1
|
Gong Y, Zhan H, Wei N, Liu M, Liu Y, Guan P, Xie Y, Deng Y, Pu Q, Lou X, Wang X, Zhang R, Wang P, Jin X, Wang X, Xu Z, Gao L, Wang X, He S, Lu Y, Hu M, Li W, Zheng K, Peng Y, Lei P, Xu H, Shi Y, Qin J, Hu H, Zhang H, Dai L. Acetylation profiling by Iseq-Kac reveals insights into HSC aging and lineage decision. Nat Chem Biol 2025:10.1038/s41589-025-01916-1. [PMID: 40419771 DOI: 10.1038/s41589-025-01916-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025]
Abstract
Profiling post-translational modifications face challenges with low-input samples. We developed Iseq-Kac (internal standard-assisted enrichment-free approach for high-throughput quantitative analysis of lysine acetylation) to profile the acetylome in as few as 103-104 cells. By using a hyperacetylated internal standard, Iseq-Kac can be used in mass spectrometry (MS) to enhance MS1 signals and facilitate MS2 fragmentation of acetylated peptides. Using Iseq-Kac, we quantified 675-1,471 acetylated peptides per analysis from 104 hematopoietic stem cells (HSCs) or multipotent progenitors. Validation by targeted MS, site-specific antibodies and functional assays linked aging-related proteome and acetylome changes to HSC lineage decision. A pronounced decrease in acetylation at H4 lysine 77 (H4K77ac) was observed in aged HSCs, linked to histone deacetylase 3 (HDAC3) activity. HDAC3 inhibition or knockdown in HSCs significantly promoted lymphocyte differentiation. Mimicking H4K77ac through H4K77Q expression enhanced B cell differentiation while repressing myeloid differentiation. Overall, Iseq-Kac enables robust low-input acetylome profiling and reveals epigenetic mechanisms underlying lineage skewing in aged HSCs.
Collapse
Affiliation(s)
- Yanqiu Gong
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huiwen Zhan
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Wei
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Min Liu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Liu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Pengbo Guan
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yusi Xie
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yujun Deng
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qianlun Pu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxian Lou
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaodong Wang
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rou Zhang
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Wang
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Xiuxiu Jin
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuxuan Wang
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiqiang Xu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Gao
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyuan Wang
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Siyu He
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Lu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Hu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wanmeng Li
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Zheng
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Peng
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lei
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yujun Shi
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Qin
- State Key Laboratory of Proteomics, Institute of Lifeomics, Beijing, China
| | - Hongbo Hu
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Chongqing International Institute for Immunology, Chongqing, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| | - Huiyuan Zhang
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Lunzhi Dai
- Center for Hematology and Immunology and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
2
|
Ishmatov A. Age, gender, and race differences in nasal morphology: Linking air conditioning and filtration efficiency to disparities in air pollution health outcomes and COVID-19 mortality. CHEMOSPHERE 2025; 377:144358. [PMID: 40153988 DOI: 10.1016/j.chemosphere.2025.144358] [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: 12/24/2024] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
COVID-19 mortality disparities underscore the critical role of environmental factors, age, sex, and racial demographics. This study investigates how individual variations in nasal morphology - specifically its air conditioning (temperature and humidity regulation) and filtration functions - may influence respiratory health and contribute to differential COVID-19 outcomes. Analysis reveals significant differences in nasal structure and function across racial, sex, and age groups, demonstrating associations with disparities in respiratory vulnerability to environmental stressors such as air pollution, infectious aerosols, and climatic conditions. Specifically, wider nasal cavities (more common in certain populations), larger male nasal passages, and age-related changes like mucosal atrophy and increased endonasal volume impair air conditioning and filtration efficiency. These morphological variations influence the nose's protective capacity, which is critical for shielding the middle and lower airways from environmental exposures. Populations with inherently reduced nasal filtration and conditioning efficiency demonstrate higher vulnerability, aligning with U.S. mortality patterns for both COVID-19 and air pollution across demographic groups. This suggests a direct link between nasal anatomy and population-level health disparities. These findings provide novel insights into the role of nasal anatomy in mediating respiratory health disparities by modulating individual responses to environmental exposures, air pollution, and pathogens. They highlight the need to address critical gaps in understanding how airway characteristics influence susceptibility to environmental stressors and to develop targeted interventions aimed at reducing health disparities.
Collapse
Affiliation(s)
- Alexander Ishmatov
- Institute for Engineering and Environmental Safety, Togliatti State University, Belorusskaya St, 14, Togliatti, 445020, Russia.
| |
Collapse
|
3
|
Xu Z, Li Z, Zhang R, Peng G, Ge J, Luo S, Liu C, Zeng L, Deng J. Pronounced effects of the sepsis-obesity paradox in elderly and male individuals without septic shock and the role of immune-inflammatory status: an analysis of MIMIC-IV data. BMC Infect Dis 2025; 25:545. [PMID: 40247198 PMCID: PMC12004873 DOI: 10.1186/s12879-025-10938-y] [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: 09/12/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Obesity has been shown to reduce short-term mortality in sepsis patients, but the main subgroups and its role in immune-related inflammatory status require further research. The aim of this study was to identify the primary beneficiaries of the sepsis-obesity paradox and to investigate the involvement of immune-inflammatory status. METHODS In this study, we analyzed data from 6602 sepsis patients from the MIMIC-IV database. Body mass index (BMI) was divided into quartiles, and mortality rates were assessed for each interval. Logistic trend tests and subgroup and restricted cubic spline (RCS) analyses were performed. Blood biochemical indicators were compared across different BMI ranges and between survivors and non-survivors. The receiver operating characteristic (ROC) curve for 28-day mortality was also evaluated. RESULTS The 28-day mortality of sepsis patients followed a U-shaped pattern with increasing BMI. Trend analysis confirmed that BMI was a significant risk factor for 28-day mortality (p < 0.05). Subgroup analysis revealed an interactive effect of BMI on 28-day mortality in elderly (≥ 65 years old), male, and non-septic shock individuals (p < 0.05). A higher BMI was associated with an increased lymphocyte proportion and decreased neutrophil proportion, neutrophil-to-lymphocyte ratio (NLR), and systemic immune-inflammation index (SII) (p < 0.05). Compared with survivors, non-survivors had lower lymphocyte proportions and higher neutrophil proportions, NLRs, and SIIs. ROC analysis revealed that the lymphocyte and neutrophil proportions, NLR, and SII had predictive value for 28-day mortality. Subgroup and RCS analyses revealed that increased BMI was associated with reduced 28-day mortality in sepsis patients, mainly in elderly, male, and septic shock individuals, with protective BMIs ranging from 27.8 ~ 41.7 kg/cm2, 28.4 ~ 37.7 kg/cm2, and > 28.6 kg/cm2, respectively. CONCLUSIONS The sepsis-obesity paradox significantly affects elderly (≥ 65 years old), male, and non-septic shock individuals, displaying a U-shaped pattern for 28-day mortality. BMI may mediate this phenomenon by influencing the body's immune-inflammatory status.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
- Department of Orthopedics, Guihang Guiyang Hospital, Guiyang, 550025, China
| | - Zhuojie Li
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Ruguo Zhang
- Department of Orthopedics, Guihang Guiyang Hospital, Guiyang, 550025, China
| | - Guoxuan Peng
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jingzhong Ge
- Department of Orthopedics, Guihang Guiyang Hospital, Guiyang, 550025, China
| | - Shanpeng Luo
- Department of Orthopedics, The Fifth Hospital of Guiyang City, Guiyang, 550004, China
| | - Chen Liu
- Department of Orthopedics, Guihang Guiyang Hospital, Guiyang, 550025, China
| | - Ling Zeng
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400042, China.
| | - Jin Deng
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
4
|
Bracken OV, De Maeyer RPH, Akbar AN. Enhancing immunity during ageing by targeting interactions within the tissue environment. Nat Rev Drug Discov 2025; 24:300-315. [PMID: 39875569 DOI: 10.1038/s41573-024-01126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Immunity declines with age. This results in a higher risk of age-related diseases, diminished ability to respond to new infections and reduced response to vaccines. The causes of this immune dysfunction are cellular senescence, which occurs in both lymphoid and non-lymphoid tissue, and chronic, low-grade inflammation known as 'inflammageing'. In this Review article, we highlight how the processes of inflammation and senescence drive each other, leading to loss of immune function. To break this cycle, therapies are needed that target the interactions between the altered tissue environment and the immune system instead of targeting each component alone. We discuss the relative merits and drawbacks of therapies that are directed at eliminating senescent cells (senolytics) and those that inhibit inflammation (senomorphics) in the context of tissue niches. Furthermore, we discuss therapeutic strategies designed to directly boost immune cell function and improve immune surveillance in tissues.
Collapse
Affiliation(s)
| | - Roel P H De Maeyer
- Division of Medicine, University College London, London, UK
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK.
| |
Collapse
|
5
|
Ozawa T, Chubachi S, Namkoong H, Nemoto S, Ikegami R, Asakura T, Tanaka H, Lee H, Fukushima T, Azekawa S, Otake S, Nakagawara K, Watase M, Masaki K, Kamata H, Harada N, Ueda T, Ueda S, Ishiguro T, Arimura K, Saito F, Yoshiyama T, Nakano Y, Muto Y, Suzuki Y, Edahiro R, Murakami K, Sato Y, Okada Y, Koike R, Ishii M, Hasegawa N, Kitagawa Y, Tokunaga K, Kimura A, Miyano S, Ogawa S, Kanai T, Fukunaga K, Imoto S. Predicting coronavirus disease 2019 severity using explainable artificial intelligence techniques. Sci Rep 2025; 15:9459. [PMID: 40108236 PMCID: PMC11923144 DOI: 10.1038/s41598-025-85733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/06/2025] [Indexed: 03/22/2025] Open
Abstract
Predictive models for determining coronavirus disease 2019 (COVID-19) severity have been established; however, the complexity of the interactions among factors limits the use of conventional statistical methods. This study aimed to establish a simple and accurate predictive model for COVID-19 severity using an explainable machine learning approach. A total of 3,301 patients ≥ 18 years diagnosed with COVID-19 between February 2020 and October 2022 were included. The discovery cohort comprised patients whose disease onset fell before October 1, 2020 (N = 1,023), and the validation cohort comprised the remaining patients (N = 2,278). Pointwise linear and logistic regression models were used to extract 41 features. Reinforcement learning was used to generate a simple model with high predictive accuracy. The primary evaluation was the area under the receiver operating characteristic curve (AUC). The predictive model achieved an AUC of ≥ 0.905 using four features: serum albumin levels, lactate dehydrogenase levels, age, and neutrophil count. The highest AUC value was 0.906 (sensitivity, 0.842; specificity, 0.811) in the discovery cohort and 0.861 (sensitivity, 0.804; specificity, 0.675) in the validation cohort. Simple and well-structured predictive models were established, which may aid in patient management and the selection of therapeutic interventions.
Collapse
Affiliation(s)
- Takuya Ozawa
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Shota Nemoto
- Industrial and Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Ryo Ikegami
- Industrial and Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Clinical Medicine (Laboratory of Bioregulatory Medicine), Kitasato University School of Pharmacy, Tokyo, Japan
- Department of Respiratory Medicine, Kitasato University, Kitasato Institute Hospital, Tokyo, Japan
| | - Hiromu Tanaka
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ho Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Fukushima
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shiro Otake
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Nakagawara
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mayuko Watase
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Katsunori Masaki
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Norihiro Harada
- Department of Respiratory Medicine, Faculty of Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tetsuya Ueda
- Department of Respiratory Medicine, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Soichiro Ueda
- JCHO (Japan Community Health Care Organization, Internal Medicine, Saitama Medical Center, Saitama, Japan
| | - Takashi Ishiguro
- Department of Respiratory Medicine, Saitama Cardiovascular and Respiratory Center, Saitama, Japan
| | - Ken Arimura
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Fukuki Saito
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan
| | | | - Yasushi Nakano
- Department of Internal Medicine, Kawasaki Municipal Ida Hospital, Kawasaki, Kanagawa, Japan
| | - Yoshikazu Muto
- Department of Infectious Diseases, Tosei General Hospital, Aichi, Japan
| | - Yusuke Suzuki
- Department of Clinical Medicine (Laboratory of Bioregulatory Medicine), Kitasato University School of Pharmacy, Tokyo, Japan
- Department of Respiratory Medicine, Kitasato University, Kitasato Institute Hospital, Tokyo, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Murakami
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yasunori Sato
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Ryuji Koike
- Health Science Research and Development Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Naoki Hasegawa
- Department of Infectious Diseases, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, the University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-0071, Japan.
| |
Collapse
|
6
|
Crivelli L, Winkler A, Keller G, Beretta S, Calandri IL, De Groote W, Fornari A, Frontera J, Kivipelto M, Lopez-Rocha AS, Mangialasche F, Munblit D, Palmer K, Guekht A, Allegri R. Impact of COVID-19 on functional, cognitive, neuropsychiatric, and health-related outcomes in patients with dementia: A systematic review. eNeurologicalSci 2025; 38:100539. [PMID: 39720103 PMCID: PMC11663964 DOI: 10.1016/j.ensci.2024.100539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/07/2024] [Accepted: 11/15/2024] [Indexed: 12/26/2024] Open
Abstract
Background This systematic review analyzes the impact of COVID-19 on dementia patients' functional, cognitive, neuropsychiatric, and health related outcomes. It hypothesizes that dementia patients infected with SARS-CoV-2experience more pronounced deterioration compared to those who are uninfected. Methods Research from 01/03/2020 to 07/10/2023 was conducted using Medline, Web of Science, and Embase databases, and adhering to PRISMA guidelines and the PICO framework. The study aimed to determine if SARS-CoV-2 infection is associated with worse outcomes in dementia patients. The protocol is registered in PROSPERO (CRD42022352481), and bias was evaluated using the Newcastle-Ottawa Scale. Results Among 198 studies reviewed, only three met the criteria. Chen et al. (2023) identified higher mortality in SARS-CoV-2-infected dementia patients, while Merla et al. (2023) observed faster cognitive decline in infected individuals with increased hospital admissions. Additionally, Cascini et al. (2022) reported an increased risk of infection and significantly elevated mortality in dementia patients, highlighting comorbidities and antipsychotic medication use as key risk factors. Conclusion These limited data suggest higher mortality and cognitive decline in dementia patients following COVID-19, underscoring the need for extensive research in this area.
