jana P, Rizvi ZA, Kshetrapal P, Bhatnagar S, Pandey AK, Awasthi A, Das B, Guchhait P. Severe SARS-CoV-2 infection in diabetes was rescued in mice supplemented with metformin and/or αKG, and patients taking metformin, via HIF1α-IFN axis. Clin Transl Med 2025; 15:e70275. [PMID: 40200582 PMCID: PMC11978732 DOI: 10.1002/ctm2.70275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Affiliation(s)
- Garima Joshi
- Department of BiotechnologyRegional Centre for Biotechnology, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Garima Verma
- Department of BiotechnologyRegional Centre for Biotechnology, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Simrandeep Kaur
- Department of BiotechnologyRegional Centre for Biotechnology, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Ellango Ramasamy
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Navya Chauhan
- Department of BiotechnologyRegional Centre for Biotechnology, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Savita Singh
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Pradipta Jana
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Zaigham Abbas Rizvi
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Pallavi Kshetrapal
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Shinjini Bhatnagar
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | | | - Amit Awasthi
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Bhabatosh Das
- Translational Health Science Technology Institute, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| | - Prasenjit Guchhait
- Department of BiotechnologyRegional Centre for Biotechnology, National Capital Region Biotech Science ClusterFaridabadHaryanaIndia
| |
Collapse
|
15
|
Sun X, Yegambaram M, Lu Q, Garcia Flores AE, Pokharel MD, Soto J, Aggarwal S, Wang T, Fineman JR, Black SM. Mitochondrial fission produces a Warburg effect via the oxidative inhibition of prolyl hydroxylase domain-2. Redox Biol 2025; 81:103529. [PMID: 39978304 PMCID: PMC11889635 DOI: 10.1016/j.redox.2025.103529] [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: 12/19/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Excessive mitochondrial fission and a shift to a Warburg phenotype are hallmarks of pulmonary hypertension (PH), although the mechanistic link between these processes remains unclear. We show that in pulmonary arterial endothelial cells (PAEC), Drp1 overexpression induces mitochondrial fission and increases glycolytic ATP production and glycolysis. This is due to mitochondrial reactive oxygen species (mito-ROS)-mediated activation of hypoxia-inducible factor-1α (HIF-1α) signaling, and this is linked to hydrogen peroxide (H2O2)-mediated inhibition of prolyl hydroxylase domain-2 (PHD2) due to its cysteine 326 oxidation and dimerization. Furthermore, these findings are validated in PAEC isolated from a lamb model of PH, which are glycolytic (Shunt PAEC), exhibit increases in both H2O2 and PHD2 dimer levels. The overexpression of catalase reversed the PHD2 dimerization, decreased HIF-1α levels, and attenuated glycolysis in Shunt PAEC. Our data suggest that reducing PHD2 dimerization could be a potential therapeutic target for PH.
Collapse
Affiliation(s)
- Xutong Sun
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Manivannan Yegambaram
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Qing Lu
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Alejandro E Garcia Flores
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Marissa D Pokharel
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jamie Soto
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Saurabh Aggarwal
- The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
16
|
Zhang J, Ma Y, To WL, Chow S, To Tang H, Wong HK, Luo J, Hoi Cheung C, Bian Z. Impact of COVID-19 infection on mortality, diabetic complications and haematological parameters in patients with diabetes mellitus: a systematic review and meta-analysis. BMJ Open 2025; 15:e090986. [PMID: 40147989 PMCID: PMC11956398 DOI: 10.1136/bmjopen-2024-090986] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES SARS-CoV-2 poses significant challenges to people living with diabetes (PLWD). This systematic review aimed to explore the impact of COVID-19 on mortality, complications associated with diabetes and haematological parameters among PLWD. DESIGN Systematic review and meta-analysis using the Grading of Recommendations Assessment, Development and Evaluation (GRADE). DATA SOURCES EMBASE, MEDLINE, Cochrane Central Register of Controlled Trials and LILACS were searched between 1 December 2019 and 14 January 2025. ELIGIBILITY CRITERIA FOR SELECTING STUDIES Eligible studies included case-control and cohort studies involving PLWD categorised into two groups: those with confirmed SARS-CoV-2 infection and those without. DATA EXTRACTION AND SYNTHESIS Meta-analyses estimated the odds ratios (ORs) and mean differences (MDs) of outcomes including mortality, intensive care unit (ICU) admission, diabetic ketoacidosis (DKA), acute kidney injury, hospitalisation length and haematological parameters. We pooled results using random-effects models and assessed study quality with the Newcastle-Ottawa Scale. A funnel plot was used to detect potential publication bias. The overall certainty of evidence was assessed using GRADE. RESULTS 25 of 7266 unique studies were eligible, including 1 154674 PLWD (561 558 with COVID-19 and 593 116 without COVID-19). SARS-CoV-2 infection in PLWD was associated with significantly increased mortality (OR 2.52, 95% CI 1.45 to 4.36, I2=99%), acute kidney injury (3.69, 95% CI 2.75 to 4.94, I2=0%), random plasma glucose in subjects with type 1 diabetes (MD 20.38 mg/dL, 95% CI 7.39 to 33.36, I2=0%), haemoglobin A1C in subjects with type 2 diabetes (0.21%, 95% CI 0.05 to 0.38, I2=13%), creatinine (0.12 mg/dL, 95% CI 0.04 to 0.19, I2=0%), C reactive protein (38.30 mg/L, 95% CI 4.79 to 71.82, I2=82%) and D-dimer (1.52 µg/mL, 95% CI 0.73 to 2.31, I2=0%). No significant differences were observed in the incidence of ICU admission and DKA, hospitalisation length, haemoglobin, leucocyte, lymphocyte, neutrophil to lymphocyte ratio, platelet, blood urea nitrogen, estimated glomerular filtration rate, procalcitonin, albumin, ferritin and bilirubin among PLWD with and without SARS-CoV-2 infection. CONCLUSIONS SARS-CoV-2 infection is associated with elevated risks of mortality and acute kidney injury and poor glycaemic control in PLWD, alongside increased levels of inflammatory and coagulation biomarkers. These findings underscore the urgent need for tailored clinical management strategies for PLWD with COVID-19. PROSPERO REGISTRATION NUMBER CRD42023418039.
Collapse
Affiliation(s)
- Jialing Zhang
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Yanfang Ma
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Chinese EQUATOR Centre, Hong Kong SAR, People's Republic of China
| | - Wing Lam To
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Sen Chow
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Hiu To Tang
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Hoi Ki Wong
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Jingyuan Luo
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Chun Hoi Cheung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| | - Zhaoxiang Bian
- Vincent V.C Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
- Chinese EQUATOR Centre, Hong Kong SAR, People's Republic of China
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, People's Republic of China
| |
Collapse
|
17
|
Eghbalifard N, Nouri N, Rouzbahani S, Bakhshi M, Ghasemi Kahrizsangi N, Golafshan F, Abbasi F. Hypoxia signaling in cancer: HIF-1α stimulated by COVID-19 can lead to cancer progression and chemo-resistance in oral squamous cell carcinoma (OSCC). Discov Oncol 2025; 16:399. [PMID: 40138101 PMCID: PMC11947373 DOI: 10.1007/s12672-025-02150-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The potential implications of Coronavirus disease-2019 (COVID-19) on oral squamous cell carcinoma (OSCC) development, chemo-resistance, tumor recurrence, and patient outcomes are explored, emphasizing the urgent need for tailored therapeutic strategies to mitigate these risks. The role of hypoxia-inducible factor 1-alpha (HIF-1α) in OSCC studies has highlighted HIF-1α as a crucial prognostic marker in OSCC, with implications for disease prognosis and patient survival. Its overexpression has been linked to aggressive subtypes in early OSCC stages, indicating its significance as an early biomarker for disease progression. Moreover, dysplastic lesions with heightened HIF-1α expression exhibit a greater propensity for malignant transformation, underscoring its role in early oral carcinogenesis. Cancer patients, including those with OSCC, face an elevated risk of severe COVID-19 complications, which may further impact cancer progression and treatment outcomes. Understanding the interplay between COVID-19 infection, HIF-1α activation, and OSCC pathogenesis is crucial for enhancing clinical management strategies. So, insights from this review shed light on the significance of HIF-1α in OSCC tumorigenesis, metastasis formation, and patient prognosis. The review underscores the need for further research to elucidate the precise mechanisms through which HIF-1α modulates cancer progression and chemo-resistance in the context of COVID-19 infection. Such knowledge is essential for developing targeted therapeutic interventions to improve outcomes for OSCC patients.
Collapse
Affiliation(s)
- Negar Eghbalifard
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nikta Nouri
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shiva Rouzbahani
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Bakhshi
- Islamic Azad University of Najaf Abad, Affiliated Hospitals, Isfahan, Iran
| | - Negin Ghasemi Kahrizsangi
- Child Growth and Development Research Center, Research Institute for Primary Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Faraz Golafshan
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Abbasi
- Department of Obstetrics and Gynecology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran.
