1
|
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
|
2
|
Liu Y, Yuan H, Hu J, Xu X, Yin S, Hu Y, Liu F. A Complex Network of Obesity-Risk Genes Revealed by Systematic Bioinformatics and Single-Cell Transcriptomic Analyses. J Obes 2025; 2025:7821115. [PMID: 40201036 PMCID: PMC11976034 DOI: 10.1155/jobe/7821115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/05/2024] [Accepted: 11/23/2024] [Indexed: 04/10/2025] Open
Abstract
The development of obesity is closely linked to genetic factors. Despite the identification of numerous genes associated with an increased risk of obesity in humans, a comprehensive understanding of their biological roles has not been achieved. In our extensive bioinformatics study, we identified 802 core genes implicated in obesity. Our protein-protein interaction (PPI) network analysis revealed that these genes form a tightly connected functional network primarily involved in neurological and metabolic regulatory processes. Moreover, our in-depth analysis of single-cell transcriptomic datasets from the human hypothalamus, pancreatic islets, adipose tissue, and liver has shed light on the distinct expression profiles of these obesity-linked genes across various tissue and cell types. This analysis also highlighted the biological processes they influence and the upstream transcriptional regulatory networks involved. Our study not only uncovers the complicated regulatory role of genetic factors in the pathogenesis and progression of obesity but also establishes a close link between the expression patterns and functional roles of these obesity-associated genes. This study provides crucial insights for advancing our understanding of the genetic mechanisms underlying obesity.
Collapse
Affiliation(s)
- Yuenan Liu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Haolin Yuan
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Junhui Hu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xu Xu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yiming Hu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Feng Liu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Otolaryngological Institute of Shanghai Jiaotong University, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
3
|
Kuruppu H, Karunananda M, Jeewandara C, Gomes L, Dissanayake DMCB, Ranatunga C, Chathurangika PH, Senatilleke N, Warnakulasuriya N, Wickramanayake RH, Wijewickrama A, Idampitiya D, Ogg GS, Malavige GN. Oxidative stress induced liver damage in dengue is exacerbated in those with obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.18.25324170. [PMID: 40166538 PMCID: PMC11957102 DOI: 10.1101/2025.03.18.25324170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Obesity and diabetes are risk factors for severe dengue. As there are limited data on the association of obesity with liver dysfunction and oxidative stress in patients with acute dengue, we investigated liver dysfunction associated with obesity, oxidative stress and inflammatory markers, in a large cohort of patients with varying severity of acute dengue. Methods 577 adults dengue patients with acute disease, presenting with a duration of illness ≤ 4 days, were enrolled and followed up from admission to discharge, with clinical and laboratory features recorded. Aspartate transaminase (AST), alanine transaminase (ALT), C-reactive protein, ferritin, 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA) levels were measured, along with the height, weight and waist circumference. Results AST, ALT, CRP and ferritin levels were significantly higher in patients with central obesity (waist circumference of ≥80cm in women or ≥90cm in men) compared to leaner individuals. ALT and CRP levels were also significantly higher in patients with a BMI of ≥ 23.9 kg/m2. 4-HNE levels significantly increased with the rise in AST levels and with ALT levels although not significant. In contrast, MDA levels gradually decreased with the rise in AST levels and ALT levels. There were no differences in 4-HNE and MDA levels in relation to clinical disease severity. However, MDA levels were significantly higher in younger individuals, and leaner individuals with a normal BMI. Furthermore, MDA levels inversely correlated with serum ferritin levels, while AST, ALT and CRP levels significantly correlated ferritin levels. Conclusions 4-HNE and MDA which are markers of lipid peroxidation, appear to play different roles in the pathogenesis of dengue, which should be further investigated for identification of therapeutic targets for treatment of dengue.
Collapse
Affiliation(s)
- Heshan Kuruppu
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Maneshka Karunananda
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Chandima Jeewandara
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Laksiri Gomes
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - D M C B Dissanayake
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Chathura Ranatunga
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Padukkage Harshani Chathurangika
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Nushara Senatilleke
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Navanjana Warnakulasuriya
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Rivindu H Wickramanayake
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | | | - Graham S Ogg
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom, University of Oxford, Oxford, United Kingdom
| | - Gathsaurie Neelika Malavige
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Silva Ramírez B, Peñuelas Urquides K, Escobedo Guajardo BL, Mata Tijerina VL, Cruz Luna JE, Corrales Pérez R, Gómez García S, González Escalante LA, Camacho Moll ME. Assessment of COVID-19 Vaccine Effectiveness Against SARS-CoV-2 Infection, Hospitalization and Death in Mexican Patients with Metabolic Syndrome from Northeast Mexico: A Multicenter Study. Vaccines (Basel) 2025; 13:244. [PMID: 40266114 PMCID: PMC11945729 DOI: 10.3390/vaccines13030244] [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: 11/25/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Metabolic syndrome (MetS) is a predisposing factor for severe COVID-19. The effectiveness of COVID-19 vaccines in patients with MetS has been poorly investigated. The aim of this study was to evaluate the effectiveness of COVID-19 vaccination before (BO) and after the Omicron (AO) SARS-CoV-2 variant in patients with MetS. Methods: This retrospective observational study was carried out in a total of 3194 patients with MetS and a COVID-19 PCR or rapid antigen test. The main outcomes were vaccine effectiveness against infection, hospitalization and death resulting from COVID-19. Results: BO, only two doses of BNT162b2 were effective against infection, this effectiveness was lost AO. Also, with two doses, BNT162b2, ChAdOx1 and CoronaVac were effective against hospitalization BO; however, AO, only BNT162b2 and CoronaVac were effective. Regarding death as an outcome of COVID-19, two doses of BNT162b2 were effective BO, whereas AO, BNT162b2 and CoronaVac were 100% effective. BO the presentation of a sore throat increased after two doses of COVID-19 vaccine regardless of the type, and the presentation of dyspnea diminished after two doses of BNT162b2 and CoronaVac. Conclusions: The SARS-CoV-2 Omicron variant has impacted vaccines' effectiveness against hospitalization and death in patients with MetS. A tailored vaccination scheme for patients with MetS should be implemented due to the varying effectiveness rates observed in our study.
Collapse
Affiliation(s)
- Beatriz Silva Ramírez
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (B.S.R.); (V.L.M.T.)
| | - Katia Peñuelas Urquides
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (K.P.U.); (L.A.G.E.)
| | - Brenda Leticia Escobedo Guajardo
- Laboratory of Molecular Research of Diseases, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico;
| | - Viviana Leticia Mata Tijerina
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (B.S.R.); (V.L.M.T.)
| | - Jorge Eleazar Cruz Luna
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Roberto Corrales Pérez
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Salvador Gómez García
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey 64000, Nuevo Leon, Mexico; (J.E.C.L.); (R.C.P.); (S.G.G.)
| | - Laura Adiene González Escalante
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico; (K.P.U.); (L.A.G.E.)
| | - María Elena Camacho Moll
- Laboratory of Molecular Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey 64720, Nuevo Leon, Mexico
| |
Collapse
|
5
|
Jiang RD, Luo YZ, Lin HF, Zheng XS, Zeng WT, Liu MQ, Deng HH, Wang Q, Lai YN, Chen Y, Guo ZS, Zeng Y, Gong QC, Qiu C, Dong M, Wang X, Wang ZY, Ji LN, Hou PP, Li Q, Shen XR, Li B, Gao Y, Zhang AH, Jiang TT, Shi AM, Zhou P, Lin XH, Deng ZQ, Li JM, Shi ZL. Impaired inflammatory resolution with severe SARS-CoV-2 infection in leptin knock out obese hamster. iScience 2025; 28:111837. [PMID: 39981511 PMCID: PMC11841202 DOI: 10.1016/j.isci.2025.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/28/2024] [Accepted: 12/13/2024] [Indexed: 02/22/2025] Open
Abstract
Comorbidities, such as obesity, increase the risk of severe COVID-19. However, the mechanisms underlying severe illnesses in individuals with obesity are poorly understood. Here, we used gene-edited leptin knock out (Leptin -/-) obese hamsters to establish a severe infection model. This model exhibits robust viral replication, severe lung lesions, pronounced clinical symptoms, and fatal infection, mirroring severe COVID-19 in patients with obesity. Using single-cell transcriptomics on lung tissues pre- and post-infection, we found that monocyte-derived alveolar macrophages (MD-AM) play a key role in lung hyper-inflammation, including two unique MD-AM cell fate branches specific to Leptin -/- hamsters. Notably, reduced Trem2-dependent efferocytosis pathways in Leptin -/- hamsters indicated weakened inflammation resolution, consistent with the scRNA-seq data from patients with obesity. In summary, our study highlights the obesity-associated mechanisms underlying severe SARS-CoV-2 infections and establishes a reliable preclinical animal model for developing obesity-specific therapeutics for critical COVID-19.
Collapse
Affiliation(s)
- Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Zhe Luo
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shuang Zheng
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hao-Hao Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Na Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zi-Shuo Guo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zeng
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Yi Wang
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Li-Na Ji
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Pan-Pan Hou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qian Li
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xu-Rui Shen
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Jiang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Zhou
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xin-Hua Lin
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Zi-Qing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
6
|
De Sanctis JB, Balda Noria G, García AH. Exploring How Adipose Tissue, Obesity, and Gender Influence the Immune Response to Vaccines: A Comprehensive Narrative Review. Int J Mol Sci 2025; 26:862. [PMID: 39859575 PMCID: PMC11765591 DOI: 10.3390/ijms26020862] [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: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
Vaccines represent an essential tool for the prevention of infectious diseases. Upon administration, a complex interaction occurs between the vaccine formulation and the recipient's immune system, ultimately resulting in protection against disease. Significant variability exists in individual and population responses to vaccination, and these differences remain the focus of the ongoing research. Notably, well-documented factors, such as age, gender, and genetic predisposition, influence immune responses. In contrast, the effects of overweight and obesity have not been as thoroughly investigated. The evidence indicates that a high body mass index (BMI) constitutes a significant risk factor for infections in general, with adipose tissue playing a crucial role in modulating the immune response. Furthermore, suboptimal levels of vaccine seroconversion have been observed among individuals with obesity. This review provides a plausible examination of the immunity and protection conferred by various vaccines in individuals with an overweight status, offering a comprehensive analysis of the mechanisms to enhance vaccination efficiency.