Collapse
Affiliation(s)
- Lucia Crivelli
- Department of Cognitive Neurology, Fleni, Montañeses 2325 (C1428AQK), Buenos Aires, Argentina
| | - Andrea Winkler
- Department of Neurology, Center for Global Health, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Community Medicine and Global Health, Institute of Health and Society, Faculty of Medicine, University of Oslo, Postboks 1130 Blindern, 0318 Oslo, Norway
- Department of Global Health and Social Medicine, Harvard Medical School, 25 Shattuck Street, 02115 Boston, MA, USA
| | - Greta Keller
- Department of Cognitive Neurology, Fleni, Montañeses 2325 (C1428AQK), Buenos Aires, Argentina
| | - Simone Beretta
- Department of Neurology, Fondazione IRCCS San Gerardo dei Tintori Monza, University of Milano Bicocca, Via G. B. Pergolesi, 33, 20900 Monza, MB, Italy
| | - Ismael Luis Calandri
- Department of Cognitive Neurology, Fleni, Montañeses 2325 (C1428AQK), Buenos Aires, Argentina
| | - Wouter De Groote
- WHO Rehabilitation Programme, Avenue Appia 20, 1211 Geneva, Switzerland
| | - Arianna Fornari
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Giovanni Celoria, 11, 20133 Milano, MI, Italy
| | | | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
- FINGERS Brain Health Institute, 22, 112 19 Stockholm, Sweden
- Medical Unit Aging, Karolinska University Hospital, 171 77 Stockholm, Sweden
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, St Dunstan's Road, London, United Kingdom
- Institute of Public Health and Clinical Nutrition and Institute of Clinical Medicine, Neurology, University of Eastern Finland, Yliopistonrinne 3, Kuopio, Finland
| | - Ana Sabsil Lopez-Rocha
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, 171 77 Stockholm, Karolinska Institutet, Sweden
| | - Francesca Mangialasche
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
- Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Daniel Munblit
- Care for Long Term Conditions Division, Florence Nightingale Faculty of Nursing, Midwifery and Palliative Care, King's College London, James Clerk Maxwell Building 57 Waterloo Road, London, United Kingdom
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Trubetskaya street, Russia
- Moscow Research and Clinical Center for Neuropsychiatry, Pirogov Russian National Research Medical University, Ulitsa Ostrovityanova, 1, 117997 Moscow, Russia
| | - Katie Palmer
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
- FINGERS Brain Health Institute, 22, 112 19 Stockholm, Sweden
| | - Alla Guekht
- Moscow Research and Clinical Center for Neuropsychiatry, Pirogov Russian National Research Medical University, Ulitsa Ostrovityanova, 1, 117997 Moscow, Russia
| | - Ricardo Allegri
- Department of Cognitive Neurology, Fleni, Montañeses 2325 (C1428AQK), Buenos Aires, Argentina
| |
Collapse
|
7
|
Miller JL, Leedale C, Kang D, Lilue J, Harder OE, Niewiesk S. Prostaglandin D2 delays CD8+ T-cell responses and respiratory syncytial virus clearance in geriatric cotton rats. J Virol 2025; 99:e0186324. [PMID: 39818970 PMCID: PMC11852932 DOI: 10.1128/jvi.01863-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
Respiratory syncytial virus (RSV) infection is associated with increased rates of severe disease, hospitalization, and death in elderly individuals. Clearance of RSV is frequently delayed within this demographic, contributing to the more severe disease course. Geriatric cotton rats mimic this prolonged clearance kinetic and serve as a useful animal model for studying age-associated immunological deficits during RSV infection. Treatment with the cyclooxygenase (COX) inhibitor ibuprofen restores RSV clearance, indicating that inflammation contributes to impaired clearance in geriatric cotton rats. Here, we further characterize a compromised immune response in geriatric cotton rats and identify an inflammatory pathway that contributes to this deficiency. Dendritic cell (DC) activation and migration to mediastinal lymph nodes are decreased during early infection in geriatric cotton rats, resulting in delayed generation of cytotoxic T cells and virus clearance. Prostaglandin D2 (PGD2), which reduces DC migration through the elevation of D-type prostanoid 1 receptor (DP1 receptor), is elevated in the airways of infected geriatric cotton rats. Reducing PGD2 production by inhibiting COX-2 or PGD2 synthase improves RSV clearance kinetics through DC activation and RSV-specific CD8+ T-cell responses in geriatric cotton rats, whereas activation of DP1 receptor through an agonist resulted in delayed viral clearance in adult cotton rats. These results indicate that PGD2 contributes to delayed antigen presentation and CD8+ T-cell responses to RSV in geriatric cotton rats. Inhibiting PGD2 generation or signaling may be a useful mechanism of therapeutic intervention in elderly individuals.IMPORTANCEElderly adults are at increased risk of severe disease resulting from infection with respiratory syncytial virus (RSV), characterized in part by delayed clearance (removal of the virus from airways). Understanding the immunological factors that lead to this delayed clearance may allow for the development of therapies to improve disease outcomes in elderly individuals infected with RSV and other respiratory viruses. Here, we describe an inflammatory pathway in geriatric cotton rats, the preferred small animal laboratory model for RSV, that impairs the generation of an effective immune response. We show that inhibiting this inflammatory pathway in geriatric cotton rats improves immune parameters and speeds clearance of RSV. These results contribute to our understanding of delayed RSV clearance in elderly individuals with possible applications for improving immune responses to RSV in clinical settings.
Collapse
Affiliation(s)
- Jonathan L. Miller
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Cameron Leedale
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Danyue Kang
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Olivia E. Harder
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
8
|
Liu X, Zhu L, Huang Z, Li Z, Duan R, Li H, Xie L, Chen X, Ding W, Chen B, Gao Y, Su J, Wang X, Su W. A dynamic peripheral immune landscape during human pregnancy. FUNDAMENTAL RESEARCH 2025; 5:391-406. [PMID: 40166108 PMCID: PMC11955049 DOI: 10.1016/j.fmre.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/28/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
Extensive immune adaptations occur during pregnancy to ensure successful delivery. However, these changes can increase the risk of disease in the mother. Here, we conducted single-cell RNA sequencing on peripheral blood mononuclear cells from pregnant women at different stages of pregnancy to elucidate the dynamic transcriptional changes in the maternal immune system. Gradual reduced cytotoxicity phenotype in highly variable cytotoxic T and natural killer cell types were observed during pregnancy. Reduced T- and B-cell response-related MHC-II and CD40 signaling as well as enhanced protolerance inducible costimulator and activin signaling may underlie the pregnancy-related weakening of adaptive immunity. Conversely, pro-inflammatory genes and pathways were upregulated in monocytes, possibly to compensate for the reduced T-cell response. Moreover, the transition from adaptive immune reduction to activation in late pregnancy in dendritic cells and CD4+ T cells was also detected. Notably, we proposed a novel view of the pro-aging effect of pregnancy from the perspective of immunity, and this effect may be restored postpartum. This work expands our knowledge of pregnancy immunity and may provide insights into the altered disease risks during pregnancy.
Collapse
Affiliation(s)
- Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Lei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Lihui Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Xiaozhen Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha 410078, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wen Ding
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Binyao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| | - Juan Su
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xianggui Wang
- Eye Center of Xiangya Hospital, Central South University, Changsha 410078, China
- Hunan Key Laboratory of Ophthalmology, Changsha 410078, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China
| |
Collapse
|
9
|
Müller L, Di Benedetto S. The impact of COVID-19 on accelerating of immunosenescence and brain aging. Front Cell Neurosci 2024; 18:1471192. [PMID: 39720706 PMCID: PMC11666534 DOI: 10.3389/fncel.2024.1471192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/29/2024] [Indexed: 12/26/2024] Open
Abstract
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has profoundly impacted global health, affecting not only the immediate morbidity and mortality rates but also long-term health outcomes across various populations. Although the acute effects of COVID-19 on the respiratory system have initially been the primary focus, it is increasingly evident that the virus can have significant impacts on multiple physiological systems, including the nervous and immune systems. The pandemic has highlighted the complex interplay between viral infection, immune aging, and brain health, that can potentially accelerate neuroimmune aging and contribute to the persistence of long COVID conditions. By inducing chronic inflammation, immunosenescence, and neuroinflammation, COVID-19 may exacerbate the processes of neuroimmune aging, leading to increased risks of cognitive decline, neurodegenerative diseases, and impaired immune function. Key factors include chronic immune dysregulation, oxidative stress, neuroinflammation, and the disruption of cellular processes. These overlapping mechanisms between aging and COVID-19 illustrate how the virus can induce and accelerate aging-related processes, leading to an increased risk of neurodegenerative diseases and other age-related conditions. This mini-review examines key features and possible mechanisms of COVID-19-induced neuroimmune aging that may contribute to the persistence and severity of long COVID. Understanding these interactions is crucial for developing effective interventions. Anti-inflammatory therapies, neuroprotective agents, immunomodulatory treatments, and lifestyle interventions all hold potential for mitigating the long-term effects of the virus. By addressing these challenges, we can improve health outcomes and quality of life for millions affected by the pandemic.
Collapse
Affiliation(s)
- Ludmila Müller
- Max Planck Institute for Human Development Center for Lifespan Psychology, Berlin, Germany
| | | |
Collapse
|
10
|
Calle-Peña ST, Diaz Tavara ED, Aguirre-Milachay E, León-Figueroa DA, Valladares-Garrido MJ. Predictors of high-flow nasal cannula failure in COVID-19 patients in a northern Peruvian hospital. BMC Pulm Med 2024; 24:414. [PMID: 39198776 PMCID: PMC11351638 DOI: 10.1186/s12890-024-03241-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
OBJECTIVES To determine predictors of high-flow nasal cannula (HFNC) failure in COVID-19 patients in a hospital in northern Peru. METHODOLOGY A retrospective cohort study was conducted during the months of March and May 2021. Data collection was based on a follow-up of 156 hospitalized patients with a diagnosis of COVID-19 who were users of HFNC. Epidemiological factors and clinical outcomes of treatment were analyzed from medical records. Epidemiological, analytical, and HFNC use-related characteristics were described using measures of absolute and relative frequencies, measures of central tendency, and dispersion. A multivariate Poisson regression analysis with robust variance and a 95% confidence interval was performed. RESULTS We found that age, SpO2/FiO2, work of breathing (WOB scale) at admission, degree of involvement, type of infiltrate on CT scan, lymphocytes, c-reactive protein, and D-dimer were significantly associated with failure of HFNC (p < 0.05). In addition, the WOB scale, PaO2/FiO2, SaO2/FiO2, and ROX index were variables that presented statistical significance (p < 0.0001). In the multivariate analysis model, a risk of failure of HFNC was determined with age > = 60 years [RRa 1.39 (1.05-1.85)] and PaO2/FiO2 score less than 100 [Rra 1.65 (0.99-2.76)]. CONCLUSIONS Predictors to failure of HFNC are age older than 60 years and minimally significantly lower PaO2/FiO2 than 100.
Collapse
Affiliation(s)
| | | | | | | | - Mario J Valladares-Garrido
- Universidad Continental, Lima, 15046, Peru.