| |
Collapse
|
18
|
Wideman SK, Wali L, Kovtunyk V, Chou S, Gusel V, Telimaa H, Najmi C, Stoeva D, Stöckl J, Gualdoni GA, Gorki AD, Radivojev S. Nebulized 2-deoxylated glucose analogues inhibit respiratory viral infection in advanced in vitro airway models. Sci Rep 2025; 15:9515. [PMID: 40108297 PMCID: PMC11923073 DOI: 10.1038/s41598-025-94476-2] [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: 12/21/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Respiratory viral infections, such as those caused by rhinoviruses (RVs) and human corona viruses (HCoV), result in a serious strain on healthcare systems and public health, underscoring an urgent need for inhaled broad-spectrum antiviral therapies. However, their development is challenging, as no standardized in vitro methodologies that can fully replicate the in vivo environment have been established. In this work, we aimed to investigate the antiviral and anti-inflammatory effect of three 2-deoxylated glucose analogues (2-DGA): 2-deoxy-D-glucose, 2-fluoro-2-deoxy-D-glucose and 2-fluoro-2-deoxy-D-mannose (2-FDM), by utilizing advanced in vitro air-liquid interface (ALI) airway models. We demonstrated that commonly used ALI models have variable susceptibility to RV, HCoV and influenza A virus (IAV) infection. Further, we showed that 2-DGA have an anti-inflammatory effect and suppress respiratory viral replication in models mimicking the upper and lower respiratory airways. Moreover, we confirmed that 2-DGA can be delivered via nebulization in vitro, highlighting their potential to be used as broad-spectrum inhaled antivirals. Finally, our results demonstrate the importance of incorporating complex in vitro methodologies, such as primary cell ALI cultures and aerosol exposure, at an early stage of drug development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Johannes Stöckl
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
19
|
Soultsioti M, de Jong AWM, Blomberg N, Tas A, Giera M, Snijder EJ, Bárcena M. Perturbation of de novo lipogenesis hinders MERS-CoV assembly and release, but not the biogenesis of viral replication organelles. J Virol 2025; 99:e0228224. [PMID: 39976449 PMCID: PMC11915874 DOI: 10.1128/jvi.02282-24] [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: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Coronaviruses hijack host cell metabolic pathways and resources to support their replication. They induce extensive host endomembrane remodeling to generate viral replication organelles and exploit host membranes for assembly and budding of their enveloped progeny virions. Because of the overall significance of host membranes, we sought to gain insight into the role of host factors involved in lipid metabolism in cells infected with Middle East respiratory syndrome coronavirus (MERS-CoV). We employed a single-cycle infection approach in combination with pharmacological inhibitors, biochemical assays, lipidomics, and light and electron microscopy. Pharmacological inhibition of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FASN), key host factors in de novo fatty acid biosynthesis, led to pronounced inhibition of MERS-CoV particle release. Inhibition of ACC led to a profound metabolic switch in Huh7 cells, altering their lipidomic profile and inducing lipolysis. However, despite the extensive changes induced by the ACC inhibitor, the biogenesis of viral replication organelles remained unaffected. Instead, ACC inhibition appeared to affect the trafficking and post-translational modifications of the MERS-CoV envelope proteins. Electron microscopy revealed an accumulation of nucleocapsids in early budding stages, indicating that MERS-CoV assembly is adversely impacted by ACC inhibition. Notably, inhibition of palmitoylation resulted in similar effects, while supplementation of exogenous palmitic acid reversed the compound's inhibitory effects, possibly reflecting a crucial need for palmitoylation of the MERS-CoV spike and envelope proteins for their role in virus particle assembly.IMPORTANCEMiddle East respiratory syndrome coronavirus (MERS-CoV) is the etiological agent of a zoonotic respiratory disease of limited transmissibility between humans. However, MERS-CoV is still considered a high-priority pathogen and is closely monitored by WHO due to its high lethality rate of around 35% of laboratory-confirmed infections. Like other positive-strand RNA viruses, MERS-CoV relies on the host cell's endomembranes to support various stages of its replication cycle. However, in spite of this general reliance of MERS-CoV replication on host cell lipid metabolism, mechanistic insights are still very limited. In our study, we show that pharmacological inhibition of acetyl-CoA carboxylase (ACC), a key enzyme in the host cell's fatty acid biosynthesis pathway, significantly disrupts MERS-CoV particle assembly without exerting a negative effect on the biogenesis of viral replication organelles. Furthermore, our study highlights the potential of ACC as a target for the development of host-directed antiviral therapeutics against coronaviruses.
Collapse
Affiliation(s)
- M. Soultsioti
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - A. W. M. de Jong
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - N. Blomberg
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - A. Tas
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - E. J. Snijder
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, the Netherlands
| | - M. Bárcena
- Section Electron Microscopy, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
20
|
Kumar N, Gond C, Singh JD, Datta A. Molecular docking, pharmacological profiling, and MD simulations of glycolytic inhibitors targeting novel SARS CoV-2 main protease and spike protein. In Silico Pharmacol 2025; 13:44. [PMID: 40093584 PMCID: PMC11908997 DOI: 10.1007/s40203-025-00336-2] [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: 05/02/2024] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
Abstract Coronavirus infection (COVID-19), designated a global health emergency by the World Health Organization in 2020, continues to spur the search for effective therapeutics. The causative agent, SARS-CoV-2, depends on viral proteins and host metabolic reprogramming for replication. This study explores the potential of glycolytic inhibitors as dual-action agents against SARS-CoV-2, explicitly targeting the main protease and the spike protein due to their critical roles in viral replication and cellular entry. These inhibitors disrupt the activity of viral proteins and host cell glycolysis, thereby preventing viral propagation. Through a combination of virtual screening, molecular docking, and molecular dynamics simulations, fluoro-deoxy-glucose folate (FDGF) and N-(2-fluoro-3-(6-O-glucosylpropyl-azomycin)) were identified as potent candidates. The docking results showed strong binding affinities, with scores of -8.6 and -7.1 kcal/mol for main protease and -9.9 and - 7.5 kcal/mol for spike receptor-binding domain bound to ACE2. Further molecular dynamic simulations confirmed the stability of the FDGF complexes, with RMSD fluctuations consistently remained within 1.6-2.9 Å over a 100 ns trajectory. Additionally, MM-GBSA binding free energy calculations revealed favorable binding energies, underscoring the stability and potential efficacy of these compounds. Overall, the findings suggest that FDGF and N-(2-fluoro-3-(6-O-glucosylpropyl-azomycin)) show promise as SARS-CoV-2 therapeutics, warranting further in vitro and in vivo validation to confirm their antiviral potential. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-025-00336-2.
Collapse
Affiliation(s)
- Nikhil Kumar
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig S. K. Mazumdar Marg, Delhi, 110054 India
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016 India
| | - Chandraprakash Gond
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig S. K. Mazumdar Marg, Delhi, 110054 India
| | - Jai Deo Singh
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016 India
| | - Anupama Datta
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig S. K. Mazumdar Marg, Delhi, 110054 India
| |
Collapse
|
21
|
Bar-Tana J. mTORC1 syndrome (TorS): unifying paradigm for PASC, ME/CFS and PAIS. J Transl Med 2025; 23:297. [PMID: 40059164 PMCID: PMC11892318 DOI: 10.1186/s12967-025-06220-z] [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: 12/08/2024] [Accepted: 02/10/2025] [Indexed: 05/13/2025] Open
Abstract
Post-acute SarS-Cov2 (PASC), Myalgia encephalomyelitis/Chronic fatigue syndrome (ME/CFS) and Post-acute infection syndrome (PAIS) consist of chronic post-acute infectious syndromes, sharing exhaustive fatigue, post exertional malaise, intermittent pain, postural tachycardia and neuro-cognitive-psychiatric dysfunction. However, the concerned shared pathophysiology is still unresolved in terms of upstream drivers and transducers. Also, risk factors which may determine vulnerability/progression to the chronic phase still remain to be defined. In lack of drivers and a cohesive pathophysiology, the concerned syndromes still remain unmet therapeutic needs. 'mTORC1 Syndrome' (TorS) implies an exhaustive disease entity driven by sustained hyper-activation of the mammalian target of rapamycin C1 (mTORC1), and resulting in a variety of disease aspects of the Metabolic Syndrome (MetS), non-alcoholic fatty liver disease, chronic obstructive pulmonary disease, some cancers, neurodegeneration and other [Bar-Tana in Trends Endocrinol Metab 34:135-145, 2023]. TorS may offer a cohesive insight of PASC, ME/CFS and PAIS drivers, pathophysiology, vulnerability and treatment options.
Collapse
Affiliation(s)
- Jacob Bar-Tana
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
22
|
Thieulent CJ, Balasuriya UBR, Tseng A, Crossland NA, Stephens JM, Dittmar W, Staszkiewicz J, Richt JA, Carossino M. Diabetes exacerbates SARS-CoV-2 replication through ineffective pulmonary interferon responses, delayed cell-mediated immunity, and disruption of leptin signaling. Front Cell Infect Microbiol 2025; 15:1513687. [PMID: 40125513 PMCID: PMC11925909 DOI: 10.3389/fcimb.2025.1513687] [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: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/25/2025] Open
Abstract
Comorbidities, including obesity and type 2 diabetes mellitus (T2DM), are associated with increased disease severity and mortality following SARS-CoV-2 infection. Here, we investigated virus-host interactions under the effects of these comorbidities in diet-induced obesity (DIO) and leptin receptor-deficient (T2DM) mice following infection with SARS-CoV-2. DIO mice, as well as their lean counterparts, showed limited susceptibility to SARS-CoV-2 infection. In contrast, T2DM mice showed exacerbated pulmonary SARS-CoV-2 replication and delayed viral clearance associated with down-regulation of innate and adaptative immune gene signatures, ineffective type I interferon response, and delayed SARS-CoV-2-specific cell-mediated immune responses. While T2DM mice showed higher and prolonged SARS-CoV-2-specific immunoglobulin isotype responses compared to their lean counterparts, neutralizing antibody levels were equivalent. By silencing the leptin receptor in vitro using a human alveolar epithelial cell line, we observed an increase in SARS-CoV-2 replication and type I interferons. Altogether, our data provides for the first time evidence that disruption of leptin receptor signaling leading to obesity and T2DM induces altered type I interferon and cell-mediated responses against SARS-CoV-2, mediating increased viral replication and delayed clearance. These data shed light on the alteration of the innate immune pathway in the lung using in-depth transcriptomic analysis and on adaptive immune responses to SARS-CoV-2 under T2DM conditions. Finally, this study provides further insight into this risk factor aggravating SARS-CoV-2 infection and understanding the underlying cellular mechanisms that could help identify potential intervention points for this at-risk population.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/physiology
- SARS-CoV-2/immunology
- Mice
- COVID-19/immunology
- COVID-19/virology
- Virus Replication
- Receptors, Leptin/genetics
- Receptors, Leptin/metabolism
- Obesity/immunology
- Obesity/complications
- Signal Transduction
- Humans
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/complications
- Leptin/metabolism
- Interferon Type I/immunology
- Interferon Type I/metabolism
- Lung/immunology
- Lung/virology
- Immunity, Cellular
- Mice, Inbred C57BL
- Immunity, Innate
- Male
- Disease Models, Animal
- Antibodies, Neutralizing/blood
- Interferons
- Mice, Knockout
Collapse
Affiliation(s)
- Côme J. Thieulent
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Anna Tseng
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Nicholas A. Crossland
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Jacqueline M. Stephens
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Wellesley Dittmar
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jaroslaw Staszkiewicz
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Juergen A. Richt
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
- Louisiana Animal Disease Diagnostic Laboratory (LSU Diagnostics), School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
23
|
Yang H, Guan L, Xue Y, Li X, Gao L, Zhang Z, Zhang H, Ma H, Liu F, Huang X, Tong Z, Li J. Longitudinal multi-omics analysis of convalescent individuals with respiratory sequelae 6-36 months after COVID-19. BMC Med 2025; 23:134. [PMID: 40038650 PMCID: PMC11881263 DOI: 10.1186/s12916-025-03971-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Approximately 10-30% of individuals continue to experience symptoms classified as post-acute sequelae of coronavirus disease 2019 (COVID-19 (PASC)). PASC is a multisystem condition primarily characterized by respiratory symptoms, such as reduced diffusing capacity for carbon monoxide (DLco). Although many studies have investigated the pathogenesis of acute COVID-19, the long-term molecular changes in COVID-19 convalescents with PASC remain poorly understood. METHODS We prospectively recruited 70 individuals who had been diagnosed with COVID-19 from 7 January 2020 to 29 May 2020 (i.e., COVID-19 convalescents); we performed follow-up visits at 6 months, 1 year, 2 years, and 3 years after hospital discharge. Thirty-five healthy controls (CONs), recruited from a physical examination center before the COVID-19 pandemic, served as a comparison group. We explored the proteomic and metabolomic profiles of 174 plasma samples from the 70 COVID-19 convalescents and 35 CONs. RESULTS We performed a comprehensive molecular analysis of COVID-19 convalescents to investigate host changes up to 3 years after hospital discharge. Our multi-omics analysis revealed activation of cytoskeletal organization and glycolysis/gluconeogenesis, as well as suppression of gas transport and adaptive immune responses, in COVID-19 convalescents. Additionally, metabolites involved in glutathione metabolism; alanine, aspartate, and glutamate metabolism; and ascorbate and aldarate metabolism were significantly upregulated in COVID-19 convalescents. Pulmonary and molecular abnormalities persisted for 3 years in COVID-19 convalescents; impaired diffusing capacity for carbon monoxide (DLco) was the most prominent feature. We used this multi-omics profile to develop a model involving one protein (heterogeneous nuclear ribonucleoprotein K (HNRNPK)) and two metabolites (arachidonoyl-EA and 1-O-(2r-hydroxy-pentadecyl)-sn-glycerol)) for identification of COVID-19 convalescents with abnormal DLco. CONCLUSIONS These data provide insights concerning molecular sequelae among COVID-19 convalescents up to 3 years after hospital discharge, clarify mechanisms driving respiratory sequelae, and support the development of a novel model to predict reduced DLco. This longitudinal multi-omics analysis may illuminate the trajectory of altered lung function in COVID-19 convalescents.