Collapse
Affiliation(s)
- Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínská 1333/5, 77900 Olomouc, Czech Republic
- Czech Advanced Technology and Research Institute, Palacky University, 77900 Olomouc, Czech Republic
| | - Germán Balda Noria
- Institute of Immunology Nicolás Enrique Bianco, Faculty of Medicine, Universidad Central de Venezuela Los Chaguaramos, Caracas 1040, Venezuela;
| | - Alexis Hipólito García
- Institute of Immunology Nicolás Enrique Bianco, Faculty of Medicine, Universidad Central de Venezuela Los Chaguaramos, Caracas 1040, Venezuela;
| |
Collapse
|
7
|
La Rosa F, Montecucco F, Liberale L, Sessarego M, Carbone F. Venous thrombosis and obesity: from clinical needs to therapeutic challenges. Intern Emerg Med 2025; 20:47-64. [PMID: 39269539 PMCID: PMC11794390 DOI: 10.1007/s11739-024-03765-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Weight bias and stigma have limited the awareness of the systemic consequences related to obesity. As the narrative evolves, obesity is emerging as a driver and enhancer of many pathological conditions. Among these, the risk of venous thromboembolism (VTE) is a critical concern linked to obesity, ranking as the third most common cardiovascular condition. Obesity is recognized as a multifactorial risk factor for VTE, influenced by genetic, demographic, behavioral, and socio-economic conditions. Despite established links, the exact incidence of obesity related VTE in the general population remains largely unknown. The complexity of distinguishing between provoked and unprovoked VTE, coupled with gaps in obesity definition and assessment still complicates a tailored risk assessment of VTE risk. Obesity reactivity, hypercoagulability, and endothelial dysfunction are driven by the so-called 'adiposopathy'. This state of chronic inflammation and metabolic disturbance amplifies thrombin generation and alters endothelial function, promoting a pro-thrombotic environment. Additionally, the inflammation-induced clot formation-also referred to as 'immunothrombosis' further exacerbates VTE risk in people living with obesity. Furthermore, current evidence highlights significant gaps in the management of obesity related VTE, particularly concerning prophylaxis and treatment efficacy of anticoagulants in people living with obesity. This review underscores the need for tailored therapeutic approaches and well-designed clinical trials to address the unique challenges posed by obesity in VTE prevention and management. Advanced research and innovative strategies are imperative to improve outcomes and reduce the burden of VTE in people living with obesity.
Collapse
Affiliation(s)
- Federica La Rosa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy.
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Marta Sessarego
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
8
|
Jia S, Yin Z, Pan H, Wang F, Liu X, Wang Q, Zhang L, Tang J, Yang H, Du J, Wang Z, Jin P, Peng Z, Tang R, Kang G, Wang X, Li S, Wang W, Li J, Shen H, Zhu F. Relative effectiveness of a heterologous booster dose with adenovirus type 5 vectored COVID-19 vaccine versus three doses of inactivated COVID-19 vaccine in adults during a nationwide outbreak of omicron predominance, in China: a retrospective, individually matched cohort-control study. Emerg Microbes Infect 2024; 13:2332660. [PMID: 38678636 PMCID: PMC11057405 DOI: 10.1080/22221751.2024.2332660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/15/2024] [Indexed: 05/01/2024]
Abstract
Effectiveness of heterologous booster regimes with ad5 vectored COVID-19 vaccine in a large, diverse population during the national-scale outbreak of SARS-CoV-2 omicron predominance in China has not been reported, yet. We conducted a large-scale cohort-control study in six provinces in China, and did a retrospective survey on the COVID-19 attack risk during this outbreak. Participant aged ≥18 years in five previous trials who were primed with 1 to 3 doses of ICV received heterologous booster with either intramuscular or orally inhaled ad5 vectored COVID-19 vaccine were included in the heterologous-trial cohort. We performed propensity score-matching at a ratio of 1:4 to match participants in the heterologous-trial cohort individually with the community individuals who received three-dose of ICV as a control (ICV-community cohort). From February 4 to April 10, 2023, 41504 (74.5%) of 55710 individuals completed the survey. The median time since the most recent vaccination to the onset of the symptoms of COVID-19 was 303.0 days (IQR 293.0-322.0). The attack rate of COVID-19 in the heterologous-trial cohort was 55.8%, while that in the ICV-community cohort was 64.6%, resulting in a relative effectiveness of 13.7% (95% CI 11.9 to 15.3). In addition, a higher relative effectiveness against COVID-19 associated outpatient visits, and admission to hospital was demonstrated, which was 25.1% (95% CI 18.9 to 30.9), and 48.9% (95% CI 27.0 to 64.2), respectively. The heterologous booster with ad5 vectored COVID-19 vaccine still offered some additional protection in preventing COVID-19 breakthrough infection versus homologous three-dose regimen with ICV, 10 months after vaccination.
Collapse
Affiliation(s)
- Siyue Jia
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Zundong Yin
- China Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Hongxing Pan
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Fuzhen Wang
- China Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xiaoqiang Liu
- Yunnan Provincial Center for Disease Control and Prevention, Kunming, People's Republic of China
| | - Qing Wang
- Chongqing Provincial Center for Disease Control and Prevention, Chongqing, People's Republic of China
| | - Li Zhang
- Shandong Provincial Center for Disease Control and Prevention, Jinan, People's Republic of China
| | - Jihai Tang
- Anhui Provincial Center for Disease Control and Prevention, Hefei, People's Republic of China
| | - Hao Yang
- Hunan Provincial Center for Disease Control and Prevention, Changsha, People's Republic of China
| | - Jiangbo Du
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhiguo Wang
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Pengfei Jin
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Zhihang Peng
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Rong Tang
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Guodong Kang
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
| | - Xuewen Wang
- Canming Medical Technology Co., Ltd, Shanghai, People's Republic of China
| | - Simin Li
- School of Public Health, Southeast University; Nanjing, People's Republic of China
| | - Weixiao Wang
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jingxin Li
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- School of Public Health, Southeast University; Nanjing, People's Republic of China
| | - Hongbing Shen
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- China Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Fengcai Zhu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, People's Republic of China
- National Vaccine Innovation Platform and Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
- School of Public Health, Southeast University; Nanjing, People's Republic of China
| |
Collapse
|
9
|
Clarke M, Mathew SM, Giles LC, Barr IG, Richmond PC, Marshall HS. The Impact of Obesity on Influenza Vaccine Immunogenicity and Antibody Transfer to the Infant During Pregnancy. Vaccines (Basel) 2024; 12:1307. [PMID: 39771969 PMCID: PMC11680122 DOI: 10.3390/vaccines12121307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Influenza vaccination is recommended for pregnant women, offering the dual benefit of protecting pregnant women and their newborn infants against influenza. This study aimed to investigate the impact of body mass index (BMI) on influenza vaccine responses in pregnant women and their newborns. METHODS Participants included pregnant women attending the Women's and Children's Hospital in South Australia between 2018 and 2021. Maternal blood samples were collected prior to and at 1 and 6 months post-influenza vaccination to measure antibody responses by hemagglutination inhibition (HI) assay. Cord blood samples were also collected. The percentages of participants achieving HI titre ≥40 were compared between obese and non-obese groups. RESULTS A total of 73 women were enrolled and received quadrivalent influenza vaccination at a mean age of 32 years (range 21-44 y) and median gestation of 24 weeks (range 11-37 weeks). BMI at vaccination was ≥30 kg/m2 for 21/73 women (29%). Most pregnant women demonstrated antibody titres ≥ 40 to all four influenza vaccine strains at 1 month post-vaccination regardless of BMI category (BMI ≥ 30 kg/m2: 19/20; 95% vs. BMI < 30 kg/m2: 47/49; 96%). At 6 months post-vaccination, 12/17 (71%) obese women compared to 36/43 (84%) non-obese women (p = 0.25) maintained HI titres ≥ 40. Cord blood serology showed HI titres ≥ 40 for 11/17 (65%) infants born to mothers with BMI ≥ 30 compared to 30/35 (86%) infants delivered by mothers with BMI < 30 kg/m2. CONCLUSIONS A high BMI did not impair influenza vaccine antibody responses in pregnant women at 1 month post-vaccination. However, at 6 months post-vaccination, and in the cord blood samples, the percentages maintaining HI titre ≥ 40 were lower for obese women than for non-obese pregnant women.
Collapse
Affiliation(s)
- Michelle Clarke
- Women’s and Children’s Health Network, North Adelaide, SA 5006, Australia; (M.C.); (S.M.M.)
- The Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Suja M. Mathew
- Women’s and Children’s Health Network, North Adelaide, SA 5006, Australia; (M.C.); (S.M.M.)
- The Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Lynne C. Giles
- School of Public Health, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Ian G. Barr
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute, Melbourne, VIC 3000, Australia;
| | - Peter C. Richmond
- Discipline of Pediatrics, University of Western Australia, Perth, WA 6009, Australia;
- Wesfarmers Centre of Vaccines and Infectious Diseases, Kids Research Institute of Australia, Perth Children’s Hospital, Perth, WA 6009, Australia
| | - Helen S. Marshall
- Women’s and Children’s Health Network, North Adelaide, SA 5006, Australia; (M.C.); (S.M.M.)
- The Robinson Research Institute, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
10
|
D'Souza M, Keeshan A, Gravel CA, Langlois MA, Cooper CL. Obesity does not influence SARS-CoV-2 humoral vaccine immunogenicity. NPJ Vaccines 2024; 9:226. [PMID: 39557875 PMCID: PMC11574036 DOI: 10.1038/s41541-024-01022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity is a recognized factor influencing immune function and infectious disease outcomes. Characterization of the influence of obesity on SARS-CoV-2 humoral vaccine immunogenicity is required to properly tailor vaccine type (mRNA, viral-vector, protein subunit vaccines) and dosing schedule. Data from a prospective cohort study collected over 34 months was used to evaluate the slope of antibody production and decay and neutralizing capacity following SARS-CoV-2 vaccination in individuals with and without obesity at baseline. Most participants were female (65.4%), white (92.4%), and received mRNA vaccines. 210 were obese and 697 non-obese. Sex and infection-acquired immunity were identified as effect modifiers for the relationship between obesity and COVID-19 vaccine humoral immunogenicity. No consistent influence of obesity on peak titres, titre retention, antibody isotype (IgG, IgM, IgA), or neutralization was identified when controlling for other key variables. It may not be necessary to consider this variable when developing SARS-CoV-2 vaccine dosing strategies.
Collapse
Affiliation(s)
| | - Alexa Keeshan
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Christopher A Gravel
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
- Department of Mathematics and Statistics, University of Ottawa, Ottawa, ON, Canada
- Data Literacy Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Curtis L Cooper
- Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Division of Infectious Diseases, Department of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
11
|
Huang Y, Wang W, Liu Y, Wang Z, Cao B. COVID-19 vaccine updates for people under different conditions. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2323-2343. [PMID: 39083202 DOI: 10.1007/s11427-024-2643-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 10/22/2024]
Abstract
SARS-CoV-2 has caused global waves of infection since December 2019 and continues to persist today. The emergence of SARS-CoV-2 variants with strong immune evasion capabilities has compromised the effectiveness of existing vaccines against breakthrough infections. Therefore, it is important to determine the best utilization strategies for different demographic groups given the variety of vaccine options available. In this review, we will discuss the protective efficacy of vaccines during different stages of the epidemic and emphasize the importance of timely updates to target prevalent variants, which can significantly improve immune protection. While it is recognized that vaccine effectiveness may be lower in certain populations such as the elderly, individuals with chronic comorbidities (e.g., diabetes with poor blood glucose control, those on maintenance dialysis), or those who are immunocompromised compared to the general population, administering multiple doses can result in a strong protective immune response that outweighs potential risks. However, caution should be exercised when considering vaccines that might trigger an intense immune response in populations prone to inflammatory flare or other complications. In conclusion, individuals with special conditions require enhanced and more effective immunization strategies to prevent infection or reinfection, as well as to avoid the potential development of long COVID.
Collapse
Affiliation(s)
- Yijiao Huang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Weiyang Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yan Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Department of Infectious Disease, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264000, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China.
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Changping Laboratory, Beijing, 102200, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, 100069, China.