- Oficina de Inteligencia Sanitaria, Red Prestacional EsSalud Lambayeque, Chiclayo, 14008, Peru.
| |
Collapse
|
11
|
Ma F. Assessing Immediate and Lasting Impacts of COVID-19-Induced Isolation on Green Space Usage Patterns. GEOHEALTH 2024; 8:e2024GH001062. [PMID: 39175506 PMCID: PMC11340692 DOI: 10.1029/2024gh001062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/08/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
The COVID-19 pandemic has profoundly influenced urban lifestyles, particularly the utilization of green spaces. While existing studies have primarily focused on the immediate effects of COVID-19-induced isolation, less attention has been given to the enduring impacts on green space usage patterns. This study addresses this gap by conducting three comprehensive surveys in Dezhou, China-before, during, and after the first wave of social isolation (December 2019, March 2020, December 2020). These surveys assessed socioeconomic conditions, commuting habits, green space usage habits, and landscape preferences, specifically focusing on usage frequency, duration of stays, and activities undertaken. Using Mann-Whitney U tests and Spearman's rho correlations, we identified significant long-term changes, including an increase in the frequency of visits by previously infrequent users, a reduction in visit durations, and a rise in high-intensity activities. These trends persisted 9 months post-isolation, highlighting the pandemic's lasting impact on green space usage and its critical role in enhancing public health and pandemic preparedness through thoughtful urban environmental design. This study not only sheds light on behavioral adaptations during a public health crisis but also offers evidence-based strategies for urban planning to bolster societal resilience in the face of future pandemics.
Collapse
Affiliation(s)
- Fengdi Ma
- Graduate School of Environmental StudiesSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
12
|
Asaba CN, Ekabe CJ, Ayuk HS, Gwanyama BN, Bitazar R, Bukong TN. Interplay of TLR4 and SARS-CoV-2: Unveiling the Complex Mechanisms of Inflammation and Severity in COVID-19 Infections. J Inflamm Res 2024; 17:5077-5091. [PMID: 39081874 PMCID: PMC11288317 DOI: 10.2147/jir.s474707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
The late 2019 emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, caused profound and unprecedented disruption to the global socio-economic structure, negatively affecting millions of lives worldwide. A typical hallmark of severe COVID-19 is hyper inflammation due to aberrant cytokine release (cytokine storm) by innate immune cells. Recent studies have revealed that SARS-CoV-2, through its spike (S) protein, can activate the body's innate immune cells via Toll-Like Receptors (TLRs), particularly TLR4. In silico studies have demonstrated that the S protein binds with high affinity to TLR4, triggering downstream signaling processes that result in pro-inflammatory cytokine release. Compared to other TLRs, such as TLR2, TLR4 plays a more significant role in initiating and sustaining the inflammatory response associated with severe COVID-19. Furthermore, interactions between the virus and target cells can enhance the cellular expression of TLR4, making cells more susceptible to viral interactions and subsequent inflammation. This increased expression of TLR4 upon viral entry creates a feedback loop, where heightened TLR4 levels lead to amplified inflammatory responses, contributing to the severity of the disease. Additionally, TLR4's potent activation of inflammatory pathways sets it apart from other TLRs, underscoring its pivotal role in the pathogenesis of COVID-19. In this review, we thoroughly explore the multitude of regulatory signaling pathways that SARS-CoV-2 employs to incite inflammation. We specifically focus on the critical impact of TLR4 activation compared to other TLRs, highlighting how TLR4's interactions with the viral S protein can exacerbate the severity of COVID-19. By delving into the mechanisms of TLR4-mediated inflammation, we aim to shed light on potential therapeutic targets that could mitigate the inflammatory damage caused by severe COVID-19. Understanding the unique role of TLR4 in the context of SARS-CoV-2 infection could pave the way for novel treatment strategies that specifically inhibit this receptor's activity, thereby reducing the overall disease burden and improving patient outcomes.
Collapse
Affiliation(s)
- Clinton Njinju Asaba
- Armand-Frappier Sante Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Cyril Jabea Ekabe
- Department of Translational Biomedical Sciences, University of Rochester, Rochester, NY, USA
| | - Humblenoble Stembridge Ayuk
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, 04318, Germany
| | | | - Razieh Bitazar
- Armand-Frappier Sante Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Terence Ndonyi Bukong
- Armand-Frappier Sante Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
13
|
Zhivaki D, Kennedy SN, Park J, Boriello F, Devant P, Cao A, Bahleda KM, Murphy S, McCabe C, Evavold CL, Chapman KL, Zanoni I, Ashenberg O, Xavier RJ, Kagan JC. Correction of age-associated defects in dendritic cells enables CD4 + T cells to eradicate tumors. Cell 2024; 187:3888-3903.e18. [PMID: 38870946 PMCID: PMC11283364 DOI: 10.1016/j.cell.2024.05.026] [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: 11/02/2022] [Revised: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
Defective host defenses later in life are associated with changes in immune cell activities, suggesting that age-specific considerations are needed in immunotherapy approaches. In this study, we found that PD-1 and CTLA4-based cancer immunotherapies are unable to eradicate tumors in elderly mice. This defect in anti-tumor activity correlated with two known age-associated immune defects: diminished abundance of systemic naive CD8+ T cells and weak migratory activities of dendritic cells (DCs). We identified a vaccine adjuvant, referred to as a DC hyperactivator, which corrects DC migratory defects in the elderly. Vaccines containing tumor antigens and DC hyperactivators induced T helper type 1 (TH1) CD4+ T cells with cytolytic activity that drive anti-tumor immunity in elderly mice. When administered early in life, DC hyperactivators were the only adjuvant identified that elicited anti-tumor CD4+ T cells that persisted into old age. These results raise the possibility of correcting age-associated immune defects through DC manipulation.
Collapse
Affiliation(s)
- Dania Zhivaki
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Stephanie N Kennedy
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Josh Park
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francesco Boriello
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Pascal Devant
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Anh Cao
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kristin M Bahleda
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Shane Murphy
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cristin McCabe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles L Evavold
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Kate L Chapman
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ramnik J Xavier
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan C Kagan
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
14
|
Ledderose C, Valsami EA, Elevado M, Liu Q, Giva B, Curatolo J, Delfin J, Abutabikh R, Junger WG. Impaired ATP hydrolysis in blood plasma contributes to age-related neutrophil dysfunction. Immun Ageing 2024; 21:45. [PMID: 38961477 PMCID: PMC11221114 DOI: 10.1186/s12979-024-00441-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The function of polymorphonuclear neutrophils (PMNs) decreases with age, which results in infectious and inflammatory complications in older individuals. The underlying causes are not fully understood. ATP release and autocrine stimulation of purinergic receptors help PMNs combat microbial invaders. Excessive extracellular ATP interferes with these mechanisms and promotes inflammatory PMN responses. Here, we studied whether dysregulated purinergic signaling in PMNs contributes to their dysfunction in older individuals. RESULTS Bacterial infection of C57BL/6 mice resulted in exaggerated PMN activation that was significantly greater in old mice (64 weeks) than in young animals (10 weeks). In contrast to young animals, old mice were unable to prevent the systemic spread of bacteria, resulting in lethal sepsis and significantly greater mortality in old mice than in their younger counterparts. We found that the ATP levels in the plasma of mice increased with age and that, along with the extracellular accumulation of ATP, the PMNs of old mice became increasingly primed. Stimulation of the formyl peptide receptors of those primed PMNs triggered inflammatory responses that were significantly more pronounced in old mice than in young animals. However, bacterial phagocytosis and killing by PMNs of old mice were significantly lower than that of young mice. These age-dependent PMN dysfunctions correlated with a decrease in the enzymatic activity of plasma ATPases that convert extracellular ATP to adenosine. ATPases depend on divalent metal ions, including Ca2+, Mg2+, and Zn2+, and we found that depletion of these ions blocked the hydrolysis of ATP and the formation of adenosine in human blood, resulting in ATP accumulation and dysregulation of PMN functions equivalent to those observed in response to aging. CONCLUSIONS Our findings suggest that impaired hydrolysis of plasma ATP dysregulates PMN function in older individuals. We conclude that strategies aimed at restoring plasma ATPase activity may offer novel therapeutic opportunities to reduce immune dysfunction, inflammation, and infectious complications in older patients.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Mark Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qing Liu
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Brennan Giva
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Julian Curatolo
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Joshua Delfin
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Reem Abutabikh
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Wolfgang G Junger
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA.
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Zhang T, Wen R, Fan H, Yu Y, Jia H, Peng Z, Zhou L, Yu G, Zhang W. Impact and potential value of immunosenescence on solid gastrointestinal tumors. Front Immunol 2024; 15:1375730. [PMID: 39007138 PMCID: PMC11239362 DOI: 10.3389/fimmu.2024.1375730] [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: 01/24/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024] Open
Abstract
Solid gastrointestinal tumors often respond poorly to immunotherapy for the complex tumor microenvironment (TME), which is exacerbated by immune system alterations. Immunosenescence is the process of increased diversification of immune genes due to aging and other factors, leading to a decrease in the recognition function of the immune system. This process involves immune organs, immune cells, and the senescence-associated secretory phenotype (SASP). The most fundamental change is DNA damage, resulting in TME remodeling. The main manifestations are worsening inflammation, increased immunosuppressive SASP production, decreased immune cell antitumor activity, and the accumulation of tumor-associated fibroblasts and myeloid-derived suppressor cells, making antitumor therapy less effective. Senotherapy strategies to remove senescent cells and block key senescence processes can have synergistic effects with other treatments. This review focuses on immunoenescence and its impact on the solid TME. We characterize the immunosenescent TME and discuss future directions for antitumor therapies targeting senescence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leqi Zhou
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanyu Yu
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Basudkar V, Gujrati G, Ajgaonkar S, Gandhi M, Mehta D, Nair S. Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging. Pharmaceuticals (Basel) 2024; 17:597. [PMID: 38794167 PMCID: PMC11123800 DOI: 10.3390/ph17050597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Inflammaging, a coexistence of inflammation and aging, is a persistent, systemic, low-grade inflammation seen in the geriatric population. Various natural compounds have been greatly explored for their potential role in preventing and treating inflammaging. Withania somnifera has been used for thousands of years in traditional medicine as a nutraceutical for its numerous health benefits including regenerative and adaptogenic effects. Recent preclinical and clinical studies on the role of Withania somnifera and its active compounds in treating aging, inflammation, and oxidative stress have shown promise for its use in healthy aging. We discuss the chemistry of Withania somnifera, the etiology of inflammaging and the protective role(s) of Withania somnifera in inflammaging in key organ systems including brain, lung, kidney, and liver as well as the mechanistic underpinning of these effects. Furthermore, we elucidate the beneficial effects of Withania somnifera in oxidative stress/DNA damage, immunomodulation, COVID-19, and the microbiome. We also delineate a putative protein-protein interaction network of key biomarkers modulated by Withania somnifera in inflammaging. In addition, we review the safety/potential toxicity of Withania somnifera as well as global clinical trials on Withania somnifera. Taken together, this is a synthetic review on the beneficial effects of Withania somnifera in inflammaging and highlights the potential of Withania somnifera in improving the health-related quality of life (HRQoL) in the aging population worldwide.
Collapse
Affiliation(s)
- Vivek Basudkar
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Gunjan Gujrati
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Saiprasad Ajgaonkar
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Manav Gandhi
- College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dilip Mehta
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Sujit Nair
- PhytoVeda Pvt. Ltd., Mumbai 400 022, India
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| |
Collapse
|
17
|
Nehar-Belaid D, Sokolowski M, Ravichandran S, Banchereau J, Chaussabel D, Ucar D. Baseline immune states (BIS) associated with vaccine responsiveness and factors that shape the BIS. Semin Immunol 2023; 70:101842. [PMID: 37717525 DOI: 10.1016/j.smim.2023.101842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Vaccines are among the greatest inventions in medicine, leading to the elimination or control of numerous diseases, including smallpox, polio, measles, rubella, and, most recently, COVID-19. Yet, the effectiveness of vaccines varies among individuals. In fact, while some recipients mount a robust response to vaccination that protects them from the disease, others fail to respond. Multiple clinical and epidemiological factors contribute to this heterogeneity in responsiveness. Systems immunology studies fueled by advances in single-cell biology have been instrumental in uncovering pre-vaccination immune cell types and genomic features (i.e., the baseline immune state, BIS) that have been associated with vaccine responsiveness. Here, we review clinical factors that shape the BIS, and the characteristics of the BIS associated with responsiveness to frequently studied vaccines (i.e., influenza, COVID-19, bacterial pneumonia, malaria). Finally, we discuss potential strategies to enhance vaccine responsiveness in high-risk groups, focusing specifically on older adults.