Collapse
Affiliation(s)
- Huqin Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lujia Guan
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yi Xue
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuyan Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Leyi Gao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhijin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Haifan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Haomiao Ma
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Fengjiao Liu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuan Huang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China.
| | - Jieqiong Li
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
24
|
Patel SH, Joseph JJ, Gandhi TR, Mehta A, Shah A. A Review of Emerging Evidence and Clinical Applications of Hyperbaric Oxygen Therapy. J Intensive Care Med 2025; 40:341-351. [PMID: 39814353 DOI: 10.1177/08850666241313136] [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] [Indexed: 01/18/2025]
Abstract
Background: Hyperbaric Oxygen Therapy (HBOT) is a medical treatment that involves administering 100% oxygen at increased atmospheric pressure to enhance oxygen delivery to tissues. Initially developed for decompression sickness, HBOT has since been utilized for a wide range of medical conditions, including severe infections, non-healing wounds, and, more recently, COVID-19. Objective: This review explores the historical development of HBOT, its principles, its emerging role in the management of and its outcome as treatment in COVID-19, particularly in mitigating inflammation, hypoxemia, and oxidative stress. Methods: A comprehensive review of the literature was conducted, analyzing case reports and case series that examined the effectiveness of HBOT in various clinical scenarios, with a focus on COVID-19. Results: HBOT has been shown to enhance tissue oxygenation, reduce inflammation, and modulate oxidative stress, thereby improving clinical outcomes in patients with severe COVID-19. The therapy's ability to increase dissolved oxygen levels in blood and tissues, independent of hemoglobin, makes it particularly beneficial in conditions like COVID-19, where hypoxemia and inflammation are prominent. Conclusion: HBOT offers a promising adjunctive treatment for severe COVID-19, with the potential to reduce mortality and improve recovery by targeting key pathophysiological processes such as hypoxemia, inflammation, and oxidative stress. Further research is warranted to optimize treatment protocols and confirm long-term benefits.
Collapse
Affiliation(s)
| | | | | | - Anita Mehta
- Anand Pharmacy College, Anand, Gujarat, India
| | - Akshay Shah
- Anand Pharmacy College, Anand, Gujarat, India
| |
Collapse
|
25
|
Jiang Y, Xu L, Zheng X, Shi H. Recent advances in nutritional metabolism studies on SARS-CoV-2 infection. INFECTIOUS MEDICINE 2025; 4:100162. [PMID: 39936106 PMCID: PMC11810712 DOI: 10.1016/j.imj.2025.100162] [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: 09/27/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025]
Abstract
In the context of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), metabolic research has become crucial for in-depth exploration of viral infection mechanisms and in searching for therapeutic strategies. This paper summarizes the interrelationships between carbohydrate, lipid, and amino acid metabolism and COVID-19 infection, discussing their roles in infection progression. SARS-CoV-2 infection leads to insulin resistance and increased glycolysis, reducing glucose utilization and shifting metabolism to use fat as an energy source. Fat is crucial for viral replication, and imbalances in amino acid metabolism may interfere with immune regulation. Consequently, metabolic changes such as hyperglycemia, hypolipidemia, and deficiency of certain amino acids following SARS-CoV-2 infection can contribute to progression toward severe conditions. These metabolic pathways not only have potential value in prediction and diagnosis but also provide new perspectives for the development of therapeutic strategies. By monitoring metabolic changes, infection severity can be predicted early, and modulating these metabolic pathways may help reduce inflammatory responses, improve immune responses, and reduce the risk of thrombosis. Research on the relationship between metabolism and SARS-CoV-2 infection provides an important scientific basis for addressing the global challenge posed by COVID-19, however, further studies are needed to validate these findings and provide more effective strategies for disease control.
Collapse
Affiliation(s)
- Yufen Jiang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Linle Xu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Xuexing Zheng
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Hongbo Shi
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
26
|
Song C, Li Q, Zhang J, Hu W. Uridine Phosphorylase 1 as a Biomarker Associated with Glycolysis in Acute Lung Injury. Inflammation 2025:10.1007/s10753-025-02270-z. [PMID: 39969741 DOI: 10.1007/s10753-025-02270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The specific pathogenesis of acute lung injury (ALI) is complex and not yet clear, and the clinical treatment methods are relatively limited. It is of great clinical significance to explore its pathogenesis and effective molecular targets. Here, we identified an ALI biomarker (UPP1) associated with uridine metabolism by a systematic bioinformatics approach. It was also confirmed to be associated with the glycolytic pathway in the mouse ALI model. In addition, drug sensitivity analysis based on the CMAP database identified three UPP1-associated drugs (CAY10585, XL147 and IOX2) that may be useful in the treatment of ALI. Molecular docking and molecular dynamics simulations further confirmed the stability of the binding between UPP1 and the three drugs. In conclusion, this study confirms that the uridine metabolism gene UPP1 associated with glycolysis is a key biomarker of ALI and provides valuable insights into the potential application of CAY10585, XL147 and IOX2 in ALI.
Collapse
Affiliation(s)
- Congkuan Song
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
- Hubei Provincial Clinical Research Center for Cancer, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China
| | - Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jinjin Zhang
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wusheng Road, Wuhan, China
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
- Hubei Provincial Clinical Research Center for Cancer, No.169 Donghu Road, Wuchang District, Wuhan, 430071, China.
| |
Collapse
|
27
|
Zhou Y, Yang Z, Zhang S, Zhang D, Luo H, Zhu D, Li G, Yang M, Hu X, Qian G, Li G, Wang L, Li S, Yu Z, Ren Z. A multicenter, real-world cohort study: effectiveness and safety of Azvudine in hospitalized COVID-19 patients with pre-existing diabetes. Front Endocrinol (Lausanne) 2025; 16:1467303. [PMID: 40046873 PMCID: PMC11879813 DOI: 10.3389/fendo.2025.1467303] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/21/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction During the Omicron infection wave, diabetic patients are susceptible to COVID-19, which is linked to a poor prognosis. However, research on the real-world effectiveness and safety of Azvudine, a common medication for COVID-19, is insufficient in those with pre-existing diabetes. Methods In this retrospective study, we included 32,864 hospitalized COVID-19 patients from 9 hospitals in Henan Province. Diabetic patients were screened and divided into the Azvudine group and the control group, via 1:1 propensity score matching. The primary outcome was all-cause mortality, and the secondary outcome was composite disease progression. Laboratory abnormal results were used for safety evaluation. Results A total of 1,417 patients receiving Azvudine and 1,417 patients receiving standard treatment were ultimately included. Kaplan-Meier curves suggested that all-cause mortality (P = 0.0026) was significantly lower in the Azvudine group than in the control group, but composite disease progression did not significantly differ (P = 0.1). Cox regression models revealed Azvudine treatment could reduce 26% risk of all-cause mortality (95% CI: 0.583-0.942, P = 0.015) versus controls, and not reduce the risk of composite disease progression (HR: 0.91, 95% CI: 0.750-1.109, P = 0.355). The results of subgroup analysis and three sensitivity analyses were consistent with the previous findings. Safety analysis revealed that the incidence rates of most adverse events were similar between the two groups. Conclusion In this study, Azvudine demonstrated good efficacy in COVID-19 patients with diabetes, with a lower all-cause mortality rate. Additionally, the safety was favorable. This study may provide a new strategy for the antiviral management of COVID-19 patients with diabetes.