- New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
12
|
Badaruddin IA, Othman H, Wan Shuaib WMA, Che Yusof MZ, Othman S, Anuar Ali MN, Mat IM, Md Mansor M, Nawi AM. Navigating Immune Landscapes: Insights From A Prospective Study. Asia Pac J Public Health 2024; 36:806-809. [PMID: 39318114 DOI: 10.1177/10105395241282621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Affiliation(s)
- Izzatul Aliaa Badaruddin
- Department of Pathology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Hanita Othman
- Department of Pathology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Wan Muhammad Azfar Wan Shuaib
- Department of Pathology, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Zakie Che Yusof
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Shahifaniza Othman
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Nazrul Anuar Ali
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Ida Mastura Mat
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Munirah Md Mansor
- Department of Diagnostic Laboratory Services, Hospital Canselor Tuanku Muhriz, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Azmawati Mohammed Nawi
- Department of Public Health, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Lam LK, Tan JT, Ooi PH, Zhang R, Chan KH, Mao X, Hung IFN, Seto WK, Yuen MF, Cheung KS. Effect of metabolic dysfunction-associated steatotic liver disease on BNT162b2 immunogenicity against the severe acute respiratory syndrome coronavirus 2 omicron variant. J Gastroenterol Hepatol 2024; 39:2386-2393. [PMID: 39152762 PMCID: PMC11618226 DOI: 10.1111/jgh.16716] [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: 04/29/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AND AIM We aimed to investigate the effect of metabolic dysfunction-associated steatotic liver disease (MASLD) on three-dose BNT162b2 immunogenicity to the omicron variant. METHODS Adult recipients of three doses of BNT162b2 were prospectively recruited between May and December 2021. The serology of the neutralizing antibody by live virus microneutralization (vMN) to the omicron variant was measured at baseline, day 180, and day 360 after the first dose. The primary outcome was seroconversion (vMN titer ≥ 10) at day 360. Exposure of interest was MASLD, defined as hepatic steatosis (controlled attenuation parameter ≥ 248 dB/m on transient elastography) plus at least one of five cardiometabolic risk factors. Subjects with prior COVID-19 were excluded. A multivariable logistic regression model was used to derive the adjusted odds ratio of seroconversion with MASLD by adjusting for age, sex, antibiotic use, and proton pump inhibitor use. RESULTS One hundred forty-eight BNT162b2 recipients (male: 48 [32.4%]; median age: 51.0 years [interquartile range, IQR: 44.5-57.3]) were recruited. The median time from the first dose to the third dose was 8.5 months (IQR: 7.9-8.9). MASLD subjects had a lower seroconversion rate than non-MASLD ones (89.6% vs 99.0%; P = 0.007). MASLD was the only independent risk factor for seroconversion (adjusted odds ratio: 0.051, 95% confidence interval: 0.002-0.440). Subgroup analysis of immunogenicity at 4 months after the third dose shows significantly lower vMN titer (13.06 [IQR: 7.69-22.20] vs 33.49 [IQR: 24.05-46.53]; P = 0.004) and seroconversion rate (76.9% vs 97.4%; P = 0.016) in MASLD than non-MASLD subjects, but not within 4 months from the third dose (vMN titer: 46.87 [IQR: 33.12-66.02] vs 41.86 [IQR: 34.47-50.91], P = 0.240; seroconversion rate: 94.3% vs 100%, P = 0.131). CONCLUSION Metabolic dysfunction-associated steatotic liver disease was a risk factor for poorer immunogenicity to the omicron variant, with a more pronounced waning effect compared among three-dose BNT162b2 recipients.
Collapse
Affiliation(s)
- Lok Ka Lam
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Jing Tong Tan
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Poh Hwa Ooi
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Ruiqi Zhang
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Kwok Hung Chan
- Department of MicrobiologyThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Xianhua Mao
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Ivan F N Hung
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
| | - Wai Kay Seto
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
- Department of MedicineThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
- State Key Laboratory of Liver ResearchThe University of Hong KongPok Fu LamHong Kong
| | - Man Fung Yuen
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongPok Fu LamHong Kong
| | - Ka Shing Cheung
- Department of Medicine, School of Clinical MedicineThe University of Hong Kong, Queen Mary HospitalPok Fu LamHong Kong
- Department of MedicineThe University of Hong Kong‐Shenzhen HospitalShenzhenChina
| |
Collapse
|
14
|
Collins TJC, Morgan PK, Man K, Lancaster GI, Murphy AJ. The influence of metabolic disorders on adaptive immunity. Cell Mol Immunol 2024; 21:1109-1119. [PMID: 39134802 PMCID: PMC11442657 DOI: 10.1038/s41423-024-01206-1] [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: 04/07/2024] [Accepted: 07/19/2024] [Indexed: 10/02/2024] Open
Abstract
The immune system plays a crucial role in protecting the body from invading pathogens and maintaining tissue homoeostasis. Maintaining homoeostatic lipid metabolism is an important aspect of efficient immune cell function and when disrupted immune cell function is impaired. There are numerous metabolic diseases whereby systemic lipid metabolism and cellular function is impaired. In the context of metabolic disorders, chronic inflammation is suggested to be a major contributor to disease progression. A major contributor to tissue dysfunction in metabolic disease is ectopic lipid deposition, which is generally caused by diet and genetic factors. Thus, we propose the idea, that similar to tissue and organ damage in metabolic disorders, excessive accumulation of lipid in immune cells promotes a dysfunctional immune system (beyond the classical foam cell) and contributes to disease pathology. Herein, we review the evidence that lipid accumulation through diet can modulate the production and function of immune cells by altering cellular lipid content. This can impact immune cell signalling, activation, migration, and death, ultimately affecting key aspects of the immune system such as neutralising pathogens, antigen presentation, effector cell activation and resolving inflammation.
Collapse
Affiliation(s)
- Thomas J C Collins
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Pooranee K Morgan
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Kevin Man
- Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
- Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Graeme I Lancaster
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
15
|
Malavige GN, Ogg GS. Immune responses and severe dengue: what have we learned? Curr Opin Infect Dis 2024; 37:349-356. [PMID: 39079180 DOI: 10.1097/qco.0000000000001040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW With the marked rise in dengue globally, developing well tolerated and effective vaccines and therapeutics is becoming more important. Here we discuss the recent developments in the understanding of immune mechanisms that lead to severe dengue and the learnings from the past, that can help us to find therapeutic targets, prognostic markers, and vaccines to prevent development of severe disease. RECENT FINDINGS The extent and duration of viraemia often appears to be associated with clinical disease severity but with some variability. However, there also appear to be significant differences in the kinetics of viraemia and nonstructural protein 1 (NS1) antigenemia and pathogenicity between different serotypes and genotypes of the DENV. These differences may have significant implications for development of treatments and in inducing robust immunity through dengue vaccines. Although generally higher levels of neutralizing antibodies are thought to protect against infection and severe disease, there have been exceptions and the specificity, breadth and functionality of the antibody responses are likely to be important. SUMMARY Although there have been many advances in our understanding of dengue pathogenesis, viral and host factors associated with occurrence of severe dengue, vascular leak and the immune correlates of protection remain poorly understood.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Graham S Ogg
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical Sciences, University of Sri Jayewardenepura, Sri Lanka
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Atef Y, Ito T, Masuda A, Kato Y, Nishimura A, Kanda Y, Kunisawa J, Kusakabe T, Nishida M. Diabetic Mice Spleen Vulnerability Contributes to Decreased Persistence of Antibody Production after SARS-CoV-2 Vaccine. Int J Mol Sci 2024; 25:10379. [PMID: 39408710 PMCID: PMC11476529 DOI: 10.3390/ijms251910379] [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: 08/17/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
During the COVID-19 pandemic, diabetic and obese patients experienced higher rates of hospital admissions, severe illness, and mortality. However, vaccinations failed to provide those vulnerable populations the same level of protection against COVID-19 severity as those without diabetic and obese phenotypes. Our study aimed to investigate how diabetes mellitus (DM) impacts the immune response following vaccination including the artificially designed trimeric SARS-CoV-2 spike (S)-protein. By using two diabetic mouse models, ob/ob mice (obese, hyperglycemic, and insulin-resistant) and STZ-treated mice (insulin-deficient and hyperglycemic), we observed a significant reduction in S-protein-specific IgG antibody titer post-vaccination in both diabetic models compared to wild-type (WT) mice. Both diabetic mouse models exhibited significant abnormalities in spleen tissue, including marked reductions in splenic weight and the size of the white pulp regions. Furthermore, the splenic T-cell and B-cell zones were notably diminished, suggesting an underlying immune dysfunction that could contribute to impaired antibody production. Notably, vaccination with the S-protein, when paired with an optimal adjuvant, did not exacerbate diabetic cardiomyopathy, blood glucose levels, or liver function, providing reassurance about the vaccine's safety. These findings offer valuable insights into potential mechanisms responsible for the decreased persistence of antibody production in diabetic patients.
Collapse
Affiliation(s)
- Yara Atef
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tomoya Ito
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akitsu Masuda
- Laboratory of Creative Science for Insect Industries, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 819-0395, Japan;
| | - Yuri Kato
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki 210-9501, Japan;
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Fukuoka 819-0395, Japan;
| | - Motohiro Nishida
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| |
Collapse
|
17
|
Ali M, Longet S, Neale I, Rongkard P, Chowdhury FUH, Hill J, Brown A, Laidlaw S, Tipton T, Hoque A, Hassan N, Hackstein CP, Adele S, Akther HD, Abraham P, Paul S, Rahman MM, Alam MM, Parvin S, Mollah FH, Hoque MM, Moore SC, Biswas SK, Turtle L, de Silva TI, Ogbe A, Frater J, Barnes E, Tomic A, Carroll MW, Klenerman P, Kronsteiner B, Chowdhury FR, Dunachie SJ. Obesity differs from diabetes mellitus in antibody and T-cell responses post-COVID-19 recovery. Clin Exp Immunol 2024; 218:78-92. [PMID: 38642547 PMCID: PMC11404124 DOI: 10.1093/cei/uxae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 01/05/2024] [Accepted: 04/18/2024] [Indexed: 04/22/2024] Open
Abstract
OBJECTIVE Obesity and type 2 diabetes (DM) are risk factors for severe coronavirus disease 2019 (COVID-19) outcomes, which disproportionately affect South Asian populations. This study aims to investigate the humoral and cellular immune responses to SARS-CoV-2 in adult COVID-19 survivors with overweight/obesity (Ov/Ob, BMI ≥ 23 kg/m2) and DM in Bangladesh. METHODS In this cross-sectional study, SARS-CoV-2-specific antibody and T-cell responses were investigated in 63 healthy and 75 PCR-confirmed COVID-19 recovered individuals in Bangladesh, during the pre-vaccination first wave of the COVID-19 pandemic in 2020. RESULTS In COVID-19 survivors, SARS-CoV-2 infection induced robust antibody and T-cell responses, which correlated with disease severity. After adjusting for age, sex, DM status, disease severity, and time since onset of symptoms, Ov/Ob was associated with decreased neutralizing antibody titers, and increased SARS-CoV-2 spike-specific IFN-γ response along with increased proliferation and IL-2 production by CD8 + T cells. In contrast, DM was not associated with SARS-CoV-2-specific antibody and T-cell responses after adjustment for obesity and other confounders. CONCLUSION Ov/Ob is associated with lower neutralizing antibody levels and higher T-cell responses to SARS-CoV-2 post-COVID-19 recovery, while antibody or T-cell responses remain unaltered in DM.