Collapse
Affiliation(s)
| | - Mark Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | | | - Damien Chaussabel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
18
|
Ouedraogo AR, Traoré L, Ouattara AK, Ouedraogo AR, Zongo SV, Savadogo M, Lallogo TD, Sombie HK, Sorgho PA, Ouedraogo TWC, Djigma FW, Lamien AS, Yonli AT, Lompo OM, Simporé J. Association of HLA-DRB1*11 and HLA-DRB1*12 gene polymorphism with COVID-19 in Burkina Faso. BMC Med Genomics 2023; 16:246. [PMID: 37845715 PMCID: PMC10577973 DOI: 10.1186/s12920-023-01684-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND The clinical manifestations of coronavirus disease (COVID-19) can vary widely, ranging from asymptomatic to severe, and may be influenced by the host genetic background. The aim of the present study was to determine the frequencies of HLA-DRB1*11 and HLA-DRB1*12 allele polymorphisms and their associations with COVID-19. METHODS In this cross-sectional study, 198 subjects were enrolled, including 150 COVID-19 positive cases and 48 subjects who tested negative for COVID-19. Participants were recruited from the emergency, intensive care, and infectious diseases departments of the Bogodogo Centre University Hospital (CHU-B) or the routine laboratory of Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA). Genomic DNA was extracted from nasopharyngeal swabs samples and multiplex PCR-SSP was used to detect the HLA-DRB1*11 and HLA-DRB1*12 alleles. The study was approved by CERS (№ 2021-02-033). RESULTS The positive cases were categorized into 38 asymptomatic (CC+), 60 symptomatic (NC+), and 52 severe cases (SC+). Females were more frequent in the overall study population (53.0%, 105/198) as well as in the negative group's CC- (68.75%, 33/48) and SC+ (57.69%, 30/52 negative groups, whereas males were more frequent in the CC+ (63.16%, 24/38) and NC+ (53.33%, 32/60) groups. The highest mean age was observed in the SC + group. A frequency of 19.19% (38/198) and 14.65% (29/198) was found for the HLA-DRB1*11 and HLA-DRB1*12 alleles, respectively. Individuals carrying the HLA-DRB1*11 allele had an approximately sixfold higher risk of asymptomatic SARS-CoV-2 infection (OR = 5.72 [1.683-19.442], p = 0.005) based on the association analysis. CONCLUSIONS Altogether, the present study reports high frequency of HLA-DRB1*11 and HLA-DRB1*12 alleles within a population from Ouagadougou, Burkina Faso. The results suggest that individuals carrying the HLA-DRB1*11 allele are more susceptible to COVID-19 infection but may not display symptoms.
Collapse
Affiliation(s)
- Alfred Rakissida Ouedraogo
- Laboratoire de Morphologie et Organogénèse (LAMO), Université Joseph KI-ZERBO, UFR/SDS, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Lassina Traoré
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
- Université Norbert ZONGO - Centre Universitaire de Manga, BP 376, Koudougou, Burkina Faso
| | - Abdoul Karim Ouattara
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
- Université Norbert ZONGO - Centre Universitaire de Manga, BP 376, Koudougou, Burkina Faso
| | - Alexis Rakiswende Ouedraogo
- Laboratoire de Morphologie et Organogénèse (LAMO), Université Joseph KI-ZERBO, UFR/SDS, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Sidnooma Véronique Zongo
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Mousso Savadogo
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Tatiana Doriane Lallogo
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Herman Karim Sombie
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA), P.O. Box 364, Ouagadougou 01, Burkina Faso
| | - Pegdwendé Abel Sorgho
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA), P.O. Box 364, Ouagadougou 01, Burkina Faso
| | - Teega-Wendé Clarisse Ouedraogo
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Florencia Wendkuuni Djigma
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso.
| | - Assita Sanou Lamien
- Laboratoire de Morphologie et Organogénèse (LAMO), Université Joseph KI-ZERBO, UFR/SDS, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Albert Théophane Yonli
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA), P.O. Box 364, Ouagadougou 01, Burkina Faso
| | - Olga Mélanie Lompo
- Laboratoire de Morphologie et Organogénèse (LAMO), Université Joseph KI-ZERBO, UFR/SDS, Ouagadougou 01, 01 BP 7021, Burkina Faso
| | - Jacques Simporé
- Université Joseph KI-ZERBO, Laboratoire de Biologie Moléculaire et de Génétique (LABIOGENE), UFR/SVT, Ouagadougou 01, 01 BP 7021, Burkina Faso
- Centre de Recherche Biomoléculaire Pietro Annigoni (CERBA), P.O. Box 364, Ouagadougou 01, Burkina Faso
| |
Collapse
|
19
|
Chen H, Luo Y, Zhang X, Luo R, Bian Y, Hou Y, Liu Y. Global research trends of immunosenescence: A bibliometric study. Heliyon 2023; 9:e20362. [PMID: 37818000 PMCID: PMC10560770 DOI: 10.1016/j.heliyon.2023.e20362] [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: 11/09/2022] [Revised: 06/26/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Background Immunosenescence, an age-related deficit in immunity, associated with multiple disorders and making the successful aging a challenge. Although nearly 4000 articles have been published, only few review articles have summarized the research status. In order to better understand the most recent advances, hotspots and development trends in immunosenescence, it is very necessary to conduct a comprehensive bibliometric analysis. Hence, commonly used bibliometric analysis software CiteSpace and VOSviewer were employed to conduct a quantitative analysis and critical evaluation of publications in this study. Methods Immunosenescence publications were screened from the Web of Science Core Collection (WoSCC). Microsoft Excel 2021, CiteSpace 5.8.R3, and VOSviewer 1.6.17 were used for bibliometric study. Results A total of 3875 publications were retrieved from WoSCC. After screening by document type (article or review) (352 publications were excluded) and language of English (85 were excluded), 3438 studies were finally used for bibliometric analysis. The literature on immunosenescence had been continuously growing since 1991, and by 2020 it has skyrocketed 312 publications from 240 in 2019. USA (1111 publications, 35.01%) was the leading country of publications, followed by ITALY (379, 11.94%) and ENGLAND (366, 11.53%). Of the authors, Pawelec G from the Tubingen University of GERMANY contributed the greatest articles (93 publications). All the keywords could be divided into five clusters, and additional potent visualization bursts revealed that "gut microbiota," "health," "dysfunction," and "nivolumab" were the active hotspots presently. Conclusion Based on the current data, we firstly concluded that there will be a dramatically rising publications on immunosenescence, and research teams from USA or GERMANY might be the best chooses for collaboration. Moreover, We particularly emphasized the development potential of mechanism and intervening strateges like "gut microbiota" and "nivolumab" in immunosenescence. We hope to provide new ideas for promoting the basic research and clinical application of immunosenescence.
Collapse
Affiliation(s)
- Hongbo Chen
- School of Nursing, Guizhou University of Traditional Chinese Medicine, Guizhou, China
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiwei Luo
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohong Zhang
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruzhen Luo
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunlong Hou
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China
| | - Yanhui Liu
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
20
|
Lin HF, Liu MQ, Jiang RD, Gong QC, Su J, Guo ZS, Chen Y, Jia JK, Dong TY, Zhu Y, Li A, Shen XR, Wang Y, Li B, Xie TT, Yang XL, Hu B, Shi ZL. Characterization of a mouse-adapted strain of bat severe acute respiratory syndrome-related coronavirus. J Virol 2023; 97:e0079023. [PMID: 37607058 PMCID: PMC10537601 DOI: 10.1128/jvi.00790-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 06/18/2023] [Indexed: 08/24/2023] Open
Abstract
Bats carry genetically diverse severe acute respiratory syndrome-related coronaviruses (SARSr-CoVs). Some of them utilize human angiotensin-converting enzyme 2 (hACE2) as a receptor and cannot efficiently replicate in wild-type mice. Our previous study demonstrated that the bat SARSr-CoV rRsSHC014S induces respiratory infection and lung damage in hACE2 transgenic mice but not wild-type mice. In this study, we generated a mouse-adapted strain of rRsSHC014S, which we named SMA1901, by serial passaging of wild-type virus in BALB/c mice. SMA1901 showed increased infectivity in mouse lungs and induced interstitial lung pneumonia in both young and aged mice after intranasal inoculation. Genome sequencing revealed mutations in not only the spike protein but the whole genome, which may be responsible for the enhanced pathogenicity of SMA1901 in wild-type BALB/c mice. SMA1901 induced age-related mortality similar to that observed in SARS and COVID-19. Drug testing using antibodies and antiviral molecules indicated that this mouse-adapted virus strain can be used to test prophylactic and therapeutic drug candidates against SARSr-CoVs. IMPORTANCE The genetic diversity of SARSr-CoVs in wildlife and their potential risk of cross-species infection highlights the importance of developing a powerful animal model to evaluate the antibodies and antiviral drugs. We acquired the mouse-adapted strain of a bat-origin coronavirus named SMA1901 by natural serial passaging of rRsSHC014S in BALB/c mice. The SMA1901 infection caused interstitial pneumonia and inflammatory immune responses in both young and aged BALB/c mice after intranasal inoculation. Our model exhibited age-related mortality similar to SARS and COVID-19. Therefore, our model will be of high value for investigating the pathogenesis of bat SARSr-CoVs and could serve as a prospective test platform for prophylactic and therapeutic candidates.
Collapse
Affiliation(s)
- Hao-Feng Lin
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mei-Qin Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ren-Di Jiang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Jia Su
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Shuo Guo
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing-Kun Jia
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tian-Yi Dong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ang Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xu-Rui Shen
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Yi Wang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Bei Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ting-Ting Xie
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xing-Lou Yang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ben Hu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zheng-Li Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
21
|
Shao C, Shi Y, Chen R, Liu X, Huang H, Zhao Y, Xu K, Chen K, Wang M, Xu Z. Risk factors associated with COVID-19 pneumonia in Chinese patients with pre-existing interstitial lung disease during the SARS-CoV-2 pandemic. J Med Virol 2023; 95:e29098. [PMID: 37707416 DOI: 10.1002/jmv.29098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/04/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
In China, the emergence of a nationally widespread epidemic infection of severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) has appeared within a month since December 7, 2022. To evaluate the risk factors for suffering from coronavirus disease 2019 (COVID-19) pneumonia due to infection with SARS-CoV-2 in different kinds of interstitial lung disease (ILD) patients with diverse immunizations, we conducted this retrospective study on 525 patients with ILDs who underwent regular follow-up in our ILD clinic. Among them, 128 ILD patients (24.4%) suffered from COVID-19 pneumonia after SARS-CoV-2 infection. Patients were older with a male predominance in the pneumonia group than in the nonpneumonia group (65.0 ± 10.0 years vs. 56.4 ± 11.7 years, p < 0.001, 55.5% vs. 39.5%, p = 0.002, respectively). Connective tissue disease-associated ILD (CTD-ILD) (25%), idiopathic pulmonary fibrosis (23.4%), and interstitial pneumonia with autoimmune features (21.1%) were the main pre-existing ILDs in the pneumonia group. In Cox multivariable analysis, only male sex and corticosteroid use were risk factors for COVID-19 pneumonia after infection. Two or three doses of vaccination were a protective factor for pre-existing ILD patients suffering from COVID-19 pneumonia. More than two doses of vaccination were strongly recommended for pre-existing ILD patients, particularly for males who were administered corticosteroids.
Collapse
Affiliation(s)
- Chi Shao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yujie Shi
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruxuan Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiangning Liu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hui Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yang Zhao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kai Xu
- Radiological Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Keqi Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zuojun Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Fonseca DLM, Filgueiras IS, Marques AHC, Vojdani E, Halpert G, Ostrinski Y, Baiocchi GC, Plaça DR, Freire PP, Pour SZ, Moll G, Catar R, Lavi YB, Silverberg JI, Zimmerman J, Cabral-Miranda G, Carvalho RF, Khan TA, Heidecke H, Dalmolin RJS, Luchessi AD, Ochs HD, Schimke LF, Amital H, Riemekasten G, Zyskind I, Rosenberg AZ, Vojdani A, Shoenfeld Y, Cabral-Marques O. Severe COVID-19 patients exhibit elevated levels of autoantibodies targeting cardiolipin and platelet glycoprotein with age: a systems biology approach. NPJ AGING 2023; 9:21. [PMID: 37620330 PMCID: PMC10449916 DOI: 10.1038/s41514-023-00118-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/12/2023] [Indexed: 08/26/2023]
Abstract
Age is a significant risk factor for the coronavirus disease 2019 (COVID-19) severity due to immunosenescence and certain age-dependent medical conditions (e.g., obesity, cardiovascular disorder, and chronic respiratory disease). However, despite the well-known influence of age on autoantibody biology in health and disease, its impact on the risk of developing severe COVID-19 remains poorly explored. Here, we performed a cross-sectional study of autoantibodies directed against 58 targets associated with autoimmune diseases in 159 individuals with different COVID-19 severity (71 mild, 61 moderate, and 27 with severe symptoms) and 73 healthy controls. We found that the natural production of autoantibodies increases with age and is exacerbated by SARS-CoV-2 infection, mostly in severe COVID-19 patients. Multiple linear regression analysis showed that severe COVID-19 patients have a significant age-associated increase of autoantibody levels against 16 targets (e.g., amyloid β peptide, β catenin, cardiolipin, claudin, enteric nerve, fibulin, insulin receptor a, and platelet glycoprotein). Principal component analysis with spectrum decomposition and hierarchical clustering analysis based on these autoantibodies indicated an age-dependent stratification of severe COVID-19 patients. Random forest analysis ranked autoantibodies targeting cardiolipin, claudin, and platelet glycoprotein as the three most crucial autoantibodies for the stratification of severe COVID-19 patients ≥50 years of age. Follow-up analysis using binomial logistic regression found that anti-cardiolipin and anti-platelet glycoprotein autoantibodies significantly increased the likelihood of developing a severe COVID-19 phenotype with aging. These findings provide key insights to explain why aging increases the chance of developing more severe COVID-19 phenotypes.