Collapse
Affiliation(s)
- Yongjian Zhou
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zecheng Yang
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shixi Zhang
- Department of Infectious Diseases, Shangqiu Municipal Hospital, Shangqiu, China
| | - Donghua Zhang
- Department of Infectious Diseases, Anyang City Fifth People’s Hospital, Anyang, China
| | - Hong Luo
- Guangshan County People’s Hospital, Xinyang, China
| | - Di Zhu
- Radiology Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangming Li
- Department of Liver Disease, the Affiliated Infectious Disease Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengzhao Yang
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaobo Hu
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowu Qian
- Department of Gastrointestinal Surgery, Nanyang Central Hospital, Nanyang, China
| | - Guotao Li
- Department of Infectious Diseases, Luoyang Central Hospital Affiliated of Zhengzhou University, Luoyang, China
| | - Ling Wang
- Department of Clinical Laboratory, Henan Provincial Chest Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Silin Li
- Department of Respiratory and Critical Care Medicine, Fengqiu County People’s Hospital, Xinxiang, China
| | - Zujiang Yu
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Ren
- Department of Infectious Diseases, State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Ribeiro GJS, Pinto ADA, Souza GC, Moriguchi EH. Association between pre-existing cardiovascular risk factors and post-acute sequelae of COVID-19 in older adults. An Sist Sanit Navar 2025; 48:e1103. [PMID: 39949251 PMCID: PMC11925477 DOI: 10.23938/assn.1103] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 03/22/2025]
Abstract
BACKGROUND The long-term health impacts of COVID-19, including post-acute sequelae of SARS-CoV-2, remain insufficiently explored, especially concerning pre-existing cardiovascular risk factors in older adults. This study examines the association between these risk factors and post-acute sequelae of SARS-CoV-2 in this population. METHODS A retrospective study of Brazilian adults aged = 60 years assessed the persistence of post-acute sequelae of SARS-CoV-2 three months after infection in 2020. Cardiovascular risk factors (obesity, smoking, high blood pressure, diabetes mellitus, hypercholesterolemia, and chronic kidney disease) were analyzed in relation to sequelae and adjusting for sociodemographic variables. Data were obtained from the Department of Epidemiological Surveillance in Roraima, Brazil. RESULTS Of the 1,322 participants (55% female; mean age 70.4 years, SD = 7.87), 61.7% (95% CI: 59.1-63.9) reported at least one post-acute sequelae of SARS-CoV-2 at the three-month follow-up. The likelihood of post-acute sequelae of SARS-CoV-2 was significantly higher in participants with diabetes mellitus (OR = 4.39; 95% CI: 3.42-5.66), tobacco use (OR = 3.93; 95% CI: 2.47-6.23), hypertension (OR = 3.62; 95% CI: 2.73-4.78), or hypercholesterolemia (OR = 3.58; 95% CI: 2.80-4.59). Chronic kidney disease (OR = 2.28; 95% CI: 1.59-3.25) and obesity (OR = 1.83; 95% CI: 1.28-2.61) were less strongly associated. CONCLUSIONS Pre-existing cardiovascular risk factors are linked to a higher likelihood of long-term COVID-19 sequelae in adults aged = 60 years old. Preventing and managing these factors are crucial for reducing the long-term effects of COVID-19, particularly during a pandemic.
Collapse
Affiliation(s)
| | | | - Gabriela Corrêa Souza
- Federal University of Rio Grande do Sul. School of Medicine. Department of Nutrition and Graduate Program in Food.
| | - Emilio Hideyuki Moriguchi
- Federal University of Rio Grande do Sul. School of Medicine. Graduate Program in Cardiology and Cardiovascular Sciences. Porto Alegre. Brazil .
| |
Collapse
|
29
|
van der Mescht MA, Steel HC, Anderson R, Rossouw TM. Vascular endothelial growth factor A: friend or foe in the pathogenesis of HIV and SARS-CoV-2 infections? Front Cell Infect Microbiol 2025; 14:1458195. [PMID: 40008234 PMCID: PMC11850333 DOI: 10.3389/fcimb.2024.1458195] [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: 07/02/2024] [Accepted: 10/28/2024] [Indexed: 02/27/2025] Open
Abstract
This review article discusses the role of vascular endothelial growth factor A (VEGF-A) in the pathogenesis of SARS-CoV-2 and HIV infection, both conditions being renowned for their impact on the vascular endothelium. The processes involved in vascular homeostasis and angiogenesis are reviewed briefly before exploring the interplay between hypoxia, VEGF-A, neuropilin-1 (NRP-1), and inflammatory pathways. We then focus on SARS-CoV-2 infection and show how the binding of the viral pathogen to the angiotensin-converting enzyme 2 receptor, as well as to NRP-1, leads to elevated levels of VEGF-A and consequences such as coagulation, vascular dysfunction, and inflammation. HIV infection augments angiogenesis via several mechanisms, most prominently, by the trans-activator of transcription (tat) protein mimicking VEGF-A by binding to its receptor, VEGFR-2, as well as upregulation of NRP-1, which enhances the interaction between VEGF-A and VEGFR-2. We propose that the elevated levels of VEGF-A observed during HIV/SARS-CoV-2 co-infection originate predominantly from activated immune cells due to the upregulation of HIF-1α by damaged endothelial cells. In this context, a few clinical trials have described a diminished requirement for oxygen therapy during anti-VEGF treatment of SARS-CoV-2 infection. The currently available anti-VEGF therapy strategies target the binding of VEGF-A to both VEGFR-1 and VEGFR-2. The blocking of both receptors could, however, lead to a negative outcome, inhibiting not only pathological, but also physiological angiogenesis. Based on the examination of published studies, this review suggests that treatment targeting selective inhibition of VEGFR-1 may be beneficial in the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | | | - Theresa M. Rossouw
- Department of Immunology, Faculty of Health Sciences, University of
Pretoria, Pretoria, South Africa
| |
Collapse
|
30
|
Shabestari M, Salari F, Azizi R, Ghadiri-Anari A, Namiranian N. Impact of COVID-19 on metabolic parameters in patients with type 2 diabetes mellitus. BMC Pulm Med 2025; 25:58. [PMID: 39901215 PMCID: PMC11789312 DOI: 10.1186/s12890-025-03529-9] [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: 10/23/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND AND AIM The Coronavirus Disease 2019 (COVID-19) pandemic has disproportionately affected individuals with Type 2 Diabetes Mellitus (T2DM), making them more susceptible to viral infections. Additionally, COVID-19 and the associated lockdown restrictions have influenced metabolic regulatory mechanisms in this population. This study aims to evaluate the impact of COVID-19 infection and lockdown measures on physiological parameters in individuals with T2DM. METHODS This retrospective cohort study included 118 individuals with a prior diagnosis of T2DM. Medical records were reviewed for laboratory tests conducted within three months before the onset of the COVID-19 pandemic in Iran. Fifty-nine patients with confirmed COVID-19 infection during the first three months of the pandemic underwent follow-up laboratory tests six months post-diagnosis. An age- and gender-matched group of 59 noninfected individuals underwent follow-up tests six months after the pandemic's onset. Clinical and laboratory parameters were analyzed and compared within each group. RESULTS In the COVID-19 positive group, significant reductions were observed in triglycerides (TG) (P = 0.001), total cholesterol (TC) (P = 0.028), body mass index (BMI) (P = 0.034), atherogenic index of plasma (AIP) (P = 0.027), triglyceride-glucose (TyG) index (P = 0.001), and triglyceride-glucose-BMI (TyG-BMI) index (P < 0.001) during the six months following infection compared to pre-pandemic levels. Other variables remained unchanged. In the COVID-19 negative group, significant reductions were noted in TC (P = 0.001) and low-density lipoprotein cholesterol (LDL-C) (P = 0.01). CONCLUSION T2DM patients with mild to moderate COVID-19 infection exhibited improvements in TC, TG, BMI, and insulin-related indices. Lockdown restrictions were associated with decreased TC and LDL-C levels in T2DM patients without a history of COVID-19 infection.
Collapse
Affiliation(s)
- Motahare Shabestari
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Reyhaneh Azizi
- Diabetic Research Center, Shahid Sadoughi University of Medical Sciences, Shahid Sadoughi Blvd, Yazd, 8917693571, Iran
| | - Akram Ghadiri-Anari
- Diabetic Research Center, Shahid Sadoughi University of Medical Sciences, Shahid Sadoughi Blvd, Yazd, 8917693571, Iran.
| | - Nasim Namiranian
- Diabetic Research Center, Shahid Sadoughi University of Medical Sciences, Shahid Sadoughi Blvd, Yazd, 8917693571, Iran
| |
Collapse
|
31
|
Antunes ASLM, Reis-de-Oliveira G, Martins-de-Souza D. Molecular overlaps of neurological manifestations of COVID-19 and schizophrenia from a proteomic perspective. Eur Arch Psychiatry Clin Neurosci 2025; 275:109-122. [PMID: 39028452 DOI: 10.1007/s00406-024-01842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024]
Abstract
COVID-19, a complex multisystem disorder affecting the central nervous system, can also have psychiatric sequelae. In addition, clinical evidence indicates that a diagnosis of a schizophrenia spectrum disorder is a risk factor for mortality in patients with COVID-19. In this study, we aimed to explore brain-specific molecular aspects of COVID-19 by using a proteomic approach. We analyzed the brain proteome of fatal COVID-19 cases and compared it with differentially regulated proteins found in postmortem schizophrenia brains. The COVID-19 proteomic dataset revealed a strong enrichment of proteins expressed by glial and neuronal cells and processes related to diseases with a psychiatric and neurodegenerative component. Specifically, the COVID-19 brain proteome enriches processes that are hallmark features of schizophrenia. Furthermore, we identified shared and distinct molecular pathways affected in both conditions. We found that brain ageing processes are likely present in both COVID-19 and schizophrenia, albeit possibly driven by distinct processes. In addition, alterations in brain cell metabolism were observed, with schizophrenia primarily impacting amino acid metabolism and COVID-19 predominantly affecting carbohydrate metabolism. The enrichment of metabolic pathways associated with astrocytic components in both conditions suggests the involvement of this cell type in the pathogenesis. Both COVID-19 and schizophrenia influenced neurotransmitter systems, but with distinct impacts. Future studies exploring the underlying mechanisms linking brain ageing and metabolic dysregulation may provide valuable insights into the complex pathophysiology of these conditions and the increased vulnerability of schizophrenia patients to severe outcomes.
Collapse
Affiliation(s)
- André S L M Antunes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | | | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, University of Campinas, Campinas, Brazil.
- D'or Institute for Research and Education, São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), Estate University of Campinas, Campinas, Brazil.