Collapse
Affiliation(s)
- Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Directorate General of Health Services, Dhaka, Bangladesh
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Patpong Rongkard
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Jennifer Hill
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stephen Laidlaw
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Tipton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ashraful Hoque
- Department of Transfusion Medicine, Sheikh Hasina National Burn & Plastics Surgery Institute, Dhaka, Bangladesh
| | - Nazia Hassan
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Hossain Delowar Akther
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Shrebash Paul
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Matiur Rahman
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Masum Alam
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Shamima Parvin
- Department of Biochemistry and Molecular Biology, Mugda Medical College, Dhaka, Bangladesh
| | - Forhadul Hoque Mollah
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Md Mozammel Hoque
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Shona C Moore
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| | - Subrata K Biswas
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Lance Turtle
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ane Ogbe
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Adriana Tomic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Miles W Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Fazle Rabbi Chowdhury
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Department of Internal Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Susanna J Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
18
|
Xue P, Merikanto I, Delale EA, Bjelajac A, Yordanova J, Chan RNY, Korman M, Mota-Rolim SA, Landtblom AM, Matsui K, Reis C, Penzel T, Inoue Y, Nadorff MR, Holzinger B, Morin CM, Espie CA, Plazzi G, De Gennaro L, Chung F, Bjorvatn B, Wing YK, Dauvilliers Y, Partinen M, Benedict C. Associations between obesity, a composite risk score for probable long COVID, and sleep problems in SARS-CoV-2 vaccinated individuals. Int J Obes (Lond) 2024; 48:1300-1306. [PMID: 38849462 PMCID: PMC11347373 DOI: 10.1038/s41366-024-01556-w] [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: 08/16/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Preliminary data suggests that obesity might hasten the decline in mRNA vaccine-induced immunity against SARS-CoV-2. However, whether this renders individuals with obesity more susceptible to long COVID symptoms post-vaccination remains uncertain. Given sleep's critical role in immunity, exploring the associations between obesity, probable long COVID symptoms, and sleep disturbances is essential. METHODS We analyzed data from a survey of 5919 adults aged 18 to 89, all of whom received two SARS-CoV-2 mRNA vaccinations. Participants were categorized into normal weight, overweight, and obesity groups based on ethnicity-specific BMI cutoffs. The probability of long COVID was evaluated using the Post-Acute Sequelae of SARS-CoV-2 (PASC) score, as our survey did not permit confirmation of acute SARS-CoV-2 infection through methods such as antibody testing. Additionally, sleep patterns were assessed through questionnaires. RESULTS Participants with obesity exhibited a significantly higher adjusted odds ratio (OR) of having a PASC score of 12 or higher, indicative of probable long COVID in our study, compared to those with normal weight (OR: 1.55, 95% CI: 1.05, 2.28). No significant difference was observed for overweight individuals (OR: 0.92 [95% CI: 0.63, 1.33]). Both obesity and probable long COVID were associated with increased odds of experiencing a heightened sleep burden, such as the presence of obstructive sleep apnea or insomnia (P < 0.001). However, no significant interaction between BMI and probable long COVID status was found. CONCLUSIONS Even post-vaccination, individuals with obesity may encounter a heightened risk of experiencing prolonged COVID-19 symptoms. However, confirming our observations necessitates comprehensive studies incorporating rigorous COVID infection testing, such as antibody assays - unavailable in our anonymous survey. Additionally, it is noteworthy that the correlation between probable long COVID and sleep disturbances appears to be independent of BMI.
Collapse
Affiliation(s)
- Pei Xue
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| | - Ilona Merikanto
- Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eva A Delale
- Institute for Anthropological Research, Zagreb, Croatia
| | - Adrijana Bjelajac
- Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Juliana Yordanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Rachel N Y Chan
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Maria Korman
- Department of Occupational Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | | | - Anne-Marie Landtblom
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Kentaro Matsui
- Department of Clinical Laboratory, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Catia Reis
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Faculdade de Ciências Humanas, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Thomas Penzel
- Sleep Medicine Center, Charite University Hospital Berlin, Berlin, Germany
| | - Yuichi Inoue
- Department of Somnology, Tokyo Medical University, Tokyo, Japan
- Japan Somnology Center, Institute of Neuropsychiatry, Tokyo, Japan
| | - Michael R Nadorff
- Department of Psychology, Mississippi State University, Mississippi, MI, USA
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Baylor, TX, USA
| | - Brigitte Holzinger
- Medical University of Vienna, Postgraduate, Schlafcoaching, Vienna, Austria
| | - Charles M Morin
- Centre de recherche CERVO/Brain Research Center, École de psychologie, Université Laval, Quebec City, Quebec, Canada
| | - Colin A Espie
- Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, QC, UK
| | - Giuseppe Plazzi
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luigi De Gennaro
- Department of Psychology, Sapienza University of Rome, Roma, Lazio, Italy
- IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Frances Chung
- Department of Anesthesiology and Pain Medicine, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Bjørn Bjorvatn
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Norwegian Competence Center for Sleep Disorders, Haukeland University Hospital, Bergen, Norway
| | - Yun Kwok Wing
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yves Dauvilliers
- Sleep-Wake Disorders Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU, Montpellier, France
- INM, University Montpellier, INSERM, Montpellier, France
| | - Markku Partinen
- Department of Clinical Neurosciences, University of Helsinki Clinicum Unit, Helsinki, Finland
- Helsinki Sleep Clinic, Terveystalo Healthcare Services, Helsinki, Finland
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
19
|
Fischer K, Lulla A, So TY, Pereyra-Gerber P, Raybould MIJ, Kohler TN, Yam-Puc JC, Kaminski TS, Hughes R, Pyeatt GL, Leiss-Maier F, Brear P, Matheson NJ, Deane CM, Hyvönen M, Thaventhiran JED, Hollfelder F. Rapid discovery of monoclonal antibodies by microfluidics-enabled FACS of single pathogen-specific antibody-secreting cells. Nat Biotechnol 2024:10.1038/s41587-024-02346-5. [PMID: 39143416 DOI: 10.1038/s41587-024-02346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/27/2024] [Indexed: 08/16/2024]
Abstract
Monoclonal antibodies are increasingly used to prevent and treat viral infections and are pivotal in pandemic response efforts. Antibody-secreting cells (ASCs; plasma cells and plasmablasts) are an excellent source of high-affinity antibodies with therapeutic potential. Current methods to study antigen-specific ASCs either have low throughput, require expensive and labor-intensive screening or are technically demanding and therefore not widely accessible. Here we present a straightforward technology for the rapid discovery of monoclonal antibodies from ASCs. Our approach combines microfluidic encapsulation of single cells into an antibody capture hydrogel with antigen bait sorting by conventional flow cytometry. With our technology, we screened millions of mouse and human ASCs and obtained monoclonal antibodies against severe acute respiratory syndrome coronavirus 2 with high affinity (<1 pM) and neutralizing capacity (<100 ng ml-1) in 2 weeks with a high hit rate (>85% of characterized antibodies bound the target). By facilitating access to the underexplored ASC compartment, the approach enables efficient antibody discovery and immunological studies into the generation of protective antibodies.
Collapse
Affiliation(s)
- Katrin Fischer
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Aleksei Lulla
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Tsz Y So
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | - Pehuén Pereyra-Gerber
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Matthew I J Raybould
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Timo N Kohler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Tomasz S Kaminski
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Robert Hughes
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | | | | | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Nicholas J Matheson
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
20
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [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: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
21
|
Runnstrom MC, Lamothe PA, Faliti CE, Cheedarla N, Moreno A, Suthar MS, Nahata R, Ravindran M, Haddad NS, Morrison-Porter A, Quehl H, Ramonell RP, Woodruff M, Anam F, Zhang R, Swenson C, Polito C, Neveu W, Patel R, Smirnova N, Nguyen DC, Kim C, Hentenaar I, Kyu S, Usman S, Ngo T, Guo Z, Wu H, Daiss JL, Park J, Manning KE, Wali B, Ellis ML, Sharma S, Holguin F, Cheedarla S, Neish AS, Roback JD, Sanz I, Eun-Hyung Lee F. Patients taking benralizumab, dupilumab, or mepolizumab have lower postvaccination SARS-CoV-2 immunity. J Allergy Clin Immunol 2024; 154:435-446. [PMID: 38878020 PMCID: PMC11305925 DOI: 10.1016/j.jaci.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/27/2024] [Accepted: 03/15/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Biologic therapies inhibiting the IL-4 or IL-5 pathways are very effective in the treatment of asthma and other related conditions. However, the cytokines IL-4 and IL-5 also play a role in the generation of adaptive immune responses. Although these biologics do not cause overt immunosuppression, their effect in primary severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunization has not been studied completely. OBJECTIVE Our aim was to evaluate the antibody and cellular immunity after SARS-CoV-2 mRNA vaccination in patients on biologics (PoBs). METHODS Patients with severe asthma or atopic dermatitis who were taking benralizumab, dupilumab, or mepolizumab and had received the initial dose of the 2-dose adult SARS-CoV-2 mRNA vaccine were enrolled in a prospective, observational study. As our control group, we used a cohort of immunologically healthy subjects (with no significant immunosuppression) who were not taking biologics (NBs). We used a multiplexed immunoassay to measure antibody levels, neutralization assays to assess antibody function, and flow cytometry to quantitate Spike-specific lymphocytes. RESULTS We analyzed blood from 57 patients in the PoB group and 46 control subjects from the NB group. The patients in the PoB group had lower levels of SARS-CoV-2 antibodies, pseudovirus neutralization, live virus neutralization, and frequencies of Spike-specific B and CD8 T cells at 6 months after vaccination. In subgroup analyses, patients with asthma who were taking biologics had significantly lower pseudovirus neutralization than did subjects with asthma who were not taking biologics. CONCLUSION The patients in the PoB group had reduced SARS-CoV-2-specific antibody titers, neutralizing activity, and virus-specific B- and CD8 T-cell counts. These results have implications when considering development of a more individualized immunization strategy in patients who receive biologic medications blocking IL-4 or IL-5 pathways.