Collapse
Affiliation(s)
- Dennyson Leandro M Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil.
| | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Alexandre H C Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Elroy Vojdani
- Regenera Medical 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA, 90025, USA
| | - Gilad Halpert
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Saint Petersburg State University Russia, Saint Petersburg, Russia
| | - Yuri Ostrinski
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Saint Petersburg State University Russia, Saint Petersburg, Russia
| | - Gabriela Crispim Baiocchi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Desirée Rodrigues Plaça
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paula P Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Shahab Zaki Pour
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Guido Moll
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Rusan Catar
- Departament of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Yael Bublil Lavi
- Scakler faculty of medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan I Silverberg
- Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Gustavo Cabral-Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Robson F Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Harald Heidecke
- CellTrend Gesellschaft mit beschränkter Haftung (GmbH), Luckenwalde, Germany
| | - Rodrigo J S Dalmolin
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Andre Ducati Luchessi
- Department of Clinical and Toxicological Analyses, Federal University of Rio Grande do Norte, R.N., Natal, Brazil
| | - Hans D Ochs
- Department of Pediatrics, University of Washington School of Medicine, and Seattle Children's Research Institute, Seattle, WA, USA
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Howard Amital
- Ariel University, Ari'el, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Medicine B, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriela Riemekasten
- Department of Rheumatology, University Medical Center Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Israel Zyskind
- Maimonides Medical Center, Brooklyn, NY, USA
- Department of Pediatrics, NYU Langone Medical Center, New York, NY, USA
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Aristo Vojdani
- Department of Immunology, Immunosciences Laboratory, Inc., Los Angeles, CA, USA
- Cyrex Laboratories, LLC 2602 S. 24th St., Phoenix, AZ, 85034, USA
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - Otavio Cabral-Marques
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, SP, Brazil.
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
- Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil.
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil.
- Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil.
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, SP, Brazil.
| |
Collapse
|
23
|
Liu Z, Huang Y, Wang D, Li M, Zhang Q, Pan C, Lin Y, Luo Y, Shi Z, Zhang P, Zheng Y. Insights gained from single-cell RNA analysis of murine endothelial cells in aging hearts. Heliyon 2023; 9:e18324. [PMID: 37554834 PMCID: PMC10404962 DOI: 10.1016/j.heliyon.2023.e18324] [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: 01/24/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
Aging is the strongest risk factor for cardiovascular disease, with progressive decline in the function of vascular endothelial cells (ECs) with age. Systematic analyses of the effects of aging on different cardiac EC types remain limited. Here, we constructed a scRNA atlas of EC transcriptomes in young and old mouse hearts. We identified 10 EC subclusters. The multidimensionally differential genes (DEGs) analysis across different EC clusters shows molecular changes with aging, showing the increase in the overall inflammatory microenvironment and the decrease in angiogenesis and cytoskeletal support capacity of aged ECs. And we performed an in-depth analysis of 3 special ECs, Immunology, Proliferating and Angiogenic. The Immunology EC seems highly associated with some immune regulatory functions, which decline with aging at different degrees. Analysis of two types of neovascular ECs, Proliferating, Angiogenic, implied that Angiogenic ECs can differentiate into multiple EC directions after initially originating from proliferating ECs. And aging leads to a decrease in the ability of vascular angiogenesis and differentiation. Finally, we summarized the effects of aging on cell signaling communication between different EC clusters. This cardiac EC atlas offers comprehensive insights into the molecular regulations of cardiovascular aging, and provides new directions for the prevention and treatment of age-related cardiovascular disease.
Collapse
Affiliation(s)
- Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100085, China
| | - Yanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dongliang Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Mengke Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Qikai Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Caineng Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yuheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yuanting Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Zhuoxing Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yingfeng Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100085, China
| |
Collapse
|
24
|
Laphanuwat P, Gomes DCO, Akbar AN. Senescent T cells: Beneficial and detrimental roles. Immunol Rev 2023; 316:160-175. [PMID: 37098109 PMCID: PMC10952287 DOI: 10.1111/imr.13206] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 04/01/2023] [Indexed: 04/27/2023]
Abstract
As the thymus involutes during aging, the T-cell pool has to be maintained by the periodic expansion of preexisting T cells during adulthood. A conundrum is that repeated episodes of activation and proliferation drive the differentiation of T cells toward replicative senescence, due to telomere erosion. This review discusses mechanisms that regulate the end-stage differentiation (senescence) of T cells. Although these cells, within both CD4 and CD8 compartments, lose proliferative activity after antigen-specific challenge, they acquire innate-like immune function. While this may confer broad immune protection during aging, these senescent T cells may also cause immunopathology, especially in the context of excessive inflammation in tissue microenvironments.
Collapse
Affiliation(s)
- Phatthamon Laphanuwat
- Division of MedicineUniversity College LondonLondonUK
- Department of PharmacologyFaculty of Medicine, Khon Kaen UniversityKhon KaenThailand
| | - Daniel Claudio Oliveira Gomes
- Division of MedicineUniversity College LondonLondonUK
- Núcleo de Doenças InfecciosasUniversidade Federal do Espírito SantoVitoriaBrazil
- Núcleo de BiotecnologiaUniversidade Federal do Espírito SantoVitoriaBrazil
| | - Arne N. Akbar
- Division of MedicineUniversity College LondonLondonUK
| |
Collapse
|
25
|
Eder J, Schumm L, Armann JP, Puhan MA, Beuschlein F, Kirschbaum C, Berner R, Toepfner N. Increased red blood cell deformation in children and adolescents after SARS-CoV-2 infection. Sci Rep 2023; 13:9823. [PMID: 37330522 PMCID: PMC10276822 DOI: 10.1038/s41598-023-35692-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/22/2023] [Indexed: 06/19/2023] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) is associated with hyperinflammation, hypercoagulability and hypoxia. Red blood cells (RBCs) play a key role in microcirculation and hypoxemia and are therefore of special interest in COVID-19 pathophysiology. While this novel disease has claimed the lives of many older patients, it often goes unnoticed or with mild symptoms in children. This study aimed to investigate morphological and mechanical characteristics of RBCs after SARS-CoV-2 infection in children and adolescents by real-time deformability-cytometry (RT-DC), to investigate the relationship between alterations of RBCs and clinical course of COVID-19. Full blood of 121 students from secondary schools in Saxony, Germany, was analyzed. SARS-CoV-2-serostatus was acquired at the same time. Median RBC deformation was significantly increased in SARS-CoV-2-seropositive compared to seronegative children and adolescents, but no difference could be detected when the infection dated back more than 6 months. Median RBC area was the same in seropositive and seronegative adolescents. Our findings of increased median RBC deformation in SARS-CoV-2 seropositive children and adolescents until 6 months post COVID-19 could potentially serve as a progression parameter in the clinical course of the disease with an increased RBC deformation pointing towards a mild course of COVID-19.
Collapse
Affiliation(s)
- Julian Eder
- Biopsychology, Technische Universität Dresden, Dresden, Germany
| | - Leonie Schumm
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jakob P Armann
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Milo A Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | | | - Reinhard Berner
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nicole Toepfner
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
26
|
Jiang QL, Wang FY, Zheng KJ. [Clinical features of children with coronavirus disease 2019 in different age groups during the epidemic of Omicron variant]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:600-605. [PMID: 37382129 DOI: 10.7499/j.issn.1008-8830.2302014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
OBJECTIVES To study the differences in the clinical features of children with coronavirus disease 2019 (COVID-19) in different age groups during the epidemic of Omicron variant. METHODS A retrospective analysis was performed on the clinical data of 211 children with COVID-19 who were admitted to the Department of General Pediatrics, Zhongshan People's Hospital, from December 9, 2022 to January 8, 2023. According to their age, they were divided into 4 groups: 1 month-<1 year (n=84), 1-<3 years group (n=64), 3-<5 years (n=29), and ≥5 years (n=34). The above groups were compared in terms of general status, clinical features, ancillary examination results, treatment, and outcome. RESULTS The children aged <3 years accounted for 70.1% (148/211) of all hospitalized children with COVID-19, and the 3-<5 years group and the ≥5 years group had a significantly higher proportion of children with underlying diseases than the 1 month-<1 year group and the 1-<3 years group (P<0.05). Compared with the other three groups, the 1 month-<1 year group had significantly higher incidence rates of dyspnea, nasal congestion/nasal discharge, diarrhea and significantly lower incidence rates of convulsion and nervous system involvement (P<0.05). Moreover, compared with the other three groups, the 1 month-<1 year group had significantly higher incidence rates of increases in bile acid and creatine kinase isoenzyme and significantly lower incidence rates of decreased platelet count, increased neutrophil percentage, and decreased lymphocyte percentage (P<0.05). The 1 month-<1 year group had a significantly higher incidence rate of mild COVID-19 than the 1-<3 years group and a significantly lower incidence rate of severe/critical COVID-19 than the other three groups (P<0.05). Compared with the other three groups, the 1 month-<1 year group had a significantly higher proportion of children receiving oxygen inhalation therapy (P<0.05). CONCLUSIONS Children with COVID-19 in different age groups have different clinical features during the epidemic of Omicron variant, especially between the children aged 1 month to <1 year and those aged ≥1 year.
Collapse
Affiliation(s)
- Qing-Lian Jiang
- Department of General Pediatrics, Zhongshan People's Hospital, Zhongshan, Guangdong 528400, China
| | - Feng-Yan Wang
- Department of General Pediatrics, Zhongshan People's Hospital, Zhongshan, Guangdong 528400, China
| | - Kai-Jun Zheng
- Department of General Pediatrics, Zhongshan People's Hospital, Zhongshan, Guangdong 528400, China
| |
Collapse
|
27
|
Jin J, Mu Y, Zhang H, Sturmlechner I, Wang C, Jadhav RR, Xia Q, Weyand CM, Goronzy JJ. CISH impairs lysosomal function in activated T cells resulting in mitochondrial DNA release and inflammaging. NATURE AGING 2023; 3:600-616. [PMID: 37118554 PMCID: PMC10388378 DOI: 10.1038/s43587-023-00399-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/15/2023] [Indexed: 04/30/2023]
Abstract
Chronic systemic inflammation is one of the hallmarks of the aging immune system. Here we show that activated T cells from older adults contribute to inflammaging by releasing mitochondrial DNA (mtDNA) into their environment due to an increased expression of the cytokine-inducible SH2-containing protein (CISH). CISH targets ATP6V1A, an essential component of the proton pump V-ATPase, for proteasomal degradation, thereby impairing lysosomal function. Impaired lysosomal activity caused intracellular accumulation of multivesicular bodies and amphisomes and the export of their cargos, including mtDNA. CISH silencing in T cells from older adults restored lysosomal activity and prevented amphisomal release. In antigen-specific responses in vivo, CISH-deficient CD4+ T cells released less mtDNA and induced fewer inflammatory cytokines. Attenuating CISH expression may present a promising strategy to reduce inflammation in an immune response of older individuals.