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil.
| |
Collapse
|
32
|
Park SG, Lee JY, Seo H, Hwang SS, Lee CK, Lee GR. Modulation of Immune Responses by Metabolic Reprogramming: The Key Role of Immunometabolism. Immune Netw 2025; 25:e15. [PMID: 40078790 PMCID: PMC11896660 DOI: 10.4110/in.2025.25.e15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Affiliation(s)
- Sung-Gyoo Park
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hyungseok Seo
- Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Korea
| | - Soo Seok Hwang
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Chong-Kil Lee
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Gap Ryol Lee
- Department of Life Science, Sogang University, Seoul 04107, Korea
| |
Collapse
|
33
|
Maison DP, Tasissa H, Deitchman A, Peluso MJ, Deng Y, Miller FD, Henrich TJ, Gerschenson M. COVID-19 clinical presentation, management, and epidemiology: a concise compendium. Front Public Health 2025; 13:1498445. [PMID: 39957982 PMCID: PMC11826932 DOI: 10.3389/fpubh.2025.1498445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Coronavirus Disease 2019, caused by severe acute respiratory coronavirus 2, has been an ever-evolving disease and pandemic, profoundly impacting clinical care, drug treatments, and understanding. In response to this global health crisis, there has been an unprecedented increase in research exploring new and repurposed drugs and advancing available clinical interventions and treatments. Given the widespread interest in this topic, this review aims to provide a current summary-for interested professionals not specializing in COVID-19-of the clinical characteristics, recommended treatments, vaccines, prevention strategies, and epidemiology of COVID-19. The review also offers a historical perspective on the pandemic to enhance understanding.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hawi Tasissa
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - F. DeWolfe Miller
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
34
|
Li Y, Li G, Li J, Luo Z, Lin Y, Lan N, Zhang X. Correlation of diabetes and adverse outcomes in hospitalized COVID-19 patients admitted to a tertiary hospital in China during a small-scale COVID-19 outbreak. PeerJ 2025; 13:e18865. [PMID: 39886017 PMCID: PMC11781264 DOI: 10.7717/peerj.18865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
Background The aim of this study was to investigate the impact of diabetes on mortality and adverse outcomes in COVID-19 patients and to analyse the associated risk factors. Methods This is a retrospective cohort study in 500 hospitalized patients with COVID-19 infection (214 with diabetes and 286 without diabetes) admitted to a tertiary hospital in China from December 2022 to February 2023. Demographic information, clinical characteristics and outcomes were collected. Survival status was investigated at discharge and at 6 months after discharge. Results The mortality rate of COVID-19 patients with diabetes was higher than the rate of non-diabetic COVID-19 patients, both at discharge, and at 6 months after discharge. Body mass index (BMI), C-reactive protein (CRP), pH, D-dimer, blood osmotic pressure, serum creatinine, white blood cell count, creatine kinase and hospitalization expenses were significantly different between diabetic group and non-diabetic group (p < 0.05). Compared with the survivors, non-survived COVID-19 patients with diabetes had worse diabetes control indicators, with random blood glucose increased by 3.58 mmol/L (p < 0.05), and fasting blood glucose increased by 2.77 mmol/L (p < 0.01). In addition, there were significant differences in age, heart rate, CRP, pH, potassium (K+), serum creatinine, white blood cell count, creatine kinase, the proportion with diabetic complications, treatment in ICU and mechanical ventilation between survivors and non-survivors of COVID-19 patients with diabetes. By multivariate logistic regression analysis, the death of COVID-19 patients with diabetes is positively correlated with age and CRP (p < 0.05), and has a trend towards significance with fasting blood glucose (p < 0.1). Conclusion Infection with COVID-19 on the basis of diabetes can significantly increase mortality, which was further associated with diabetes control indicators.
Collapse
Affiliation(s)
- Yu Li
- Department of Endocrinology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guanni Li
- Department of Endocrinology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiahong Li
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Zirui Luo
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yaxuan Lin
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Ning Lan
- Department of Endocrinology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaodan Zhang
- Department of Endocrinology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
35
|
Lu Y, Wang Y, Ruan T, Wang Y, Ju L, Zhou M, Liu L, Yao D, Yao M. Immunometabolism of Tregs: mechanisms, adaptability, and therapeutic implications in diseases. Front Immunol 2025; 16:1536020. [PMID: 39917294 PMCID: PMC11798928 DOI: 10.3389/fimmu.2025.1536020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Immunometabolism is an emerging field that explores the intricate interplay between immune cells and metabolism. Regulatory T cells (Tregs), which maintain immune homeostasis in immunometabolism, play crucial regulatory roles. The activation, differentiation, and function of Tregs are influenced by various metabolic pathways, such as the Mammalian targets of rapamycin (mTOR) pathway and glycolysis. Correspondingly, activated Tregs can reciprocally impact these metabolic pathways. Tregs also possess robust adaptive capabilities, thus enabling them to adapt to various microenvironments, including the tumor microenvironment (TME). The complex mechanisms of Tregs in metabolic diseases are intriguing, particularly in conditions like MASLD, where Tregs are significantly upregulated and contribute to fibrosis, while in diabetes, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA), they show downregulation and reduced anti-inflammatory capacity. These phenomena suggest that the differentiation and function of Tregs are influenced by the metabolic environment, and imbalances in either can lead to the development of metabolic diseases. Thus, moderate differentiation and inhibitory capacity of Tregs are critical for maintaining immune system balance. Given the unique immunoregulatory abilities of Tregs, the development of targeted therapeutic drugs may position them as novel targets in immunotherapy. This could contribute to restoring immune system balance, resolving metabolic dysregulation, and fostering innovation and progress in immunotherapy.
Collapse
|
36
|
Li M, Yuan H, Yang X, Lei Y, Lian J. Glutamine-glutamate centered metabolism as the potential therapeutic target against Japanese encephalitis virus-induced encephalitis. Cell Biosci 2025; 15:6. [PMID: 39844330 PMCID: PMC11755858 DOI: 10.1186/s13578-024-01340-3] [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: 08/01/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Japanese encephalitis (JE) induced by Japanese encephalitis virus (JEV) infection is the most prevalent diagnosed epidemic viral encephalitis globally. The underlying pathological mechanisms remain largely unknown. Given that viruses are obligate intracellular parasites, cellular metabolic reprogramming triggered by viral infection is intricately related to the establishment of infection and progression of disease. Therefore, uncovering and manipulating the metabolic reprogramming that underlies viral infection will help elucidate the pathogenic mechanisms and develop novel therapeutic strategies. METHODS Metabolomics analysis was performed to comprehensively delineate the metabolic profiles in JEV-infected mice brains and neurons. Metabolic flux analysis, quantitative real-time PCR, western blotting and fluorescence immunohistochemistry were utilized to describe detailed glutamine-glutamate metabolic profiles during JEV infection. Exogenous addition of metabolites and associated compounds and RNA interference were employed to manipulate glutamine-glutamate metabolism to clarify its effects on viral replication. The survival rate, severity of neuroinflammation, and levels of viral replication were assessed to determine the efficacy of glutamine supplementation in JEV-challenged mice. RESULTS Here, we have delineated a novel perspective on the pathogenesis of JE by identifying an aberrant low flux in glutamine-glutamate metabolism both in vivo and in vitro, which was critical in the establishment of JEV infection and progression of JE. The perturbed glutamine-glutamate metabolism induced neurotransmitter imbalance and created an immune-inhibitory state with increased gamma-aminobutyric acid/glutamate ratio, thus facilitating efficient viral replication both in JEV-infected neurons and the brain of JEV-infected mice. In addition, viral infection restrained the utilization of glutamine via the glutamate-α-ketoglutaric acid axis in neurons, thus avoiding the adverse effects of glutamine oxidation on viral propagation. As the conversion of glutamine to glutamate was inhibited after JEV infection, the metabolism of glutathione (GSH) was simultaneously impaired, exacerbating oxidative stress in JEV-infected neurons and mice brains and promoting the progression of JE. Importantly, the supplementation of glutamine in vivo alleviated the intracranial inflammation and enhanced the survival of JEV-challenged mice. CONCLUSION Altogether, our study highlights an aberrant glutamine-glutamate metabolism during JEV infection and unveils how this facilitates viral replication and promotes JE progression. Manipulation of these metabolic alterations may potentially be exploited to develop therapeutic approaches for JEV infection.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Hang Yuan
- Pathogenic Biology, Medical College of Yan'an University, Yan'an, 716000, China
| | - Xiaofei Yang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yingfeng Lei
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China.
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
37
|
Adha SA, Afifah NN, Latarissa IR, Iftinan GN, Kusuma ASW, Febriyanti RM, Barliana MI, Lestari K. Herbal Medicines as Complementary Therapy for Managing Complications in COVID-19 Patients with Diabetes Mellitus. Diabetes Metab Syndr Obes 2025; 18:135-146. [PMID: 39840393 PMCID: PMC11746946 DOI: 10.2147/dmso.s498774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/26/2024] [Indexed: 01/23/2025] Open
Abstract
Diabetes mellitus (DM) is recognized and classified as a group of conditions marked by persistent high blood glucose levels. It is also an inflammatory condition that may influence concurrent disease states, including Coronavirus Disease 2019 (COVID-19). Currently, no effective drug has been found to treat COVID-19, especially in DM patients. Many herbal medicines, such as the well-known Andrographis paniculata, have been explored as drugs and complementary therapies due to their antidiabetic, antibacterial, antiviral, anti-inflammatory, and immunomodulatory effects. This study aimed to examine the potential of herbal medicines as complementary therapy in DM patients with COVID-19 complications, drawing from in-vitro and in-vivo investigations. This study analyzed articles published within the last 15 years using keywords including "herbal medicines", "COVID-19", "Diabetes Mellitus", "antidiabetics", "antiviral", and "anti-inflammatory". The results showed that several herbal medicines could serve as complementary therapy for DM patients with COVID-19 complications. These include Andrographis paniculata, Ageratum conyzoides, Artocarpus altilis, Centella asiatica, Momordica charantia, Persea gratissima, Phyllanthus urinaria, Physalis angulata, Tinospora cordifolia, and Zingiber zerumbet. Herbal medicines may serve as a complementary therapy for DM patients with COVID-19, but these claims need experimental validation in infection models and among affected patients.