Collapse
Affiliation(s)
- Martin C Runnstrom
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Department of Medicine, Atlanta Veterans Affairs Healthcare System, Atlanta, Ga
| | - Pedro A Lamothe
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Caterina E Faliti
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Lowance Center for Human Immunology, Atlanta, Ga
| | - Narayanaiah Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Alberto Moreno
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Yerkes National Primate Research Center, Atlanta, Ga
| | - Mehul S Suthar
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Ga; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Rishika Nahata
- Emory College of Arts and Sciences, Emory University, Atlanta, Ga
| | - Mayuran Ravindran
- J. Willis Hurst Internal Medicine Residency Program, Emory University School of Medicine, Atlanta, Ga
| | - Natalie S Haddad
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Andrea Morrison-Porter
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Hannah Quehl
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Richard P Ramonell
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Matthew Woodruff
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Lowance Center for Human Immunology, Atlanta, Ga
| | - Fabliha Anam
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Lowance Center for Human Immunology, Atlanta, Ga
| | - Rebeca Zhang
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Ga
| | - Colin Swenson
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Carmen Polito
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Wendy Neveu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Rahulkumar Patel
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Natalia Smirnova
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Doan C Nguyen
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Lowance Center for Human Immunology, Atlanta, Ga
| | - Caroline Kim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Ian Hentenaar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Shuya Kyu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Sabeena Usman
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Thuy Ngo
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Zhenxing Guo
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Ga
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Ga
| | - John L Daiss
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Jiwon Park
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Kelly E Manning
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Ga; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Bursha Wali
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Ga; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Madison L Ellis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Ga; Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Ga
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colo
| | - Fernando Holguin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colo
| | - Suneethamma Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Ga
| | - John D Roback
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Ga
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, Ga; Lowance Center for Human Immunology, Atlanta, Ga
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Ga.
| |
Collapse
|
22
|
Wratil PR, Le Thi TG, Osterman A, Badell I, Huber M, Zhelyazkova A, Wichert SP, Litwin A, Hörmansdorfer S, Strobl F, Grote V, Jebrini T, Török HP, Hornung V, Choukér A, Koletzko B, Adorjan K, Koletzko S, Keppler OT. Dietary habits, traveling and the living situation potentially influence the susceptibility to SARS-CoV-2 infection: results from healthcare workers participating in the RisCoin Study. Infection 2024; 52:1425-1437. [PMID: 38436913 PMCID: PMC11289231 DOI: 10.1007/s15010-024-02201-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/25/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE To explore occupational and non-occupational risk and protective factors for the coronavirus disease 2019 (COVID-19) in healthcare workers (HCWs). METHODS Serum specimens and questionnaire data were obtained between October 7 and December 16, 2021 from COVID-19-vaccinated HCWs at a quaternary care hospital in Munich, Germany, and were analyzed in the RisCoin Study. RESULTS Of 3,696 participants evaluated, 6.6% have had COVID-19 at least once. Multivariate logistic regression analysis identified working in patient care occupations (7.3% had COVID-19, 95% CI 6.4-8.3, Pr = 0.0002), especially as nurses, to be a potential occupation-related COVID-19 risk factor. Non-occupational factors significantly associated with high rates of the disease were contacts to COVID-19 cases in the community (12.8% had COVID-19, 95% CI 10.3-15.8, Pr < 0.0001), being obese (9.9% had COVID-19, 95% CI 7.1-13.5, Pr = 0.0014), and frequent traveling abroad (9.4% had COVID-19, 95% CI 7.1-12.3, Pr = 0.0088). On the contrary, receiving the basic COVID-19 immunization early during the pandemic (5.9% had COVID-19, 95% CI 5.1-6.8, Pr < 0.0001), regular smoking (3.6% had COVID-19, 95% CI 2.1-6.0, Pr = 0.0088), living with the elderly (3.0% had COVID-19, 95% CI 1.0-8.0, Pr = 0.0475), and frequent consumption of ready-to-eat meals (2.6% had COVID-19, 95% CI 1.1-5.4, Pr = 0.0045) were non-occupational factors potentially protecting study participants against COVID-19. CONCLUSION The newly discovered associations between the living situation, traveling as well as dietary habits and altered COVID-19 risk can potentially help refine containment measures and, furthermore, contribute to new mechanistic insights that may aid the protection of risk groups and vulnerable individuals.
Collapse
Affiliation(s)
- Paul R Wratil
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Thu Giang Le Thi
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Andreas Osterman
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Irina Badell
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Melanie Huber
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Pettenkoferstr. 9a, 80336, Munich, Germany
| | - Ana Zhelyazkova
- Institut für Notfallmedizin und Medizinmanagement (INM), LMU University Hospital, LMU Munich, Munich, Germany
| | - Sven P Wichert
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Anna Litwin
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | | | - Frances Strobl
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Veit Grote
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Tarek Jebrini
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany
| | - Helga P Török
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, LMU Munich, Munich, Germany
| | - Alexander Choukér
- Department of Anesthesiology, Laboratory of Translational Research Stress and Immunity, LMU University Hospital, LMU Munich, Munich, Germany
| | - Berthold Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany
| | - Kristina Adorjan
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstraße 7, 80336, Munich, Germany.
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany.
- Center for International Health (CIH), LMU Munich, Munich, Germany.
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
| | - Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Lindwurmstraße 4, 80337, Munich, Germany.
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland.
| | - Oliver T Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, LMU Munich, Pettenkoferstr. 9a, 80336, Munich, Germany.
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
23
|
Amini A, Klenerman P, Provine NM. Role of mucosal-associated invariant T cells in coronavirus disease 2019 vaccine immunogenicity. Curr Opin Virol 2024; 67:101412. [PMID: 38838550 PMCID: PMC11511680 DOI: 10.1016/j.coviro.2024.101412] [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: 03/18/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are an unconventional T cell population that are highly abundant in humans. They possess a semi-invariant T cell receptor (TCR) that recognises microbial metabolites formed during riboflavin biosynthesis, presented on a nonpolymorphic MHC-like molecule MR1. MAIT cells possess an array of effector functions, including type 1, type 17, and tissue repair activity. Deployment of these functions depends on the stimuli they receive through their TCR and/or cytokine receptors. Strong cytokine signalling, such as in response to vaccination, can bypass TCR triggering and provokes a strong proinflammatory response. Although data are still emerging, multiple aspects of MAIT cell biology are associated with modulation of immunity induced by the coronavirus disease 2019 mRNA and adenovirus vector vaccines. In this review, we will address how MAIT cells may play a role in immunogenicity of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and how these cells can be harnessed as cellular adjuvants.
Collapse
Affiliation(s)
- Ali Amini
- Translational Gastroenterology Unit, Nuffield Department of Medicine - Experimental Medicine, University of Oxford, UK
| | - Paul Klenerman
- Translational Gastroenterology Unit, Nuffield Department of Medicine - Experimental Medicine, University of Oxford, UK; Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, UK; Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK.
| | - Nicholas M Provine
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK; Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, UK.
| |
Collapse
|
24
|
Rajeev D, MacIver NJ. Metformin as a Therapeutic Agent for Obesity-Associated Immune Dysfunction. J Nutr 2024; 154:2534-2542. [PMID: 38972391 DOI: 10.1016/j.tjnut.2024.07.001] [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: 03/15/2024] [Revised: 05/16/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Obesity is associated with impaired immune function, characterized by inflammation, and leading to poor response to infection, impaired vaccine response, increased susceptibility to autoimmune disease, and increased risk of cancer and cancer mortality. Worse, there is evidence that weight loss alone may be insufficient to reverse the immune dysfunction caused by obesity. It is therefore critically important to identify alternative therapeutic approaches to decrease the negative effects of obesity-associated inflammation. In this article, we will review evidence that the antidiabetic drug metformin may be considered as a therapeutic agent for obesity-associated immune dysfunction. Metformin has immunomodulatory effects, stimulating or suppressing the immune response in both a cell-specific and disease-specific manner. Although the mechanism of action of metformin on the immune system remains to be fully elucidated, there is strong evidence that metformin enters select immune cells and disrupts electron transport, leading to both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent effects on immune cell differentiation and cytokine production. These effects of metformin on immune cells have been shown to improve immune responses to infection, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Devika Rajeev
- Department of Nutrition, University of North Carolina at Chapel Hill, NC, United States
| | - Nancie J MacIver
- Department of Nutrition, University of North Carolina at Chapel Hill, NC, United States; Department of Pediatrics, University of North Carolina at Chapel Hill, NC, United States; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, NC, United States.
| |
Collapse
|
25
|
Protopapas K, Thomas K, Moschopoulos CD, Oktapoda E, Marousi E, Marselou E, Stamoulis N, Filis C, Kazakou P, Oikonomopoulou C, Zampetas G, Efstratiadou O, Chavatza K, Kavatha D, Antoniadou A, Papadopoulos A. Breakthrough COVID-19 Infections after Booster SARS-CoV-2 Vaccination in a Greek Cohort of People Living with HIV during the Delta and Omicron Waves. Biomedicines 2024; 12:1614. [PMID: 39062187 PMCID: PMC11274973 DOI: 10.3390/biomedicines12071614] [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: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION Currently approved SARS-CoV-2 vaccines have been proven effective in protecting against severe COVID-19; however, they show variable efficacy against symptomatic infection and disease transmission. We studied the breakthrough COVID-19 infection (BTI) after booster vaccination against SARS-CoV-2 in people living with HIV (PWH). METHODS This was a retrospective, single-center, descriptive cohort study involving PWH, who were followed in the HIV Clinic of "Attikon" University Hospital in Athens, Greece. A BTI was defined as a case of laboratory-confirmed COVID-19 occurring at least 14 days after the third (booster) vaccine dose. RESULTS We studied 733 PWH [males: 89%, mean age: 45.2 ± 11.3 years, mean BMI: 26.1 ± 4.1, HIV stage at diagnosis (CDC classification): A/B/C = 80/9/11%, MSM: 72.6%] with well-controlled HIV infection. At least one comorbidity was recorded in 54% of cases. A history of ≥1 vaccination was reported by 90%, with 75% having been vaccinated with ≥3 vaccines. Four hundred and two (55%) PWH had a history of COVID-19 and 302 (41.2%) had a BTI, with only 15 (3.7%) needing hospitalization. Only one patient was admitted to the ICU, and no death was reported. Regarding BTI after booster dose, increased age (OR = 0.97, 95% CI: 0.96-0.99, per 1-year increase), and COVID-19 infection prior to booster dose (OR = 0.38, 95% CI: 0.21-0.68) were associated with a lower likelihood for BTI, whereas higher BMI (OR = 1.04, 95% CI: 1.01-1.08) and MSM as a mode of HIV transmission were associated with increased risk (OR = 2.59, 95% CI: 1.47-4.56). The incidence rate of total COVID-19 and BTI followed the epidemic curve of the general population, with the highest incidence recorded in June 2022. CONCLUSIONS A significant proportion of PWH with well-controlled HIV infection experienced a BTI, with the majority of them having mild infection. These data, which include the period of Omicron variant predominance, confirm the importance of vaccination in the protection against severe COVID-19.
Collapse
|
26
|
Chambers ES, Cai W, Vivaldi G, Jolliffe DA, Perdek N, Li W, Faustini SE, Gibbons JM, Pade C, Richter AG, Coussens AK, Martineau AR. Influence of individuals' determinants including vaccine type on cellular and humoral responses to SARS-CoV-2 vaccination. NPJ Vaccines 2024; 9:87. [PMID: 38778017 PMCID: PMC11111746 DOI: 10.1038/s41541-024-00878-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Vaccine development targeting SARS-CoV-2 in 2020 was of critical importance in reducing COVID-19 severity and mortality. In the U.K. during the initial roll-out most individuals either received two doses of Pfizer COVID-19 vaccine (BNT162b2) or the adenovirus-based vaccine from Oxford/AstraZeneca (ChAdOx1-nCoV-19). There are conflicting data as to the impact of age, sex and body habitus on cellular and humoral responses to vaccination, and most studies in this area have focused on determinants of mRNA vaccine immunogenicity. Here, we studied a cohort of participants in a population-based longitudinal study (COVIDENCE UK) to determine the influence of age, sex, body mass index (BMI) and pre-vaccination anti-Spike (anti-S) antibody status on vaccine-induced humoral and cellular immune responses to two doses of BNT162b2 or ChAdOx-n-CoV-19 vaccination. Younger age and pre-vaccination anti-S seropositivity were both associated with stronger antibody responses to vaccination. BNT162b2 generated higher neutralising and anti-S antibody titres to vaccination than ChAdOx1-nCoV-19, but cellular responses to the two vaccines were no different. Irrespective of vaccine type, increasing age was also associated with decreased frequency of cytokine double-positive CD4+T cells. Increasing BMI was associated with reduced frequency of SARS-CoV-2-specific TNF+CD8% T cells for both vaccines. Together, our findings demonstrate that increasing age and BMI are associated with attenuated cellular and humoral responses to SARS-CoV-2 vaccination. Whilst both vaccines induced T cell responses, BNT162b2 induced significantly elevated humoral immune response as compared to ChAdOx-n-CoV-19.