Collapse
Affiliation(s)
- Jun Jin
- Multiscale Research Institute for Complex Systems, Fudan University, Shanghai, China.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
| | - Yunmei Mu
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Huimin Zhang
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | - Chenyao Wang
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Rohit R Jadhav
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Qiong Xia
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Cornelia M Weyand
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Jorg J Goronzy
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Division of Rheumatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
28
|
Zhang C, Guo ZF, Liu W, Kazama K, Hu L, Sun X, Wang L, Lee H, Lu L, Yang XF, Summer R, Sun J. PIMT is a novel and potent suppressor of endothelial activation. eLife 2023; 12:e85754. [PMID: 37070640 PMCID: PMC10112892 DOI: 10.7554/elife.85754] [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: 12/22/2022] [Accepted: 04/02/2023] [Indexed: 04/19/2023] Open
Abstract
Proinflammatory agonists provoke the expression of cell surface adhesion molecules on endothelium in order to facilitate leukocyte infiltration into tissues. Rigorous control over this process is important to prevent unwanted inflammation and organ damage. Protein L-isoaspartyl O-methyltransferase (PIMT) converts isoaspartyl residues to conventional methylated forms in cells undergoing stress-induced protein damage. The purpose of this study was to determine the role of PIMT in vascular homeostasis. PIMT is abundantly expressed in mouse lung endothelium and PIMT deficiency in mice exacerbated pulmonary inflammation and vascular leakage to LPS(lipopolysaccharide). Furthermore, we found that PIMT inhibited LPS-induced toll-like receptor signaling through its interaction with TNF receptor-associated factor 6 (TRAF6) and its ability to methylate asparagine residues in the coiled-coil domain. This interaction was found to inhibit TRAF6 oligomerization and autoubiquitination, which prevented NF-κB transactivation and subsequent expression of endothelial adhesion molecules. Separately, PIMT also suppressed ICAM-1 expression by inhibiting its N-glycosylation, causing effects on protein stability that ultimately translated into reduced EC(endothelial cell)-leukocyte interactions. Our study has identified PIMT as a novel and potent suppressor of endothelial activation. Taken together, these findings suggest that therapeutic targeting of PIMT may be effective in limiting organ injury in inflammatory vascular diseases.
Collapse
Affiliation(s)
- Chen Zhang
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Zhi-Fu Guo
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Wennan Liu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Kyosuke Kazama
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Louis Hu
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Xiaobo Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Lu Wang
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Hyoungjoo Lee
- Quantitative Proteomics Resource Center, University of PennsylvaniaPhiladelphiaUnited States
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Ross Summer
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
29
|
Abstract
The inflammaging concept was introduced in 2000 by Prof. Franceschi. This was an evolutionary or rather a revolutionary conceptualization of the immune changes in response to a lifelong stress. This conceptualization permitted to consider the lifelong proinflammatory process as an adaptation which could eventually lead to either beneficial or detrimental consequences. This dichotomy is influenced by both the genetics and the environment. Depending on which way prevails in an individual, the outcome may be healthy longevity or pathological aging burdened with aging-related diseases. The concept of inflammaging has also revealed the complex, systemic nature of aging. Thus, this conceptualization opens the way to consider age-related processes in their complexity, meaning that not only the process but also all counter-processes should be considered. It has also opened the way to add new concepts to the original one, leading to better understanding of the nature of inflammaging and of aging itself. Finally, it showed the way towards potential multimodal interventions involving a holistic approach to optimize the aging process towards a healthy longevity.
Collapse
|
30
|
Li Z, Tian M, Wang G, Cui X, Ma J, Liu S, Shen B, Liu F, Wu K, Xiao X, Zhu C. Senotherapeutics: An emerging approach to the treatment of viral infectious diseases in the elderly. Front Cell Infect Microbiol 2023; 13:1098712. [PMID: 37065192 PMCID: PMC10094634 DOI: 10.3389/fcimb.2023.1098712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2023] [Indexed: 03/31/2023] Open
Abstract
In the context of the global COVID-19 pandemic, the phenomenon that the elderly have higher morbidity and mortality is of great concern. Existing evidence suggests that senescence and viral infection interact with each other. Viral infection can lead to the aggravation of senescence through multiple pathways, while virus-induced senescence combined with existing senescence in the elderly aggravates the severity of viral infections and promotes excessive age-related inflammation and multiple organ damage or dysfunction, ultimately resulting in higher mortality. The underlying mechanisms may involve mitochondrial dysfunction, abnormal activation of the cGAS-STING pathway and NLRP3 inflammasome, the role of pre-activated macrophages and over-recruited immune cells, and accumulation of immune cells with trained immunity. Thus, senescence-targeted drugs were shown to have positive effects on the treatment of viral infectious diseases in the elderly, which has received great attention and extensive research. Therefore, this review focused on the relationship between senescence and viral infection, as well as the significance of senotherapeutics for the treatment of viral infectious diseases.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Guolei Wang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xianghua Cui
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun’e Ma
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuan Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Chengliang Zhu, ; Xuan Xiao,
| |
Collapse
|
31
|
Chen Y, Song W, Li C, Wang J, Liu F, Ye Z, Ren P, Tong Y, Li J, Ou Z, Lee ACY, Cai JP, Wong BHY, Chan JFW, Yuen KY, Zhang AJX, Chu H. COVID-19 mRNA vaccine protects against SARS-CoV-2 Omicron BA.1 infection in diet-induced obese mice through boosting host innate antiviral responses. EBioMedicine 2023; 89:104485. [PMID: 36857860 PMCID: PMC9970285 DOI: 10.1016/j.ebiom.2023.104485] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Obesity is a worldwide epidemic and is considered a risk factor of severe manifestation of Coronavirus Disease 2019 (COVID-19). The pathogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses to infection, re-infection, and vaccination in individuals with obesity remain incompletely understood. METHODS Using the diet-induced obese (DIO) mouse model, we studied SARS-CoV-2 Alpha- and Omicron BA.1-induced disease manifestations and host immune responses to infection, re-infection, and COVID-19 mRNA vaccination. FINDINGS Unlike in lean mice, Omicron BA.1 and Alpha replicated to comparable levels in the lungs of DIO mice and resulted in similar degree of tissue damages. Importantly, both T cell and B cell mediated adaptive immune responses to SARS-CoV-2 infection or COVID-19 mRNA vaccination are impaired in DIO mice, leading to higher propensity of re-infection and lower vaccine efficacy. However, despite the absence of neutralizing antibody, vaccinated DIO mice are protected from lung damage upon Omicron challenge, accompanied with significantly more IFN-α and IFN-β production in the lung tissue. Lung RNAseq and subsequent experiments indicated that COVID-19 mRNA vaccination in DIO mice boosted antiviral innate immune response, including the expression of IFN-α, when compared to the nonvaccinated controls. INTERPRETATION Our findings suggested that COVID-19 mRNA vaccination enhances host innate antiviral responses in obesity which protect the DIO mice to a certain degree when adaptive immunity is suboptimal. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Wenchen Song
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Can Li
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jiaxuan Wang
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Feifei Liu
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhanhong Ye
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Peidi Ren
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Yihan Tong
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Junhua Li
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Zhihua Ou
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Andrew Chak-Yiu Lee
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jian-Piao Cai
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Bosco Ho-Yin Wong
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan Province, People's Republic of China; and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Academician Workstation of Hainan Province, Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan Province, People's Republic of China; and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jin-Xia Zhang
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China.
| | - Hin Chu
- Department of Microbiology, State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; Department of Infectious Disease and Microbiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, People's Republic of China.
| |
Collapse
|
32
|
Wang C, Hao X, Chen S. Calling for improved pulmonary and critical care medicine in China and beyond. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:1-2. [PMID: 39170875 PMCID: PMC11332819 DOI: 10.1016/j.pccm.2023.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 08/23/2024]
Affiliation(s)
- Chen Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiuyuan Hao
- Editorial Department of Chinese Medical Journal Pulmonary and Critical Care Medicine, Chinese Medical Association, Beijing 100052, China
| | - Simiao Chen
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Heidelberg Institute of Global Health, Faculty of Medicine and University Hospital, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
33
|
Kountouras J, Tzitiridou-Chatzopoulou M, Papaefthymiou A, Chatzopoulos D, Doulberis M. COVID-19 mRNA Vaccine Effectiveness against Elderly Frail People. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020202. [PMID: 36837403 PMCID: PMC9962607 DOI: 10.3390/medicina59020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/07/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
The frail, elderly population is often characterized by poor immunogenicity post COVID-19 mRNA vaccination. "Inflame-ageing" and "immune-senescence" are pathogenetic mechanisms that might explain this phenomenon. Complex interplay with cytokines and microbiota is also implicated in this inflammatory cascade. The abovementioned population, although very important from immunologic perspective, has barely been included in the mRNA vaccination clinical trials.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Correspondence: (J.K.); (M.D.)
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Gastroenterology, University Hospital of Larisa, 41110 Larisa, Greece
| | - Dimitrios Chatzopoulos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
- Correspondence: (J.K.); (M.D.)
| |
Collapse
|
34
|
Aguirre-Milachay E, León-Figueroa DA, Chumán-Sánchez M, Romani L, Runzer-Colmenares FM. Factors associated with mortality in patients hospitalized for COVID-19 admitted to a tertiary hospital in Lambayeque, Peru, during the first wave of the pandemic. PLoS One 2023; 18:e0285133. [PMID: 37167338 PMCID: PMC10174592 DOI: 10.1371/journal.pone.0285133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 04/15/2023] [Indexed: 05/13/2023] Open
Abstract
INTRODUCTION COVID-19 caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has spread worldwide, becoming a long-term pandemic. OBJECTIVES To analyze the factors associated with mortality in patients hospitalized for COVID-19 in a tertiary hospital in the Lambayeque region of Peru. METHODS A retrospective cohort study of patients with a diagnosis of COVID-19, hospitalized in a hospital in northern Peru, was conducted from March to September 2020. RESULTS Of the 297 patients studied, 69% were women, the mean age was 63.99 years (SD = ±15.33 years). Hypertension was the most frequent comorbidity (36.67%), followed by diabetes mellitus (24.67%) and obesity (8.33%). The probability of survival at 3 days of ICU stay was 65.3%, at 7 days 24.2%, and 0% on day 14. Risk factors associated with mortality in patients hospitalized for COVID-19 are age, male sex, tachypnea, low systolic blood pressure, low peripheral oxygen saturation, impaired renal function, elevated IL-6 and elevated D-dimer. CONCLUSIONS Mortality in hospitalized patients with COVID-19 was 51.18 per 100 persons, Mortality was found to be associated with hypertension, type of infiltrating, and sepsis.
Collapse
Affiliation(s)
- Edwin Aguirre-Milachay
- Servicio de Geriatría, Departamento de Medicina, Hospital Nacional Almanzor Aguinaga Asenjo, Chiclayo, Peru
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo, Peru
| | - Darwin A León-Figueroa
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo, Peru
- Emerge, Unidad de Investigación en Enfermedades Emergentes y Cambio Climático, Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Marisella Chumán-Sánchez
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo, Peru
- Sociedad Científica de Estudiantes de Medicina Veritas (SCIEMVE), Chiclayo, Perú
| | - Luccio Romani
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo, Peru
- Emerge, Unidad de Investigación en Enfermedades Emergentes y Cambio Climático, Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | |
Collapse
|
35
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [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: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
36
|
Interaction of aging and Immunosenescence: New therapeutic targets of aging. Int Immunopharmacol 2022; 113:109397. [DOI: 10.1016/j.intimp.2022.109397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
37
|
Inhaled particulate accumulation with age impairs immune function and architecture in human lung lymph nodes. Nat Med 2022; 28:2622-2632. [PMID: 36411343 PMCID: PMC9835154 DOI: 10.1038/s41591-022-02073-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 10/03/2022] [Indexed: 11/22/2022]
Abstract
Older people are particularly susceptible to infectious and neoplastic diseases of the lung and it is unclear how lifelong exposure to environmental pollutants affects respiratory immune function. In an analysis of human lymph nodes (LNs) from 84 organ donors aged 11-93 years, we found a specific age-related decline in lung-associated, but not gut-associated, LN immune function linked to the accumulation of inhaled atmospheric particulate matter. Increasing densities of particulates were found in lung-associated LNs with age, but not in the corresponding gut-associated LNs. Particulates were specifically contained within CD68+CD169- macrophages, which exhibited decreased activation, phagocytic capacity, and altered cytokine production compared with non-particulate-containing macrophages. The structures of B cell follicles and lymphatic drainage were also disrupted in lung-associated LNs with particulates. Our results reveal that the cumulative effects of environmental exposure and age may compromise immune surveillance of the lung via direct effects on immune cell function and lymphoid architecture.