Collapse
Affiliation(s)
- Syah Akbarul Adha
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Nadiya Nurul Afifah
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Irma Rahayu Latarissa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Ghina Nadhifah Iftinan
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Arif Satria Wira Kusuma
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Raden Maya Febriyanti
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Keri Lestari
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
38
|
Sánchez‐García S, Povo‐Retana A, Marin S, Madurga S, Fariñas M, Aleixandre N, Castrillo A, de la Rosa JV, Alvarez‐Lucena C, Landauro‐Vera R, Prieto P, Cascante M, Boscá L. Immunometabolic Effect of Nitric Oxide on Human Macrophages Challenged With the SARS-CoV2-Induced Cytokine Storm. A Fluxomic Approach. Adv Healthc Mater 2025; 14:e2401688. [PMID: 39502019 PMCID: PMC11694080 DOI: 10.1002/adhm.202401688] [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: 05/25/2024] [Revised: 10/04/2024] [Indexed: 01/03/2025]
Abstract
The cytokine storm associated with SARS-CoV-2 infection is one of the most distinctive pathological signatures in COVID-19 patients. Macrophages respond to this pro-inflammatory challenge by reprogramming their functional and metabolic phenotypes. Interestingly, human macrophages fail to express the inducible form of the NO synthase (NOS2) in response to pro-inflammatory activation and, therefore, NO is not synthesized by these cells. The contribution of exogenously added NO, via a chemical NO-donor, on the immunometabolic changes associated with the cytokine storm is investigated. By using metabolic, transcriptomic, and functional assays the effect of NO in human macrophages is evaluated and found specific responses. Moreover, through integrative fluxomic analysis, pathways modified by NO that contribute to the expression of a particular phenotype in human macrophages are identified, which includes a decrease in mitochondrial respiration and TCA with a slight increase in the glycolytic flux. A significant ROS increase and preserved cell viability are observed in the presence of NO, which may ease the inflammatory response and host defense. Also, NO reverses the cytokine storm-induced itaconate accumulation. These changes offer additional clues to understanding the potential crosstalk between NO and the COVID-19 cytokine storm-dependent signaling pathways.
Collapse
Affiliation(s)
- Sergio Sánchez‐García
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Adrián Povo‐Retana
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Sergio Madurga
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Marco Fariñas
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
| | - Nuria Aleixandre
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Department of Material Science and Physical Chemistry & Research Institute of Theoretical and Computational Chemistry (IQTCUB)University of BarcelonaBarcelona08028Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Juan V. de la Rosa
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| | - Carlota Alvarez‐Lucena
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Rodrigo Landauro‐Vera
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Patricia Prieto
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Departamento de Farmacología, Farmacognosia y BotánicaFacultad de Farmacia, Universidad Complutense de MadridMadrid28040Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine‐Institute of Biomedicine (IBUB), Faculty of BiologyUniversitat de BarcelonaBarcelona08028Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols‐Morreale, Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de MadridArturo Duperier 4Madrid28029Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)Av. Monforte de Lemos 3–5, P‐11Madrid28029Spain
- Unidad de Biomedicina (Unidad Asociada al CSIC) de la Universidad de Las Palmas de Gran CanariaLas Palmas35016Spain
| |
Collapse
|
39
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
40
|
de Oliveira EC, Tibúrcio R, Duarte G, Lago A, de Melo L, Nunes S, Davanzo GG, Martins AJ, Ribeiro BV, Mothé D, Menezes JBP, Veras P, Tavares N, Moraes-Vieira PM, Brodskyn CI. Pharmacological inhibition of key metabolic pathways attenuates Leishmania spp infection in macrophages. PLoS Negl Trop Dis 2025; 19:e0012763. [PMID: 39775223 PMCID: PMC11756801 DOI: 10.1371/journal.pntd.0012763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 01/23/2025] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Macrophages represent a fundamental component of the innate immune system that play a critical role in detecting and responding to pathogens as well as danger signals. Leishmania spp. infections lead to a notable alteration in macrophage metabolism, whereby infected cells display heightened energy metabolism that is linked to the integrity of host mitochondria. However, little is known about how different species of Leishmania manipulate host metabolism. Here, we demonstrate that despite differences in their mechanisms for evading host immune responses, L. amazonensis and L. braziliensis induce comparable disruptions in key metabolic pathways. We found that infected macrophages exhibited an overall elevation in energy metabolism regardless of the parasite strain, evidenced by the elevation in glycolysis and oxygen consumption rates, along with increased proton leak and decreased ATP production. We also analyzed the effects of both Leishmania spp. strain infection on mitochondria function, further revealing that infected cells display heightened mitochondrial mass and membrane potential. To investigate the metabolic pathways required for Leishmania amastigotes to persist in BMDMs, we pre-treated cells with small molecule drugs that target major metabolic pathways, revealing that perturbations in several metabolic processes affected parasite survival in a strain-independent manner. Treatments with inhibitors of the oxidative phosphorylation and glycolysis substantially reduced parasite loads. Collectively, our findings suggest that L.amazonensis and L.braziliensis exploit host cell metabolic pathways similarly to survive in macrophages.
Collapse
Affiliation(s)
| | - Rafael Tibúrcio
- University of California San Francisco, Department of Medicine, San Francisco, California, United States
| | - Gabriela Duarte
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Amanda Lago
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Léon de Melo
- University of Calgary, Departments of Microbiology, Immunology and Infectious Diseases, Calgary, Alberta, Canada
| | - Sara Nunes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Gustavo Gastão Davanzo
- University of Campinas, Department of Genetics, Evolution, Microbiology and Immunology, São Paulo, Brazil
| | - Ana Júlia Martins
- University of Campinas, Department of Genetics, Evolution, Microbiology and Immunology, São Paulo, Brazil
| | | | - Deborah Mothé
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | | - Patrícia Veras
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Natalia Tavares
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Pedro M. Moraes-Vieira
- University of Campinas, Department of Genetics, Evolution, Microbiology and Immunology, São Paulo, Brazil
| | | |
Collapse
|
41
|
Li X, Wu Y, Zhang M, Wang F, Yin H, Zhang Y, Zhao S, Ma J, Lv M, Lu C. A new peptide inhibitor of C1QBP exhibits potent anti-tumour activity against triple negative breast cancer by impairing mitochondrial function and suppressing homologous recombination repair. Clin Transl Med 2025; 15:e70162. [PMID: 39748215 PMCID: PMC11695203 DOI: 10.1002/ctm2.70162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
C1QBP exhibits heightened expression across a spectrum of tumours, thereby fostering their proliferation and metastasis, rendering it a pivotal therapeutic target. Nevertheless, to date, no pharmacological agents capable of directly targeting and inducing the degradation of C1QBP have been identified. In this study, we have unveiled a new peptide, PDBAG1, derived from the precursor protein GPD1, employing a peptidomics-based drug screening strategy. PDBAG1 has demonstrated substantial efficacy in suppressing triple-negative breast cancer (TNBC) both in vitro and in vivo. Its mechanism of action involves mitochondrial impairment and the inhibition of oxidative phosphorylation (OXPHOS), achieved through direct binding to C1QBP, thereby promoting its ubiquitin-dependent degradation. Concomitantly, due to metabolic adaptability, we have observed an up-regulation of glycolysis to compensate for OXPHOS inhibition. We observed an aberrant phenomenon wherein the hypoxia signalling pathway in tumour cells exhibited significant activation under normoxic conditions following PDBAG1 treatment. Through size-exclusion chromatography (SEC) and isothermal titration calorimetry (ITC) assays, we have validated that PDBAG1 is capable of binding C1QBP with a Kd value of 334 nM. Furthermore, PDBAG1 inhibits homologous recombination repair proteins and facilitates synergism with poly-ADP-ribose polymerase inhibitors in cancer therapy. This underscores that PDBAG1 ultimately induces insurmountable survival stress through multiple mechanisms while concurrently engendering therapeutic vulnerabilities specific to TNBC. KEY POINTS: The newly discovered peptide PDBAG1 is the first small molecule substance found to directly target and degrade C1QBP, demonstrating significant tumour inhibitory effects and therapeutic potential.
Collapse
Affiliation(s)
- Xingxing Li
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Yue Wu
- The State Key Laboratory of Pharmaceutical BiotechnologyDivision of ImmunologyMedical SchoolNanjing UniversityNanjingChina
| | - Min Zhang
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Fengliang Wang
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Hong Yin
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Yanrong Zhang
- Nanjing Women and Children's Healthcare InstituteWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Shuli Zhao
- General Clinical Research CenterNanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jiehua Ma
- Nanjing Women and Children's Healthcare InstituteWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Mingming Lv
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
- Nanjing Women and Children's Healthcare InstituteWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| | - Cheng Lu
- Department of BreastWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjingChina
| |
Collapse
|
42
|
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
|
43
|
Yang QW, Yue CL, Chen M, Ling YY, Dong Q, Zhou YX, Cao Y, Ding YX, Zhao X, Huang H, Zhang ZH, Hu L, Xu XH. Daphnetin may protect from SARS-CoV-2 infection by reducing ACE2. Sci Rep 2024; 14:30682. [PMID: 39730426 DOI: 10.1038/s41598-024-79734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/12/2024] [Indexed: 12/29/2024] Open
Abstract
To combat the SARS-CoV-2 pandemic, innovative prevention strategies are needed, including reducing ACE2 expression on respiratory cells. This study screened approved drugs in China for their ability to downregulate ACE2. Daphnetin (DAP) was found to significantly reduce ACE2 mRNA and protein levels in PC9 cells. DAP exerts its inhibitory effects on ACE2 expression by targeting HIF-1α and JAK2, thereby impeding the transcription of the ACE2 gene. The SARS-CoV-2 pseudovirus infection assay confirmed that DAP-treated PC9 cells exhibited decreased susceptibility to viral infection. At therapeutic doses, DAP effectively lowers ACE2 expression in the respiratory systems of mice and humans. This suggests that DAP, already approved for other conditions, could be a new preventive measure against SARS-CoV-2, offering a cost-effective and accessible way to reduce SARS-CoV-2 spread.