Collapse
Affiliation(s)
- Emma S Chambers
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - Weigang Cai
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Giulia Vivaldi
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - David A Jolliffe
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Natalia Perdek
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Wenhao Li
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Sian E Faustini
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Joseph M Gibbons
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentristry, Queen Mary University of London, London, E1 2AT, UK
| | - Corinna Pade
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentristry, Queen Mary University of London, London, E1 2AT, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anna K Coussens
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7925, South Africa
| | - Adrian R Martineau
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| |
Collapse
|
27
|
Cherry N, Adisesh A, Burstyn I, Charlton C, Chen Y, Durand-Moreau Q, Labrèche F, Ruzycki S, Turnbull L, Zadunayski T, Yasui Y. Determinants of SARS-CoV-2 IgG response and decay in Canadian healthcare workers: A prospective cohort study. Vaccine 2024; 42:1168-1178. [PMID: 38278628 DOI: 10.1016/j.vaccine.2024.01.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Healthcare workers (HCWs) from an interprovincial Canadian cohort gave serial blood samples to identify factors associated with anti-receptor binding domain (anti-RBD) IgG response to the SARS-CoV-2 virus. METHODS Members of the HCW cohort donated blood samples four months after their first SARS-CoV-2 immunization and again at 7, 10 and 13 months. Date and type of immunizations and dates of SARS-CoV-2 infection were collected at each of four contacts, together with information on immunologically-compromising conditions and current therapies. Blood samples were analyzed centrally for anti-RBD IgG and anti-nucleocapsid IgG (Abbott Architect, Abbott Diagnostics). Records of immunization and SARS-CoV-2 testing from public health agencies were used to assess the impact of reporting errors on estimates from the random-effects multivariable model fitted to the data. RESULTS 2752 of 4567 vaccinated cohort participants agreed to donate at least one blood sample. Modelling of anti-RBD IgG titer from 8903 samples showed an increase in IgG with each vaccine dose and with first infection. A decrease in IgG titer was found with the number of months since vaccination or infection, with the sharpest decline after the third dose. An immunization regime that included mRNA1273 (Moderna) resulted in higher anti-RBD IgG. Participants reporting multiple sclerosis, rheumatoid arthritis or taking selective immunosuppressants, tumor necrosis factor inhibitors, calcineurin inhibitors and antineoplastic agents had lower anti-RBD IgG. Supplementary analyses showed higher anti-RBD IgG in those reporting side-effects of vaccination, no relation of anti-RBD IgG to obesity and lower titers in women immunized in early or mid-pregnancy. Sensitivity analysis results suggested no important bias in the self-report data. CONCLUSION Creation of a prospective cohort was central to the credibility of results presented here. Serial serology assessments, with longitudinal analysis, provided effect estimates with enhanced accuracy and a clearer understanding of medical and other factors affecting response to vaccination.
Collapse
Affiliation(s)
- Nicola Cherry
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada.
| | - Anil Adisesh
- Division2 Division of Occupational Medicine, Department of Medicine, University of Toronto, C. David Naylor Building, 6 Queen's Park Crescent West, Toronto, ON M5S 3H2, Canada
| | - Igor Burstyn
- Department of Environmental and Occupational Health, Drexel University, Philadelphia, PA 19104, USA
| | - Carmen Charlton
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Yan Chen
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| | - Quentin Durand-Moreau
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - France Labrèche
- Research Department, Institut de recherche Robert-Sauvé en santé et en sécurité du travail, 505 de Maisonneuve Blvd, West Montreal, QC H3A 3C2, Canada
| | - Shannon Ruzycki
- Department of Medicine, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, AB T2N 4N1, Canada
| | - LeeAnn Turnbull
- Alberta Precision Laboratories, 84440 112 St, Edmonton, AB T6G 2I2, Canada
| | - Tanis Zadunayski
- Division of Preventive Medicine, University of Alberta, 5-22 University Terrace, Edmonton, AB T6G 2T4, Canada
| | - Yutaka Yasui
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 735, Memphis, TN 38105, USA
| |
Collapse
|
28
|
Pyrillou K, Humphry M, Kitt LA, Rodgers A, Nus M, Bennett MR, Smith KG, Lyons PA, Mallat Z, Clarke MC. Loss of T follicular regulatory cell-derived IL-1R2 augments germinal center reactions via increased IL-1. JCI Insight 2024; 9:e174005. [PMID: 38329807 PMCID: PMC11143922 DOI: 10.1172/jci.insight.174005] [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: 07/18/2023] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Inappropriate immune activity is key in the pathogenesis of multiple diseases, and it is typically driven by excess inflammation and/or autoimmunity. IL-1 is often the effector owing to its powerful role in both innate and adaptive immunity, and, thus, it is tightly controlled at multiple levels. IL-1R2 antagonizes IL-1, but effects of losing this regulation are unknown. We found that IL-1R2 resolves inflammation by rapidly scavenging free IL-1. Specific IL-1R2 loss in germinal center (GC) T follicular regulatory (Tfr) cells increased the GC response after a first, but not booster, immunization, with an increase in T follicular helper (Tfh) cells, GC B cells, and antigen-specific antibodies, which was reversed upon IL-1 blockade. However, IL-1 signaling is not obligate for GC reactions, as WT and Il1r1-/- mice showed equivalent phenotypes, suggesting that GC IL-1 is normally restrained by IL-1R2. Fascinatingly, germline Il1r2-/- mice did not show this phenotype, but conditional Il1r2 deletion in adulthood recapitulated it, implying that compensation during development counteracts IL-1R2 loss. Finally, patients with ulcerative colitis or Crohn's disease had lower serum IL-1R2. All together, we show that IL-1R2 controls important aspects of innate and adaptive immunity and that IL-1R2 level may contribute to human disease propensity and/or progression.
Collapse
Affiliation(s)
- Katerina Pyrillou
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Melanie Humphry
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Lauren A. Kitt
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Amanda Rodgers
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Meritxell Nus
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Martin R. Bennett
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Kenneth G.C. Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ziad Mallat
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| | - Murray C.H. Clarke
- Section of CardioRespiratory Medicine, Heart and Lung Research Institute, and
| |
Collapse
|
29
|
Jokiranta ST, Miettinen S, Salonen S, Kareinen L, Uusitalo R, Korhonen EM, Virtanen J, Kivistö I, Aaltonen K, Mosselhy DA, Lääveri T, Kantele A, Arstila TP, Jarva H, Vapalahti O, Heinonen S, Kekäläinen E. Stable Levels of Antibodies Against Unrelated Toxoid Vaccines After COVID-19: COVID-19 Infection Does Not Affect Toxoid Vaccine Antibody Levels. Pathog Immun 2024; 8:74-87. [PMID: 38347963 PMCID: PMC10860543 DOI: 10.20411/pai.v8i2.627] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024] Open
Abstract
Background Lymphopenia is common in COVID-19. This has raised concerns that COVID-19 could affect the immune system akin to measles infection, which causes immune amnesia and a reduction in protective antibodies. Methods We recruited COVID-19 patients (n = 59) in Helsinki, Finland, and collected plasma samples on 2 to 3 occasions during and after infection. We measured IgG antibodies to diphtheria toxin, tetanus toxoid, and pertussis toxin, along with total IgG, SARS-CoV-2 spike protein IgG, and neutralizing antibodies. We also surveyed the participants for up to 17 months for long-term impaired olfaction as a proxy for prolonged post-acute COVID-19 symptoms. Results No significant differences were found in the unrelated vaccine responses while the serological response against COVID-19 was appropriate. During the acute phase of the disease, the SARSCoV-2 IgG levels were lower in outpatients when compared to inpatients. SARS-CoV-2 serology kinetics matched expectations. In the acute phase, anti-tetanus and anti-diphtheria IgG levels were lower in patients with prolonged impaired olfaction during follow up than in those without. Conclusions We could not detect significant decline in overall humoral immunity during or after COVID-19 infection. In severe COVID-19, there appears to be a temporary decline in total IgG levels.
Collapse
Affiliation(s)
- Suvi T. Jokiranta
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Simo Miettinen
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Sami Salonen
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Lauri Kareinen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Finnish Food Authority, Helsinki, Finland
| | - Ruut Uusitalo
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Essi M. Korhonen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jenni Virtanen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilkka Kivistö
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kirsi Aaltonen
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dina A. Mosselhy
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Microbiological Unit, Fish Diseases Department, Animal Health Research Institute, ARC, Dokki, Giza 12618, Egypt
| | - Tinja Lääveri
- Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Aalto University, Department of Computer Science, School of Science, Espoo, Finland
| | - Anu Kantele
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Meilahti Infectious Diseases and Vaccine Research Center, MeiVac, Department of Infectious Diseases, University of Helsinki, Helsinki, Finland
| | - T. Petteri Arstila
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hanna Jarva
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Viral Zoonosis Research Unit, Medicum, Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
- Department of Virology, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Santtu Heinonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Eliisa Kekäläinen
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
30
|
Bedston S, Almaghrabi F, Patterson L, Agrawal U, Woolford L, Anand SN, Joy M, Crawford A, Goudie R, Byford R, Abbasizanjani H, Smith D, Laidlaw L, Akbari A, Sullivan C, Bradley DT, Lyons RA, de Lusignan S, Hobbs FR, Robertson C, Sheikh SA, Shi T. Risk of severe COVID-19 outcomes after autumn 2022 COVID-19 booster vaccinations: a pooled analysis of national prospective cohort studies involving 7.4 million adults in England, Northern Ireland, Scotland and Wales. THE LANCET REGIONAL HEALTH. EUROPE 2024; 37:100816. [PMID: 38162515 PMCID: PMC10757260 DOI: 10.1016/j.lanepe.2023.100816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/17/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024]
Abstract
Background UK COVID-19 vaccination policy has evolved to offering COVID-19 booster doses to individuals at increased risk of severe Illness from COVID-19. Building on our analyses of vaccine effectiveness of first, second and initial booster doses, we aimed to identify individuals at increased risk of severe outcomes (i.e., COVID-19 related hospitalisation or death) post the autumn 2022 booster dose. Methods We undertook a national population-based cohort analysis across all four UK nations through linked primary care, vaccination, hospitalisation and mortality data. We included individuals who received autumn 2022 booster doses of BNT162b2 (Comirnaty) or mRNA-1273 (Spikevax) during the period September 1, 2022 to December 31, 2022 to investigate the risk of severe COVID-19 outcomes. Cox proportional hazard models were used to estimate adjusted hazard ratios (aHR) and 95% confidence intervals (CIs) for the association between demographic and clinical factors and severe COVID-19 outcomes after the autumn booster dose. Analyses were adjusted for age, sex, body mass index (BMI), deprivation, urban/rural areas and comorbidities. Stratified analyses were conducted by vaccine type. We then conducted a fixed-effect meta-analysis to combine results across the four UK nations. Findings Between September 1, 2022 and December 31, 2022, 7,451,890 individuals ≥18 years received an autumn booster dose. 3500 had severe COVID-19 outcomes (2.9 events per 1000 person-years). Being male (male vs female, aHR 1.41 (1.32-1.51)), older adults (≥80 years vs 18-49 years; 10.43 (8.06-13.50)), underweight (BMI <18.5 vs BMI 25.0-29.9; 2.94 (2.51-3.44)), those with comorbidities (≥5 comorbidities vs none; 9.45 (8.15-10.96)) had a higher risk of COVID-19 hospitalisation or death after the autumn booster dose. Those with a larger household size (≥11 people within household vs 2 people; 1.56 (1.23-1.98)) and from more deprived areas (most deprived vs least deprived quintile; 1.35 (1.21-1.51)) had modestly higher risks. We also observed at least a two-fold increase in risk for those with various chronic neurological conditions, including Down's syndrome, immunodeficiency, chronic kidney disease, cancer, chronic respiratory disease, or cardiovascular disease. Interpretation Males, older individuals, underweight individuals, those with an increasing number of comorbidities, from a larger household or more deprived areas, and those with specific underlying health conditions remained at increased risk of COVID-19 hospitalisation and death after the autumn 2022 vaccine booster dose. There is now a need to focus on these risk groups for investigating immunogenicity and efficacy of further booster doses or therapeutics. Funding National Core Studies-Immunity, UK Research and Innovation (Medical Research Council and Economic and Social Research Council), Health Data Research UK, the Scottish Government, and the University of Edinburgh.