Collapse
|
38
|
Ganguli S, Howlader S, Dey K, Barua S, Islam MN, Begum A, Sobahan MA, Chakraborty RR, Hawlader MDH, Biswas PK. Association of food habit with the COVID-19 severity and hospitalization: A cross-sectional study among the recovered individuals in Bangladesh. Nutr Health 2022; 28:771-782. [PMID: 36066026 PMCID: PMC9716059 DOI: 10.1177/02601060221124068] [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] [Indexed: 11/15/2022]
Abstract
Background: It was assumed that dietary habits might influence the status of COVID-19 patients. Aim: We aimed at the identification of association of dietary habits with the COVID-19 severity and hospitalization. Methods: It was a retrospective cross-sectional study (n = 1025). We used bivariate and multivariate analyses to correlate the association between self-reported dietary patterns and COVID-19 severity and hospitalization. Results: Dietary habits (black tea, milked tea, pickles, black caraway seeds, honey, fish, fruits, vegetables, garlic, onion and turmeric) were identified with lower risk of COVID-19 severity and hospitalization. Interestingly, the consumption frequency (one-, two- or three-times/day) of rice - the staple food in Bangladesh - was not associated with COVID-19 severity and hospitalization for comorbid patients. In contrast, a moderate rice-eating habit (two times/day) was strongly associated with the lower risk of severity and hospitalization for non-comorbid patients. However, for both comorbid and non-comorbid patients, consumption of black tea, milked tea, pickles and honey were associated with a lower likelihood of severity and hospitalization. Overall, a high consumption (three-times/day) of fish, fruits and vegetables, a moderate consumption of garlic, onion and turmeric spices and a daily intake of black/milked tea, and honey were associated with reduced risk of COVID-19 severity and hospitalization. Conclusions: To reduce the severity of COVID-19, a habitual practice of intaking black tea, milked tea, black caraway seeds and honey along with dietary habit (rice, fish and vegetables) and with a moderate consumption of ginger, garlic, onion, mixed aromatic spices (cinnamon + cardamom + cloves) and turmeric might be suggested.
Collapse
Affiliation(s)
- Sumon Ganguli
- Biomaterials Research Laboratory,
Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
| | - Sabbir Howlader
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
| | - Kamol Dey
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
| | - Suman Barua
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
| | - Md. Nazrul Islam
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
- School of Pharmacy, The University of
Queensland, Queensland, Australia
| | - Afroza Begum
- Department of Statistics, University of Chittagong, Chattogram, Bangladesh
| | - Md. Abdus Sobahan
- Department of Applied Chemistry and
Chemical Engineering, Faculty of Science, University of Chittagong, Chattogram, Bangladesh
| | - Rivu Raj Chakraborty
- Department of Surgery, Rangamati
Medical College and Hospital, Rangamati, Bangladesh
| | | | - Paritosh Kumar Biswas
- Department of Microbiology and
Veterinary Public Health, Chattogram Veterinary and Animal Sciences University,
Chattogram, Bangladesh
| |
Collapse
|
39
|
Liu Y, Xia K, Liu S, Wang W, Li G. Ginseng as a Key Immune Response Modulator in Chinese Medicine: From Antipandemic History to COVID-19 Management. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 51:19-34. [PMID: 36419254 DOI: 10.1142/s0192415x23500027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The cytokine storm plays an indispensable role in the severe and critical illness and death of the COVID-19 vulnerable population. Thus, suppressing the cytokine storm is of great significance. Ginseng is a traditional Chinese herb originally used for improving physiological conditions and ameliorating disease. Common throughout the history of ancient Chinese medicine is utilizing ginseng as a major ingredient to successfully fight various pandemics, and the most famous decoction is Renshen Baidu powder. In recent years, ginseng has been observed to provide preventive and therapeutic benefits in the treatment of various conditions by suppressing hyper-inflammation, inhibiting virus intrusion, and balancing the host's immunity. This paper summarizes the ancient Chinese medicine books' recordings of, the clinical practice of, and the laboratory exploration of ginseng for the treatment of pandemics and COVID-19. Ginseng and its active ingredients were found to downregulate inflammatory cytokines, upregulate anti-inflammatory cytokines, stimulate the secretion of the antiviral cytokine IFN-[Formula: see text], prevent viral entry and replication, and improve viral clearance. Furthermore, ginseng modulates both natural and acquired immunity during viral infection. Collectively, we propose that ginseng can act as a key immune response modulator against the cytokine storm of COVID-19. This paper may provide a new approach to discover specific medications using ginseng to combat COVID-19.
Collapse
Affiliation(s)
- Yanyi Liu
- Department of Respiratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China.,Beijing University of Chinese Medicine, Beijing 100029, P. R. China
| | - Kun Xia
- Department of Respiratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China
| | - Shixu Liu
- Department of Respiratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China
| | - Wei Wang
- Department of Respiratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China
| | - Guangxi Li
- Department of Respiratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China
| |
Collapse
|
40
|
Rossi E, Mutti L, Morrione A, Giordano A. Neuro-Immune Interactions in Severe COVID-19 Infection. Pathogens 2022; 11:1256. [PMID: 36365007 PMCID: PMC9699641 DOI: 10.3390/pathogens11111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus that has affected the world since 2019. Interstitial pneumonia is the most common clinical presentation, but additional symptoms have been reported, including neurological manifestations. Severe forms of infection, especially in elderly patients, present as an excessive inflammatory response called "cytokine storm", which can lead to acute respiratory distress syndrome (ARDS), multiorgan failure and death. Little is known about the relationship between symptoms and clinical outcomes or the characteristics of virus-host interactions. The aim of this narrative review is to highlight possible links between neurological involvement and respiratory damage mediated by pathological inflammatory pathways in SARS-CoV-2 infection. We will focus on neuro-immune interactions and age-related immunity decline and discuss some pathological mechanisms that contribute to negative outcomes in COVID-19 patients. Furthermore, we will describe available therapeutic strategies and their effects on COVID-19 neurological symptoms.
Collapse
Affiliation(s)
- Elena Rossi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Italian Group for Research and Therapy for Mesothelioma (GIMe), 27058 Voghera, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
41
|
Hagiya H, Hikita T, Habu T, Asada M, Yorifuji T, Toyooka S, Otsuka F, Nakayama M. Poor vaccine responsiveness towards third-dose mRNA vaccine of COVID-19 in Japanese older people. J Infect 2022; 85:436-480. [PMID: 35835411 PMCID: PMC9272660 DOI: 10.1016/j.jinf.2022.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Hideharu Hagiya
- Department of General Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan.
| | - Takao Hikita
- Office of Innovative Medicine, Organization for Research Strategy and Development, Okayama University, Okayama, Japan; Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Tomohiro Habu
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masaki Asada
- Departments of Medical Education, Kurashiki Central Hospital, Japan
| | - Takashi Yorifuji
- Department of Epidemiology, Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Fumio Otsuka
- Department of General Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Masanori Nakayama
- Office of Innovative Medicine, Organization for Research Strategy and Development, Okayama University, Okayama, Japan; Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany.
| |
Collapse
|
42
|
Luo S, Liang Y, Wong THT, Schooling CM, Au Yeung SL. Identifying factors contributing to increased susceptibility to COVID-19 risk: a systematic review of Mendelian randomization studies. Int J Epidemiol 2022; 51:1088-1105. [PMID: 35445260 PMCID: PMC9047195 DOI: 10.1093/ije/dyac076] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND To summarize modifiable factors for coronavirus disease 2019 (COVID-19) suggested by Mendelian randomization studies. METHODS In this systematic review, we searched PubMed, EMBASE and MEDLINE, from inception to 15 November 2021, for Mendelian randomization studies in English. We selected studies that assessed associations of genetically predicted exposures with COVID-19-related outcomes (severity, hospitalization and susceptibility). Risk of bias of the included studies was evaluated based on the consideration of the three main assumptions for instrumental variable analyses. RESULTS We identified 700 studies through systematic search, of which 50 Mendelian randomization studies were included. Included studies have explored a wide range of socio-demographic factors, lifestyle attributes, anthropometrics and biomarkers, predisposition to diseases and druggable targets in COVID-19 risk. Mendelian randomization studies suggested that increases in smoking, obesity and inflammatory factors were associated with higher risk of COVID-19. Predisposition to ischaemic stroke, combined bipolar disorder and schizophrenia, attention-deficit and hyperactivity disorder, chronic kidney disease and idiopathic pulmonary fibrosis was potentially associated with higher COVID-19 risk. Druggable targets, such as higher protein expression of histo-blood group ABO system transferase (ABO), interleukin (IL)-6 and lower protein expression of 2'-5' oligoadenylate synthetase 1 (OAS1) were associated with higher risk of COVID-19. There was no strong genetic evidence supporting the role of vitamin D, glycaemic traits and predisposition to cardiometabolic diseases in COVID-19 risk. CONCLUSION This review summarizes modifiable factors for intervention (e.g. smoking, obesity and inflammatory factors) and proteomic signatures (e.g. OAS1 and IL-6) that could help identify drugs for treating COVID-19.
Collapse
Affiliation(s)
- Shan Luo
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ying Liang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tommy Hon Ting Wong
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Catherine Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Environmental, Occupational, and Geospatial Health Sciences, School of Public Health and Health Policy, City University of New York, New York, USA
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
43
|
Jo N, Zhang R, Ueno H, Yamamoto T, Weiskopf D, Nagao M, Yamanaka S, Hamazaki Y. Aging and CMV Infection Affect Pre-existing SARS-CoV-2-Reactive CD8 + T Cells in Unexposed Individuals. FRONTIERS IN AGING 2022; 2:719342. [PMID: 35822004 PMCID: PMC9261342 DOI: 10.3389/fragi.2021.719342] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022]
Abstract
Age is a major risk factor for COVID-19 severity, and T cells play a central role in anti-SARS-CoV-2 immunity. Because SARS-CoV-2-cross-reactive T cells have been detected in unexposed individuals, we investigated the age-related differences in pre-existing SARS-CoV-2-reactive T cells. SARS-CoV-2-reactive CD4+ T cells from young and elderly individuals were mainly detected in the central memory fraction and exhibited similar functionalities and numbers. Naïve-phenotype SARS-CoV-2-reactive CD8+ T cell populations decreased markedly in the elderly, while those with terminally differentiated and senescent phenotypes increased. Furthermore, senescent SARS-CoV-2-reactive CD8+ T cell populations were higher in cytomegalovirus seropositive young individuals compared to seronegative ones. Our findings suggest that age-related differences in pre-existing SARS-CoV-2-reactive CD8+ T cells may explain the poor outcomes in elderly patients and that cytomegalovirus infection is a potential factor affecting CD8+ T cell immunity against SARS-CoV-2. Thus, this study provides insights for developing effective therapeutic and vaccination strategies for the elderly.
Collapse
Affiliation(s)
- Norihide Jo
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Alliance Laboratory for Advanced Medical Research, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Rui Zhang
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hideki Ueno
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Miki Nagao
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Yamanaka
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA, United States
| | - Yoko Hamazaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Laboratory of Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
44
|
LipoxinA4 as a Potential Prognostic Marker of COVID-19. J Lipids 2022; 2022:8527305. [PMID: 35812307 PMCID: PMC9259546 DOI: 10.1155/2022/8527305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
This pilot study aimed to determine early changes of LXA4 levels among the hospitalized patients confirmed as COVID-19 cases following the clinical management and its correlation with commonly used inflammatory markers, including erythrocyte sedimentation rate (ESR), c-reactive protein (CRP), and ferritin. Thirty-one adult hospitalized patients infected with the non-severe COVID-19 were included. LXA4 levels were measured at the baseline and 48-72 hours after hospitalization. Accordingly, ESR and CRP levels were collected on the first day of hospitalization. Moreover, the maximum serum ferritin levels were determined during the five days. LXA4 levels significantly increased at 48-72 hours compared to the baseline. ESR, CRP, and ferritin levels were positively correlated with the increased LXA4. In contrast, aging was shown to negatively correlate with the increased LXA4 levels. LXA4 may be known as a valuable marker to assess the treatment response among non-elderly patients with non-severe COVID-19. Furthermore, LXA4 could be considered as a potential treatment option under inflammatory conditions. Further studies are necessary to clarify LXA4 role in COVID-19 pathogenesis, as well as the balance between such pro-resolving mediators and inflammatory parameters.