Collapse
Affiliation(s)
- Qian-Wen Yang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Chang-Ling Yue
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Meng Chen
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yun-Yun Ling
- Department of Chemistry, Wannan Medical College, Wuhu, 241002, China
| | - Qi Dong
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Ying-Xin Zhou
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yin Cao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yan-Xia Ding
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Xu Zhao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Hai Huang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhao-Huan Zhang
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| | - Xiao-Hui Xu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
44
|
Marin D, Fernandez GJ, Hernandez JC, Taborda N. A systems biology approach unveils different gene expression control mechanisms governing the immune response genetic program in peripheral blood mononuclear cells exposed to SARS-CoV-2. PLoS One 2024; 19:e0314754. [PMID: 39637135 PMCID: PMC11620636 DOI: 10.1371/journal.pone.0314754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024] Open
Abstract
COVID-19 and other pandemic viruses continue being important for public health and the global economy. Therefore, it is essential to explore the pathogenesis of COVID-19 more deeply, particularly its association with inflammatory and antiviral processes. In this study, we used the RNA-seq technique to analyze mRNA and non-coding RNA profiles of human peripheral blood mononuclear cells (PBMCs) from healthy individuals after SARS-CoV-2 in vitro exposure, to identify pathways related to immune response and the regulatory post-transcriptional mechanisms triggered that can serve as possible complementary therapeutic targets. Our analyses show that SARS-CoV-2 induced a significant regulation in the expression of 790 genes in PBMCs, of which 733 correspond to mRNAs and 57 to non-coding RNAs (lncRNAs). The immune response, antiviral response, signaling, cell proliferation and metabolism are the main biological processes involved. Among these, the inflammatory response groups the majority of regulated genes with an increase in the expression of chemokines involved in the recruitment of monocytes, neutrophils and T-cells. Additionally, it was observed that exposure to SARS-CoV-2 induces the expression of genes related to the IL-27 pathway but not of IFN-I or IFN-III, indicating the induction of ISGs through this pathway rather than the IFN genes. Moreover, several lncRNA and RNA binding proteins that can act in the cis-regulation of genes of the IL-27 pathway were identified. Our results indicate that SARS-CoV-2 can regulate the expression of multiple genes in PBMCs, mainly related to the inflammatory and antiviral response. Among these, lncRNAs establish an important mechanism in regulating the immune response to the virus. They could contribute to developing severe forms of COVID-19, constituting a possible therapeutic target.
Collapse
Affiliation(s)
- Damariz Marin
- GIOM, Facultad de Odontología, Universidad Cooperativa de Colombia, Medellín, Colombia
| | - Geysson Javier Fernandez
- Biología y Control de Enfermedades Infecciosas (BCEI), Universidad de Antioquia- UdeA, Medellín, Colombia
| | - Juan C. Hernandez
- Infettare, Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín, Colombia
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia- UdeA, Medellín, Colombia
| | - Natalia Taborda
- Corporación Universitaria Remington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| |
Collapse
|
45
|
Topper MJ, Guarnieri JW, Haltom JA, Chadburn A, Cope H, Frere J, An J, Borczuk A, Sinha S, Kim J, Park J, Butler D, Meydan C, Foox J, Bram Y, Richard SA, Epsi NJ, Agan B, Chenoweth JG, Simons MP, Tribble D, Burgess T, Dalgard C, Heise MT, Moorman NJ, Baxter VK, Madden EA, Taft-Benz SA, Anderson EJ, Sanders WA, Dickmander RJ, Beigel K, Widjaja GA, Janssen KA, Lie T, Murdock DG, Angelin A, Soto Albrecht YE, Olali AZ, Cen Z, Dybas J, Priebe W, Emmett MR, Best SM, Kelsey Johnson M, Trovao NS, Clark KB, Zaksas V, Meller R, Grabham P, Schisler JC, Moraes-Vieira PM, Pollett S, Mason CE, Syrkin Wurtele E, Taylor D, Schwartz RE, Beheshti A, Wallace DC, Baylin SB. Lethal COVID-19 associates with RAAS-induced inflammation for multiple organ damage including mediastinal lymph nodes. Proc Natl Acad Sci U S A 2024; 121:e2401968121. [PMID: 39602262 PMCID: PMC11626201 DOI: 10.1073/pnas.2401968121] [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: 02/15/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
Lethal COVID-19 outcomes are attributed to classic cytokine storm. We revisit this using RNA sequencing of nasopharyngeal and 40 autopsy samples from patients dying of SARS-CoV-2. Subsets of the 100 top-upregulated genes in nasal swabs are upregulated in the heart, lung, kidney, and liver, but not mediastinal lymph nodes. Twenty-two of these are "noncanonical" immune genes, which we link to components of the renin-angiotensin-activation-system that manifest as increased fibrin deposition, leaky vessels, thrombotic tendency, PANoptosis, and mitochondrial dysfunction. Immunohistochemistry of mediastinal lymph nodes reveals altered architecture, excess collagen deposition, and pathogenic fibroblast infiltration. Many of the above findings are paralleled in animal models of SARS-CoV-2 infection and human peripheral blood mononuclear and whole blood samples from individuals with early and later SARS-CoV-2 variants. We then redefine cytokine storm in lethal COVID-19 as driven by upstream immune gene and mitochondrial signaling producing downstream RAAS (renin-angiotensin-aldosterone system) overactivation and organ damage, including compromised mediastinal lymph node function.
Collapse
Affiliation(s)
- Michael J. Topper
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Joseph W. Guarnieri
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Jeffrey A. Haltom
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Amy Chadburn
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY10065
| | - Henry Cope
- School of Medicine, University of Nottingham, DerbyDE22 3DT, United Kingdom
| | - Justin Frere
- Icahn School of Medicine, Mount Sinai, New York, NY10023
| | - Julia An
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | | | | | | | | | | | - Cem Meydan
- Weill Cornell Medicine, New York, NY10065
| | | | - Yaron Bram
- Weill Cornell Medicine, New York, NY10065
| | - Stephanie A. Richard
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Nusrat J. Epsi
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Brian Agan
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Josh G. Chenoweth
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Mark P. Simons
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - David Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - Timothy Burgess
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
| | - Clifton Dalgard
- Department of Anatomy, Physiology & Genetics, Uniformed Services University, Bethesda, MD20814
| | | | | | | | | | | | | | | | | | - Katherine Beigel
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Gabrielle A. Widjaja
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kevin A. Janssen
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Timothy Lie
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Deborah G. Murdock
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Alessia Angelin
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Yentli E. Soto Albrecht
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- The University of Pennsylvania, Philadelphia, PA19104
| | - Arnold Z. Olali
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Zimu Cen
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Joseph Dybas
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Waldemar Priebe
- COVID-19 International Research Team, Medford, MA02155
- University of Texas Monroe Dunaway Anderson Cancer Center, Houston, TX77030
| | - Mark R. Emmett
- COVID-19 International Research Team, Medford, MA02155
- University of Texas Medical Branch, Galveston, TX77555
| | - Sonja M. Best
- COVID-19 International Research Team, Medford, MA02155
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, National Institute of Allergy and Infectious Diseases, NIH, Rocky Mountain Laboratories, Hamilton, MT59840
| | - Maya Kelsey Johnson
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Nidia S. Trovao
- COVID-19 International Research Team, Medford, MA02155
- Fogarty International Center, NIH, Bethesda, MD20892
| | - Kevin B. Clark
- COVID-19 International Research Team, Medford, MA02155
- Cures Within Reach, Chicago, IL60602
- Champions Service, Computational Sciences Support Network, Multi-Tier Assistance, Training, and Computational Help Track, NSF's Advanced Cyberinfrastructure Coordination Ecosystem: Services and Support, Carnegie-Mellon University, Pittsburgh, PA15213
| | - Victoria Zaksas
- COVID-19 International Research Team, Medford, MA02155
- Center for Translational Data Science, University of Chicago, Chicago, IL60615
- Clever Research Lab, Springfield, IL62704
| | - Robert Meller
- COVID-19 International Research Team, Medford, MA02155
- Morehouse School of Medicine, Atlanta, GA30310
| | - Peter Grabham
- COVID-19 International Research Team, Medford, MA02155
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, NY19103
| | - Jonathan C. Schisler
- COVID-19 International Research Team, Medford, MA02155
- University of North Carolina, Chapel Hill, NC27599
| | - Pedro M. Moraes-Vieira
- COVID-19 International Research Team, Medford, MA02155
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil13083-862
| | - Simon Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD20817
| | - Christopher E. Mason
- COVID-19 International Research Team, Medford, MA02155
- Weill Cornell Medicine, New York, NY10065
- New York Genome Center, New York, NY10013
| | - Eve Syrkin Wurtele
- COVID-19 International Research Team, Medford, MA02155
- Center for Metabolic Biology, Bioinformatics and Computational Biology, and Genetics Development, and Cell Biology, Iowa State University, Ames, IA50011
- Center for Bioinformatics and Computational Biology Iowa State University, Ames, IA50011
- Center for Genetics Development, and Cell Biology Iowa State University, Ames, IA50011
| | - Deanne Taylor
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
| | - Robert E. Schwartz
- COVID-19 International Research Team, Medford, MA02155
- Weill Cornell Medicine, New York, NY10065
| | - Afshin Beheshti
- COVID-19 International Research Team, Medford, MA02155
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Blue Marble Space Institute of Science, Seattle, WA98104
- McGowan Institute for Regenerative Medicine and Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA15219
| | - Douglas C. Wallace
- COVID-19 International Research Team, Medford, MA02155
- The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Center for Mitochondrial and Epigenomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, University of Pennsylvania, Philadelphia, PA19104
| | - Stephen B. Baylin
- COVID-19 International Research Team, Medford, MA02155
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Van Andel Institute, Grand Rapids, MI49503
| |
Collapse
|
46
|
Mou X, Luo F, Zhang W, Cheng Q, Hepojoki J, Zhu S, Liu Y, Xiong H, Guo D, Yu J, Chen L, Li Y, Hou W, Chen S. SARS-CoV-2 NSP16 promotes IL-6 production by regulating the stabilization of HIF-1α. Cell Signal 2024; 124:111387. [PMID: 39251053 DOI: 10.1016/j.cellsig.2024.111387] [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: 05/05/2024] [Revised: 08/23/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiologic agent of coronavirus disease 2019 (COVID-19). Severe and fatal COVID-19 cases often display cytokine storm i.e. significant elevation of pro-inflammatory cytokines and acute respiratory distress syndrome (ARDS) with systemic hypoxia. Understanding the mechanisms of these pathogenic manifestations would be essential for the prevention and especially treatment of COVID-19 patients. Here, using a dual luciferase reporter assay for hypoxia-response element (HRE), we initially identified SARS-CoV-2 nonstructural protein 5 (NSP5), NSP16, and open reading frame 3a (ORF3a) to upregulate hypoxia-inducible factor-1α (HIF-1α) signaling. Further experiments showed NSP16 to have the most prominent effect on HIF-1α, thus contributing to the induction of COVID-19 associated pro-inflammatory response. We demonstrate that NSP16 interrupts von Hippel-Lindau (VHL) protein interaction with HIF-1α, thereby inhibiting ubiquitin-dependent degradation of HIF-1α and allowing it to bind HRE region in the IL-6 promoter region. Taken together, the findings imply that SARS-CoV-2 NSP16 induces HIF-1α expression, which in turn exacerbates the production of IL-6.