Collapse
Affiliation(s)
- Stuart Bedston
- Population Data Science, Swansea University Medical School, Faculty of Medicine, Health, and Life Science, Swansea University, Swansea, UK
| | - Fatima Almaghrabi
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Lynsey Patterson
- Centre for Public Health, Queen's University Belfast, Belfast, UK
- Public Health Agency, Belfast, UK
| | - Utkarsh Agrawal
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Lana Woolford
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Sneha N. Anand
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Mark Joy
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Anna Crawford
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Rosalind Goudie
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Rachel Byford
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Hoda Abbasizanjani
- Population Data Science, Swansea University Medical School, Faculty of Medicine, Health, and Life Science, Swansea University, Swansea, UK
| | - Deb Smith
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Lynn Laidlaw
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ashley Akbari
- Population Data Science, Swansea University Medical School, Faculty of Medicine, Health, and Life Science, Swansea University, Swansea, UK
| | | | - Declan T. Bradley
- Centre for Public Health, Queen's University Belfast, Belfast, UK
- Public Health Agency, Belfast, UK
| | - Ronan A. Lyons
- Population Data Science, Swansea University Medical School, Faculty of Medicine, Health, and Life Science, Swansea University, Swansea, UK
| | - Simon de Lusignan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - F.D. Richard Hobbs
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Chris Robertson
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, Scotland, UK
- Public Health Scotland, Glasgow, Scotland, UK
| | - Sir Aziz Sheikh
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ting Shi
- Usher Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
31
|
Lee SK, Lim Y, Jeong S, Han HW. COVID-19-related cardiovascular disease risk due to weight gain: a nationwide cohort study. Eur J Med Res 2024; 29:2. [PMID: 38167158 PMCID: PMC10762936 DOI: 10.1186/s40001-023-01569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) is a significant contributor to morbidity and mortality worldwide, with CVD and post-acute COVID-19 associated CVD increasing. It remains unknown whether COVID-19 patients with weight gain are at a high risk for CVD events. Therefore, the primary objective of this study is to investigate the association between weight control and the risk of CVD following COVID-19. METHODS The study included 2,024,728 adults who participated in two rounds of health screening between 2017 and 2020. The final cohort, which included 70,996 participants in the COVID-19 group and 212,869 participants in the control group. The adjusted hazard ratio of BMI change to CVD risk was calculated using Cox proportional hazards regression. RESULTS We identified a total of 2869 cases of CVD (861 events for COVID-19 group and 2,008 events for the control group). Compared to individuals with a stable BMI, COVID-19 patients without obesity had an increased risk of CVD (adjusted hazard ratio [aHR] = 2.28; 95% confidence interval [CI], 1.15-4.53; p-value = 0.018). Additionally, non-COVID-19 patients with obesity also exhibited a higher risk of CVD (aHR = 1.58; 95% CI, 1.01-2.47; p-value = 0.046). CONCLUSION In conclusion, people who gained weight during the pandemic, regardless of their weight category, had a significantly higher risk of CVD associated with COVID-19 compared to those who maintained their weight before the pandemic.
Collapse
Affiliation(s)
- Su Kyoung Lee
- Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Yohwan Lim
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, 335 Pangyo-Ro, Seongnam, 13448, Republic of Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, 335 Pangyo-Ro, Seongnam, 13448, Republic of Korea.
| | - Hyun Wook Han
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, 335 Pangyo-Ro, Seongnam, 13448, Republic of Korea.
| |
Collapse
|
32
|
Vashishtha VM, Kumar P. The durability of vaccine-induced protection: an overview. Expert Rev Vaccines 2024; 23:389-408. [PMID: 38488132 DOI: 10.1080/14760584.2024.2331065] [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: 01/18/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
INTRODUCTION Current vaccines vary widely in both their efficacy against infection and disease, and the durability of the efficacy. Some vaccines provide practically lifelong protection with a single dose, while others provide only limited protection following annual boosters. What variables make vaccine-induced immune responses last? Can breakthroughs in these factors and technologies help us produce vaccines with better protection and fewer doses? The durability of vaccine-induced protection is now a hot area in vaccinology research, especially after COVID-19 vaccines lost their luster. It has fueled discussion on the eventual utility of existing vaccines to society and bolstered the anti-vaxxer camp. To sustain public trust in vaccines, lasting vaccines must be developed. AREAS COVERED This review summarizes licensed vaccines' protection. It analyses immunological principles and vaccine and vaccinee parameters that determine longevity of antibodies. The review concludes with challenges and the way forward to improve vaccine durability. EXPERT OPINION Despite enormous advances, we still lack essential markers and reliable correlates of lasting protection. Most research has focused on humoral immune responses, but we must also focus on innate, mucosal, and cellular responses - their assessment, correlates, determinants, and novel adjuvants. Suitable vaccine designs and platforms for durable immunity must be found.
Collapse
Affiliation(s)
- Vipin M Vashishtha
- Department of Pediatrics, Mangla Hospital & Research Center, Shakti Chowk, Bijnor, Uttar Pradesh, India
| | - Puneet Kumar
- Department of Pediatrician, Kumar Child Clinic, New Delhi, India
| |
Collapse
|
33
|
Bian L, Fu Q, Gan Z, Wu Z, Song Y, Xiong Y, Hu F, Zheng L. Fluorescence-Quenching Lateral Flow Immunoassay for "Turn-On" and Sensitive Detection of Anti-SARS-Cov-2 Neutralizing Antibodies in Human Serum. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305774. [PMID: 38032112 PMCID: PMC10811470 DOI: 10.1002/advs.202305774] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/26/2023] [Indexed: 12/01/2023]
Abstract
The titer of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies (NAbs) in the human body is an essential reference for evaluating the acquired protective immunity and resistance to SARS-CoV-2 infection. In this study, a fluorescence-quenching lateral flow immunoassay (FQ-LFIA) is established for quantitative detection of anti-SARS-CoV-2 NAbs in the sera of individuals who are vaccinated or infected within 10 min. The ultrabright aggregation-induced emission properties encapsulated in nanoparticles, AIE490 NP, are applied in the established FQ-LFIA with gold nanoparticles to achieve a fluorescence "turn-on" competitive immunoassay. Under optimized conditions, the FQ-LFIA quantitatively detected 103 positive and 50 negative human serum samples with a limit of detection (LoD) of 1.29 IU mL-1 . A strong correlation is present with the conventional pseudovirus-based virus neutralization test (R2 = 0.9796, P < 0.0001). In contrast, the traditional LFIA with a "turn-off" mode can only achieve a LoD of 11.06 IU mL-1 . The FQ-LFIA showed excellent sensitivity to anti-SARS-CoV-2 NAbs. The intra- and inter-assay precisions of the established method are below 15%. The established FQ-LFIA has promising potential as a rapid and quantitative method for detecting anti-SARS-CoV-2 NAbs. FQ-LFIA can also be used to detect various biomarkers.
Collapse
Affiliation(s)
- Lun Bian
- Biomaterials Research CenterSchool of Biomedical EngineeringSouthern Medical UniversityGuangzhou510515China
| | - Qiangqiang Fu
- Department of Laboratory MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Zhuoheng Gan
- Biomaterials Research CenterSchool of Biomedical EngineeringSouthern Medical UniversityGuangzhou510515China
| | - Ze Wu
- Department of Laboratory MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yuchen Song
- Biomaterials Research CenterSchool of Biomedical EngineeringSouthern Medical UniversityGuangzhou510515China
| | - Yufeng Xiong
- Department of Laboratory MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Fang Hu
- Biomaterials Research CenterSchool of Biomedical EngineeringSouthern Medical UniversityGuangzhou510515China
- Division of Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510282China
| | - Lei Zheng
- Department of Laboratory MedicineNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
34
|
Beyerlein A, Weigert M, Katz K, Küchenhoff H, Hartl W. Long-Term Trends in the Protection Against Severe Courses of COVID-19 by Vaccination. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:873-878. [PMID: 37963066 PMCID: PMC10859745 DOI: 10.3238/arztebl.m2023.0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND The long-term course of protection against severe COVID-19 courses by vaccine-induced or hybrid immunity in Germany is unclear. METHODS We studied 146 457 cases aged 60-99 years in the German federal state of Bavaria who were immunized against COVID-19 and tested positive for it from February 2022 to January 2023. We calculated adjusted hazard ratios for a severe course (hospitalization or death due to COVID-19) for different intervals between the onset of full primary or booster immunity and the date of the infection. RESULTS 3342 (2.3%) severe courses of COVID-19 were observed in the first 60 days after the infection. The risk of a severe course rose with the interval between the onset of immune protection and the infection (adjusted hazard ratios and 95% confidence intervals at 6, 9, 12, and 15 months: 1.14 [1.08; 1.20]; 1.33 [1.24; 1.42]; 1.39 [1.25; 1.54]; 1.61 [1.35; 1.93]). The risk rose more slowly when mRNA-based vaccines were used exclusively. In a previous study, we observed 82% initial efficacy in cases aged 60 and above who received a booster vaccination (compared to unvaccinated cases) and an absolute risk reduction of 2.1%. If one extrapolates these findings to the current study, the residual efficacy and absolute risk reduction are found to be approximately 71% and 1.8% (respectively) at 6 months, and 32% and 0.8% at 15 months. CONCLUSION These results indicate that, during the Omicron wave, the protection of older persons against a severe COVID-19 course gradually declined from six months after vaccination onward. The limitations of this study include confounders that could not be taken into account, possible misclassification of the cause of death, and selection bias due to missing information about vaccination status and severe COVID-19 courses.