Collapse
Affiliation(s)
- Xiaoli Mou
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Fan Luo
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, 00290 Helsinki, Finland
| | - Weihao Zhang
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Qi Cheng
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, 00290 Helsinki, Finland
| | - Shaowei Zhu
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Yuanyuan Liu
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Hairong Xiong
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China
| | - Deyin Guo
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Jingyou Yu
- Guangzhou Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong 510320, China
| | - Liangjun Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Wei Hou
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; School of Public Health, Wuhan University, Wuhan, Hubei 430071, China; School of Ecology and Environment, Tibet University, Lhasa, Tibet 850000, China; Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong 518057, China.
| | - Shuliang Chen
- State Key Laboratory of Virology, Institute of Medical Virology, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei 430071, China; Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, Hubei 430071, China.
| |
Collapse
|
47
|
Mokry RL, Purdy JG. Glucose-independent human cytomegalovirus replication is supported by metabolites that feed upper glycolytic branches. Proc Natl Acad Sci U S A 2024; 121:e2412966121. [PMID: 39560652 PMCID: PMC11621781 DOI: 10.1073/pnas.2412966121] [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: 06/27/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024] Open
Abstract
Viruses with broad tissue distribution and cell tropism successfully replicate in various nutrient environments in the body. Several viruses reprogram metabolism for viral replication. However, many studies focus on metabolic reprogramming in nutrient-rich conditions that do not recapitulate physiological environments in the body. Here, we investigated how viruses may replicate when a metabolite thought to be essential for replication is limited. We use human cytomegalovirus infection in glucose-free conditions as a model to determine how glucose supports virus replication and how physiologically relevant nutrients contribute to glucose-independent virus production. We find that glucose supports viral genome synthesis, viral protein production and glycosylation, and infectious virus production. Notably, supplement of glucose-free cultures with uridine, ribose, or UDP-GlcNAc-metabolites that feed upper glycolytic branches like the pentose phosphate pathway-results in partially restored virus replication, including low levels of infectious virus production. Supplementing lower glycolysis in glucose-free cultures using pyruvate fails to restore virus replication. These results indicate that nutrients can compensate for glucose via feeding upper glycolytic branches to sustain low levels of virus production. More broadly, our findings suggest that viruses may successfully replicate in diverse metabolic niches, including those in the body with low glucose levels, through alternative nutrient usage.
Collapse
Affiliation(s)
- Rebekah L. Mokry
- BIO5 Institute, University of Arizona, Tucson, AZ85719
- Department of Immunobiology, University of Arizona, Tucson, AZ85724
| | - John G. Purdy
- BIO5 Institute, University of Arizona, Tucson, AZ85719
- Department of Immunobiology, University of Arizona, Tucson, AZ85724
- Cancer Biology Interdisciplinary Program, University of Arizona, Tucson, AZ85724
| |
Collapse
|
48
|
Uchimido R, Kami K, Yamamoto H, Yokoe R, Tsuchiya I, Nukui Y, Goto Y, Hanafusa M, Fujiwara T, Wakabayashi K. Longitudinal Metabolomics Reveals Metabolic Dysregulation Dynamics in Patients with Severe COVID-19. Metabolites 2024; 14:656. [PMID: 39728437 DOI: 10.3390/metabo14120656] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objective: A dysregulated metabolism has been studied as a key aspect of the COVID-19 pathophysiology, but its longitudinal progression in severe cases remains unclear. In this study, we aimed to investigate metabolic dysregulation over time in patients with severe COVID-19 requiring mechanical ventilation (MV). Methods: In this single-center, prospective, observational study, we obtained 236 serum samples from 118 adult patients on MV in an ICU. The metabolite measurements were performed using capillary electrophoresis Fourier transform mass spectrometry, and we categorized the sampling time points into three time zones to align them with the disease progression: time zone 1 (T1) (the hyperacute phase, days 1-3 post-MV initiation), T2 (the acute phase, days 4-14), and T3 (the chronic phase, days 15-30). Using volcano plots and enrichment pathway analyses, we identified the differential metabolites (DMs) and enriched pathways (EPs) between the survivors and non-survivors for each time zone. The DMs and EPs were further grouped into early-stage, late-stage, and consistent groups based on the time zones in which they were detected. Results: With the 566 annotated metabolites, we identified 38 DMs and 17 EPs as the early-stage group, which indicated enhanced energy production in glucose, amino acid, and fatty acid metabolisms in non-survivors. As the late-stage group, 84 DMs and 10 EPs showed upregulated sphingolipid, taurine, and tryptophan-kynurenine metabolisms with downregulated steroid hormone synthesis in non-survivors. Three DMs and 23 EPs in the consistent group showed more pronounced dysregulation in the dopamine and arachidonic acid metabolisms across all three time zones in non-survivors. Conclusions: This study elucidated the temporal differences in metabolic dysregulation between survivors and non-survivors of severe COVID-19, offering insights into its longitudinal progression and disease mechanisms.
Collapse
Affiliation(s)
- Ryo Uchimido
- Department of Intensive Care Medicine, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8510, Japan
| | - Kenjiro Kami
- Human Metabolome Technologies, Inc., 246-2 Mizukami Kakuganji, Tsuruoka City 997-0052, Japan
| | - Hiroyuki Yamamoto
- Human Metabolome Technologies, Inc., 246-2 Mizukami Kakuganji, Tsuruoka City 997-0052, Japan
| | - Ryo Yokoe
- Department of Intensive Care Medicine, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8510, Japan
| | - Issei Tsuchiya
- Department of Intensive Care Medicine, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8510, Japan
| | - Yoko Nukui
- Department of Infection Control and Laboratory Medicine, Kyoto Prefectural University of Medicine, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Kyoto 602-8566, Japan
| | - Yuki Goto
- Department of Tokyo Metropolitan Health Policy Advisement, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8519, Japan
| | - Mariko Hanafusa
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo 104-0045, Japan
| | - Takeo Fujiwara
- Department of Public Health, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8519, Japan
| | - Kenji Wakabayashi
- Department of Intensive Care Medicine, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo City 113-8510, Japan
| |
Collapse
|
49
|
He K, Li Y, Xiong W, Xing Y, Gao W, Du Y, Kong W, Chen L, Yang X, Dai Z. Sevoflurane exposure accelerates the onset of cognitive impairment via promoting p-Drp1 S616-mediated mitochondrial fission in a mouse model of Alzheimer's disease. Free Radic Biol Med 2024; 225:699-710. [PMID: 39490772 DOI: 10.1016/j.freeradbiomed.2024.10.301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Sevoflurane is an inhalational anesthetic widely used in clinical settings. Accumulating evidence has shown that sevoflurane exposure may impair cognitive function, potentially contributing to Alzheimer's disease (AD)-related changes. However, the underlying mechanism remains poorly understood. In the present study, 4-month-old 5xFAD mice were used to investigate the effect of sevoflurane exposure on cognitive decline by Y-maze test and novel object recognition test. We found that sevoflurane exposure promoted the appearance of cognitive impairment of 5xFAD mice, accompanied with the deterioration of Aβ accumulation, synaptic defects, and neuroinflammation. Additionally, sevoflurane was also found to aggravate mitochondrial fission of 5xFAD mice, as indicated by the further upregulated expression of p-Drp1S616. Moreover, sevoflurane significantly increased mitochondrial damage and dysfunction of AD models both in vitro and in vivo experiments. Seahorse XF analysis further indicated that sevoflurane exposure facilitated a metabolic shift from oxidative phosphorylation to glycolysis. Further rescue experiments revealed that a key mechanism underlying sevoflurane-induced cognitive impairment was the excessive mitochondrial fission, as supported by the result that the mitochondrial fission inhibitor Mdivi-1 counteracted the sevoflurane-mediated deteriorative effects in 5xFAD mice. These findings provided evidence for a new mechanism of sevoflurane exposure accelerating AD-related cognitive decline.
Collapse
Affiliation(s)
- Kaiwu He
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China; Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Youzhi Li
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Wei Xiong
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Yanmei Xing
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Wenli Gao
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Yuting Du
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Wei Kong
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China
| | - Lixin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
| | - Zhongliang Dai
- Department of Anesthesiology, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen, 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China.
| |
Collapse
|
50
|
Zhang Y, Chang L, Xin X, Qiao Y, Qiao W, Ping J, Xia J, Su J. Influenza A virus-induced glycolysis facilitates virus replication by activating ROS/HIF-1α pathway. Free Radic Biol Med 2024; 225:910-924. [PMID: 39491735 DOI: 10.1016/j.freeradbiomed.2024.10.304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
As a highly contagious acute respiratory disease, influenza A virus (A/WSN/1933) poses a huge threat to human health and public health. influenza A virus proliferation relies on glucose metabolism in host cells, yet the effects of influenza A virus on glucose metabolism and the underlying molecular mechanisms remain unclear. Here, we created models of WSN virus-infected mice and A549 cells, along with analyzing metabolomics and transcriptomics data, to investigate how WSN virus infection affects host cell glucose metabolism and specific mechanisms. Analysis of metabolites and gene expression showed that WSN virus infection triggers glycolysis in A549 cells, with notable upregulation of hexokinase 2 (HK2), lactate dehydrogenase A (LDHA), hypoxia-inducible factor-1 alpha (HIF-1α), and elevated lactate levels. Additionally, it leads to mitochondrial impairment and heightened reactive oxygen species (ROS) generation. Elevated levels of glucose may enhance the replication of WSN virus, whereas inhibitors of glycolysis can reduce it. Enhancement of HIF-1α activation facilitated replication of WSN virus through stimulation of lactate synthesis, with the primary influence of glycolysis on WSN virus replication being mediated by ROS/HIF-1α signaling. Mice given HIF-1α inhibitor PTX-478 or glycolysis inhibitor 2-Deoxyglucose (2-DG) exhibited reduced lactate levels and decreased WSN virus replication, along with mitigated weight loss and lung damage. In summary, WSN virus-induced glycolysis has been demonstrated to enhance virus replication through the activation of the ROS/HIF-1α pathway, suggesting potential new targets for combating the virus.
Collapse
Affiliation(s)
- Yijia Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lifeng Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xin Xin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yixuan Qiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wenna Qiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jihui Ping
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jun Xia
- Institute of Veterinary Medicine, Xinjiang Academy of Animal Science, Key Laboratory for Prevention and Control of Herbivorous Animal Diseases of the Ministry of Agriculture and Rural Affairs & Xinjiang Animal Disease Research Key Laboratory, 830000, China.
| | - Juan Su
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|