Collapse
Affiliation(s)
| | - Maximilian Weigert
- Statistical Consulting Unit StaBLab, CODAG, Institute of Statistics, LMU Munich
- Munich Center for Machine Learning (MCML), Munich
| | - Katharina Katz
- Bavarian Health and Food Safety Authority, Oberschleißheim
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, CODAG, Institute of Statistics, LMU Munich
- Munich Center for Machine Learning (MCML), Munich
- *Joint last authors
| | - Wolfgang Hartl
- Surgical Intensive Care Medicine, Department of General, Visceral, and Transplantation Surgery, University Hospital, Großhadern Campus, LMU Munich
- *Joint last authors
| |
Collapse
|
35
|
Tong MZW, Sng JDJ, Carney M, Cooper L, Brown S, Lineburg KE, Chew KY, Collins N, Ignacio K, Airey M, Burr L, Joyce BA, Jayasinghe D, McMillan CLD, Muller DA, Adhikari A, Gallo LA, Dorey ES, Barrett HL, Gras S, Smith C, Good‐Jacobson K, Short KR. Elevated BMI reduces the humoral response to SARS-CoV-2 infection. Clin Transl Immunology 2023; 12:e1476. [PMID: 38050635 PMCID: PMC10693902 DOI: 10.1002/cti2.1476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
Objective Class III obesity (body mass index [BMI] ≥ 40 kg m-2) significantly impairs the immune response to SARS-CoV-2 vaccination. However, the effect of an elevated BMI (≥ 25 kg m-2) on humoral immunity to SARS-CoV-2 infection and COVID-19 vaccination remains unclear. Methods We collected blood samples from people who recovered from SARS-CoV-2 infection approximately 3 and 13 months of post-infection (noting that these individuals were not exposed to SARS-CoV-2 or vaccinated in the interim). We also collected blood samples from people approximately 5 months of post-second dose COVID-19 vaccination (the majority of whom did not have a prior SARS-CoV-2 infection). We measured their humoral responses to SARS-CoV-2, grouping individuals based on a BMI greater or less than 25 kg m-2. Results Here, we show that an increased BMI (≥ 25 kg m-2), when accounting for age and sex differences, is associated with reduced antibody responses after SARS-CoV-2 infection. At 3 months of post-infection, an elevated BMI was associated with reduced antibody titres. At 13 months of post-infection, an elevated BMI was associated with reduced antibody avidity and a reduced percentage of spike-positive B cells. In contrast, no significant association was noted between a BMI ≥ 25 kg m-2 and humoral immunity to SARS-CoV-2 at 5 months of post-secondary vaccination. Conclusions Taken together, these data showed that elevated BMI is associated with an impaired humoral immune response to SARS-CoV-2 infection. The impairment of infection-induced immunity in individuals with a BMI ≥ 25 kg m-2 suggests an added impetus for vaccination rather than relying on infection-induced immunity.
Collapse
Affiliation(s)
- Marcus ZW Tong
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Julian DJ Sng
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Meagan Carney
- School of Mathematics and PhysicsThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Cooper
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Samuel Brown
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Katie E Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Keng Yih Chew
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Neve Collins
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Kirsten Ignacio
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Megan Airey
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Lucy Burr
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
- Department of Respiratory MedicineMater HealthBrisbaneQLDAustralia
| | - Briony A Joyce
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Dhilshan Jayasinghe
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Christopher LD McMillan
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - David A Muller
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| | - Anurag Adhikari
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Linda A Gallo
- School of HealthUniversity of the Sunshine CoastPetrieQLDAustralia
| | - Emily S Dorey
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
| | - Helen L Barrett
- Mater ResearchThe University of QueenslandSouth BrisbaneQLDAustralia
- University of New South Wales MedicineKensingtonNSWAustralia
- Obstetric MedicineRoyal Hospital for WomenRandwickNSWAustralia
| | - Stephanie Gras
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Department of Biochemistry and ChemistryLa Trobe Institute for Molecular Science, La Trobe UniversityBundooraVICAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Infection and Inflammation ProgramQIMR Berghofer Medical Research InstituteHerstonQLDAustralia
| | - Kim Good‐Jacobson
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVICAustralia
- Immunity Program, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Kirsty R Short
- School of Chemistry and Molecular BiosciencesThe University of QueenslandSt LuciaQLDAustralia
- Australian Infectious Diseases Research CentreThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
36
|
Moser J, Emous M, Heeringa P, Rodenhuis-Zybert IA. Mechanisms and pathophysiology of SARS-CoV-2 infection of the adipose tissue. Trends Endocrinol Metab 2023; 34:735-748. [PMID: 37673763 DOI: 10.1016/j.tem.2023.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023]
Abstract
Obesity is an independent risk factor for severe COVID-19, yet there remains a lack of consensus on the mechanisms underlying this relationship. A hypothesis that has garnered considerable attention suggests that SARS-CoV-2 disrupts adipose tissue function, either through direct infection or by indirect mechanisms. Indeed, recent reports have begun to shed some light on the important role that the adipose tissue plays during the acute phase of infection, as well as mediating long-term sequelae. In this review, we examine the evidence of extrapulmonary dissemination of SARS-CoV-2 to the adipose tissue. We discuss the mechanisms, acute and long-term implications, and possible management strategies to limit or ameliorate severe disease and long-term metabolic disturbances.
Collapse
Affiliation(s)
- Jill Moser
- Department of Critical Care, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Marloes Emous
- Center Obesity Northern Netherlands (CON), Department of Surgery, Medical Center Leeuwarden, Leeuwarden, the Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Izabela A Rodenhuis-Zybert
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
37
|
O'Meara TR, Nanishi E, McGrath ME, Barman S, Dong D, Dillen C, Menon M, Seo HS, Dhe-Paganon S, Ernst RK, Levy O, Frieman MB, Dowling DJ. Reduced SARS-CoV-2 mRNA vaccine immunogenicity and protection in mice with diet-induced obesity and insulin resistance. J Allergy Clin Immunol 2023; 152:1107-1120.e6. [PMID: 37595760 PMCID: PMC10841117 DOI: 10.1016/j.jaci.2023.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Obesity and type 2 diabetes mellitus (T2DM) are associated with an increased risk of severe outcomes from infectious diseases, including coronavirus disease 2019. These conditions are also associated with distinct responses to immunization, including an impaired response to widely used severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccines. OBJECTIVE We sought to establish a connection between reduced immunization efficacy via modeling the effects of metabolic diseases on vaccine immunogenicity that is essential for the development of more effective vaccines for this distinct vulnerable population. METHODS A murine model of diet-induced obesity and insulin resistance was used to model the effects of comorbid T2DM and obesity on vaccine immunogenicity and protection. RESULTS Mice fed a high-fat diet (HFD) developed obesity, hyperinsulinemia, and glucose intolerance. Relative to mice fed a normal diet, HFD mice vaccinated with a SARS-CoV-2 mRNA vaccine exhibited significantly lower anti-spike IgG titers, predominantly in the IgG2c subclass, associated with a lower type 1 response, along with a 3.83-fold decrease in neutralizing titers. Furthermore, enhanced vaccine-induced spike-specific CD8+ T-cell activation and protection from lung infection against SARS-CoV-2 challenge were seen only in mice fed a normal diet but not in HFD mice. CONCLUSIONS The study demonstrated impaired immunity following SARS-CoV-2 mRNA immunization in a murine model of comorbid T2DM and obesity, supporting the need for further research into the basis for impaired anti-SARS-CoV-2 immunity in T2DM and investigation of novel approaches to enhance vaccine immunogenicity among those with metabolic diseases.
Collapse
Affiliation(s)
- Timothy R O'Meara
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Etsuro Nanishi
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Marisa E McGrath
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - Soumik Barman
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Danica Dong
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Carly Dillen
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - Manisha Menon
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Mass; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Mass
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, Md
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Broad Institute of MIT and Harvard, Cambridge, Mass
| | - Matthew B Frieman
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Md
| | - David J Dowling
- Precision Vaccines Program, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
38
|
Ferreira IATM, Lee CYC, Foster WS, Abdullahi A, Dratva LM, Tuong ZK, Stewart BJ, Ferdinand JR, Guillaume SM, Potts MOP, Perera M, Krishna BA, Peñalver A, Cabantous M, Kemp SA, Ceron-Gutierrez L, Ebrahimi S, Lyons P, Smith KGC, Bradley J, Collier DA, McCoy LE, van der Klaauw A, Thaventhiran JED, Farooqi IS, Teichmann SA, MacAry PA, Doffinger R, Wills MR, Linterman MA, Clatworthy MR, Gupta RK. Atypical B cells and impaired SARS-CoV-2 neutralization following heterologous vaccination in the elderly. Cell Rep 2023; 42:112991. [PMID: 37590132 DOI: 10.1016/j.celrep.2023.112991] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
Suboptimal responses to a primary vaccination course have been reported in the elderly, but there is little information regarding the impact of age on responses to booster third doses. Here, we show that individuals 70 years or older (median age 73, range 70-75) who received a primary two-dose schedule with AZD1222 and booster third dose with mRNA vaccine achieve significantly lower neutralizing antibody responses against SARS-CoV-2 spike pseudotyped virus compared with those younger than 70 (median age 66, range 54-69) at 1 month post booster. Impaired neutralization potency and breadth post third dose in the elderly is associated with circulating "atypical" spike-specific B cells expressing CD11c and FCRL5. However, when considering individuals who received three doses of mRNA vaccine, we did not observe differences in neutralization or enrichment in atypical B cells. This work highlights the finding that AdV and mRNA COVID-19 vaccine formats differentially instruct the memory B cell response.
Collapse
Affiliation(s)
- Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Colin Y C Lee
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - William S Foster
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - John R Ferdinand
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Stephane M Guillaume
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Martin O P Potts
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Benjamin A Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ana Peñalver
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Mia Cabantous
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Soraya Ebrahimi
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Paul Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Bradley
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami A Collier
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Agatha van der Klaauw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - Paul A MacAry
- National University of Singapore, Singapore, Singapore
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Mark R Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michelle A Linterman
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
39
|
Chauvin C, Retnakumar SV, Bayry J. Obesity negatively impacts maintenance of antibody response to COVID-19 vaccines. Cell Rep Med 2023; 4:101117. [PMID: 37467723 PMCID: PMC10394250 DOI: 10.1016/j.xcrm.2023.101117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Severe obesity accelerates the decline of neutralizing antibodies to COVID-19 vaccines contributing to increased risk of hospitalization from breakthrough SARS-CoV-2 infections.1 These findings have repercussion on the vaccination policy for SARS-CoV-2 variants and other infectious diseases like influenza in obese population.
Collapse
Affiliation(s)
- Camille Chauvin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Sruthi Vijaya Retnakumar
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India.
| |
Collapse
|
40
|
COVID vaccines falter in people with severe obesity. Nature 2023; 617:654. [PMID: 37198470 DOI: 10.1038/d41586-023-01590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
|