Collapse
|
45
|
Coskun E, Kalil R, Yin A, Wehmeyer G, Hoffman K, Kopparam R, Phongtankuel V, Goyal P, Kaner R, Siegler E. Choices and Outcomes of the Oldest Old Admitted During the First Wave of COVID-19 in New York City. J Palliat Care 2022; 37:298-309. [PMID: 35502860 PMCID: PMC9066242 DOI: 10.1177/08258597221098130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objectives: Morbidity and mortality are higher in older adults with COVID-19, but their decisions about aggressive care, severity of disease, and outcomes during the first surge in New York City are not well characterized. We sought to determine if the oldest patients chose intubation and comfort care at different rates compared to younger geriatric patients. We also studied outcomes among patients admitted with severe disease and those who chose aggressive versus comfort care. Methods: This retrospective analysis used electronic health record data from patients 65 years and older at two medical centers in New York City admitted between 3/5/2020 and 5/15/2020. The primary outcome was comfort care orders, and secondary outcomes included death, palliative care consultation, goals of care discussion, code status, and ventilator weaning. Results: Of the 854 patients, 214 were in the oldest old (OO, age > = 85) group, 269 middle old (MO, age 75-84), and 371 young old (YO, age 65-74). Among those with serious disease, the OO were more likely to choose comfort care (45% vs. 21% MO and 6.8% YO), less likely to be intubated (17% vs. 37% MO and 44% YO), more likely to have a palliative care consult, more likely to be DNR/DNI on admission (60% vs. 17% MO and 9.3%% YO), and more likely to die during admission (65% vs. 42% MO and 21% YO) (all p-values < 0.001). Of all 216 intubated patients, 78% of the OO died, versus 66% of the MO and 36% of the YO (p = <0.001). Conclusions: Adults 85 and above admitted with COVID-19 were more likely to forego intubation and die with comfort-based care. Irrespective of intubation choice, patients 85 and older had a markedly poorer prognosis than other cohorts over 65.
Collapse
Affiliation(s)
- Emily Coskun
- Division of Geriatrics and Palliative Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA,Emily Coskun, Division of Geriatrics and Palliative Medicine, New York-Presbyterian Weill Cornell Medical Center, 525 East 68 St. New York, New York 10065.
| | - Ramsey Kalil
- Department of Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Andrew Yin
- Department of Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Graham Wehmeyer
- Department of Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Katherine Hoffman
- Division of Biostatistics and Epidemiology, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Rohini Kopparam
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Veerawat Phongtankuel
- Division of Geriatrics and Palliative Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Parag Goyal
- Division of Cardiology, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Robert Kaner
- Division of Pulmonary and Critical Care Medicine and Genetic Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| | - Eugenia Siegler
- Division of Geriatrics and Palliative Medicine, New York-Presbyterian Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
46
|
Aging and the COVID-19 pandemic: The inter-related roles of biology, physical wellbeing, social norms and global health systems. Maturitas 2022; 167:99-104. [PMCID: PMC9328837 DOI: 10.1016/j.maturitas.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
|
47
|
Yang JM, Moon SY, Lee JY, Agalliu D, Yon DK, Lee SW. COVID-19 Morbidity and Severity in Patients With Age-Related Macular Degeneration: A Korean Nationwide Cohort Study. Am J Ophthalmol 2022; 239:159-169. [PMID: 34102151 PMCID: PMC8179838 DOI: 10.1016/j.ajo.2021.05.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE To determine the potential association between age-related macular degeneration (AMD), a representative chronic age-related degenerative disease of the retina associated with inflammation and aging, and susceptibility to SARS-CoV-2 infection and severe COVID-19 outcomes. DESIGN Nationwide cohort study with propensity-score matching. METHODS A population-based nationwide cohort in Korea was examined. Data were obtained from the Health Insurance Review & Assessment Service of Korea, including all patients aged ≥40 years who underwent SARS-CoV-2 testing in South Korea between January 1, 2020 and May 15, 2020 (excluding self-referral). The primary outcome was SARS-CoV-2 test positivity and the secondary outcome was severe clinical outcome of COVID-19. RESULTS The unmatched cohort consisted of 135,435 patients who were tested for SARS-CoV-2: 4531 patients (3.3%) tested positive for SARS-CoV-2 and 5493 (4.1%) had AMD. After propensity score matching, exudative AMD was associated with an increased likelihood of susceptibility to SARS-CoV-2 infection (adjusted odds ratio [aOR], 1.50; 95% confidence interval [CI], 1.03-2.25), and a considerably greater risk of severe clinical outcomes of COVID-19 (aOR, 2.26; 95% CI, 1.02-5.26), but not any AMD and non-exudative AMD. CONCLUSIONS In a Korean nationwide cohort, data suggest that clinicians should be aware of the greater risk of susceptibility to severe clinical outcomes of COVID-19 in patients with exudative AMD. These findings provide an improved understanding of the relationship between the pathogenesis of COVID-19 and chronic neurological disorders.
Collapse
Affiliation(s)
- Jee Myung Yang
- From Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine (J.M.Y, J.Y.L); Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang, Republic of Korea; Department of Data Science, Sejong University College of Software Convergence, Seoul, Republic of Korea (S.Y.M, D.K.Y, S.W.L)
| | - Sung Yong Moon
- Department of Data Science, Sejong University College of Software Convergence, Seoul, Republic of Korea (S.Y.M, D.K.Y, S.W.L)
| | - Joo Yong Lee
- From Department of Ophthalmology, Asan Medical Center, University of Ulsan College of Medicine (J.M.Y, J.Y.L)
| | - Dritan Agalliu
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA (D.A)
| | - Dong Keon Yon
- Department of Data Science, Sejong University College of Software Convergence, Seoul, Republic of Korea (S.Y.M, D.K.Y, S.W.L); Center for Digital Health, Medical Science Research Institute, Kyung Hee University College of Medicine, Seoul, Republic of Korea (D.K.Y).
| | - Seung Won Lee
- Department of Data Science, Sejong University College of Software Convergence, Seoul, Republic of Korea (S.Y.M, D.K.Y, S.W.L); Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea (S.W.L).
| |
Collapse
|
48
|
Ntouros PA, Kravvariti E, Vlachogiannis NI, Pappa M, Trougakos IP, Terpos E, Tektonidou MG, Souliotis VL, Sfikakis PP. Oxidative stress and endogenous DNA damage in blood mononuclear cells may predict anti-SARS-CoV-2 antibody titers after vaccination in older adults. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166393. [PMID: 35314351 PMCID: PMC8930778 DOI: 10.1016/j.bbadis.2022.166393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Immune senescence in the elderly has been associated with chronic oxidative stress and DNA damage accumulation. Herein we tested the hypothesis that increased endogenous DNA damage and oxidative stress in peripheral blood mononuclear cells of older adults associate with diminished humoral immune response to SARS-CoV-2 vaccination. Increased oxidative stress and DNA double-strand breaks (DSBs) were detected in 9 non-immunocompromised individuals aged 80-96 years compared to 11 adults aged 27-44 years, before, as well as on days 1 and 14 after the first dose, and on day 14 after the second dose of the BNT162B2-mRNA vaccine (all p < 0.05). SARS-CoV-2 vaccination induced a resolvable increase in oxidative stress and DNA damage, but individual DSB-repair efficiency was unaffected by vaccination irrespective of age, confirming vaccination safety. Individual titers of anti-Spike-Receptor Binding Domain (S-RBD)-IgG antibodies, and the neutralizing capacity of circulating anti-SARS-CoV-2 antibodies, measured on day 14 after the second dose in all participants, correlated inversely with the corresponding pre-vaccination endogenous oxidative stress and DSB levels (all p < 0.05). In particular, a strong inverse correlation of individual pre-vaccination DSB levels with both the respective anti-S-RBD-IgG antibodies titers (r = -0.867) and neutralizing capacity of circulating anti-SARS-CoV-2 antibodies (r = -0.983) among the 9 older adults was evident. These findings suggest that humoral responses to SARS-CoV-2 vaccination may be weaker when immune cells are under oxidative and/or genomic stress. Whether such measurements may serve as biomarkers of vaccine efficacy in older adults warrants further studies.
Collapse
Affiliation(s)
- Panagiotis A Ntouros
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| | - Evrydiki Kravvariti
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece; Postgraduate Medical Studies in the Physiology of Aging and Geriatric Syndromes, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos I Vlachogiannis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria Pappa
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Vassilis L Souliotis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece; Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece; Postgraduate Medical Studies in the Physiology of Aging and Geriatric Syndromes, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
49
|
Junqueira C, Crespo Â, Ranjbar S, de Lacerda LB, Lewandrowski M, Ingber J, Parry B, Ravid S, Clark S, Schrimpf MR, Ho F, Beakes C, Margolin J, Russell N, Kays K, Boucau J, Das Adhikari U, Vora SM, Leger V, Gehrke L, Henderson LA, Janssen E, Kwon D, Sander C, Abraham J, Goldberg MB, Wu H, Mehta G, Bell S, Goldfeld AE, Filbin MR, Lieberman J. FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature 2022; 606:576-584. [PMID: 35385861 PMCID: PMC10071495 DOI: 10.1038/s41586-022-04702-4] [Citation(s) in RCA: 385] [Impact Index Per Article: 128.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2022] [Indexed: 12/26/2022]
Abstract
SARS-CoV-2 can cause acute respiratory distress and death in some patients1. Although severe COVID-19 is linked to substantial inflammation, how SARS-CoV-2 triggers inflammation is not clear2. Monocytes and macrophages are sentinel cells that sense invasive infection to form inflammasomes that activate caspase-1 and gasdermin D, leading to inflammatory death (pyroptosis) and the release of potent inflammatory mediators3. Here we show that about 6% of blood monocytes of patients with COVID-19 are infected with SARS-CoV-2. Monocyte infection depends on the uptake of antibody-opsonized virus by Fcγ receptors. The plasma of vaccine recipients does not promote antibody-dependent monocyte infection. SARS-CoV-2 begins to replicate in monocytes, but infection is aborted, and infectious virus is not detected in the supernatants of cultures of infected monocytes. Instead, infected cells undergo pyroptosis mediated by activation of NLRP3 and AIM2 inflammasomes, caspase-1 and gasdermin D. Moreover, tissue-resident macrophages, but not infected epithelial and endothelial cells, from lung autopsies from patients with COVID-19 have activated inflammasomes. Taken together, these findings suggest that antibody-mediated SARS-CoV-2 uptake by monocytes and macrophages triggers inflammatory cell death that aborts the production of infectious virus but causes systemic inflammation that contributes to COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Caroline Junqueira
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Ângela Crespo
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Shahin Ranjbar
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Luna B de Lacerda
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Mercedes Lewandrowski
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jacob Ingber
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Blair Parry
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA
| | - Sagi Ravid
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Sarah Clark
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marie Rose Schrimpf
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Felicia Ho
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Caroline Beakes
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA
| | - Justin Margolin
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA
| | - Nicole Russell
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA
| | - Kyle Kays
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA
| | - Julie Boucau
- Ragon Institute, Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA, USA
| | - Upasana Das Adhikari
- Ragon Institute, Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA, USA
| | - Setu M Vora
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Valerie Leger
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lee Gehrke
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lauren A Henderson
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Erin Janssen
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Douglas Kwon
- Ragon Institute, Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard Medical School, Cambridge, MA, USA
| | - Chris Sander
- cBio Center, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marcia B Goldberg
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Center for Bacterial Pathogenesis, Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Hao Wu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gautam Mehta
- Institute for Liver and Digestive Health, University College London, London, UK
- Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Steven Bell
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Anne E Goldfeld
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael R Filbin
- Emergency Medicine, Institute for Patient Care, Massachusetts General Hospital, Boston, MA, USA.
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Aloul KM, Nielsen JE, Defensor EB, Lin JS, Fortkort JA, Shamloo M, Cirillo JD, Gombart AF, Barron AE. Upregulating Human Cathelicidin Antimicrobial Peptide LL-37 Expression May Prevent Severe COVID-19 Inflammatory Responses and Reduce Microthrombosis. Front Immunol 2022; 13:880961. [PMID: 35634307 PMCID: PMC9134243 DOI: 10.3389/fimmu.2022.880961] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/11/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is characterized by hyperactivation by inflammatory cytokines and recruitment of macrophages, neutrophils, and other immune cells, all hallmarks of a strong inflammatory response that can lead to severe complications and multi-organ damage. Mortality in COVID-19 patients is associated with a high prevalence of neutrophil extracellular trap (NET) formation and microthrombosis that are exacerbated by hyperglycemia, diabetes, and old age. SARS-CoV-2 infection in humans and non-human primates have revealed long-term neurological consequences of COVID-19, possibly concomitant with the formation of Lewy bodies in the brain and invasion of the nervous system via the olfactory bulb. In this paper, we review the relevance of the human cathelicidin LL-37 in SARS-CoV-2 infections. LL-37 is an immunomodulatory, host defense peptide with direct anti-SARS-CoV-2 activity, and pleiotropic effects on the inflammatory response, neovascularization, Lewy body formation, and pancreatic islet cell function. The bioactive form of vitamin D and a number of other compounds induce LL-37 expression and one might predict its upregulation, could reduce the prevalence of severe COVID-19. We hypothesize upregulation of LL-37 will act therapeutically, facilitating efficient NET clearance by macrophages, speeding endothelial repair after inflammatory tissue damage, preventing α-synuclein aggregation, and supporting blood-glucose level stabilization by facilitating insulin release and islet β-cell neogenesis. In addition, it has been postulated that LL-37 can directly bind the S1 domain of SARS-CoV-2, mask angiotensin converting enzyme 2 (ACE2) receptors, and limit SARS-CoV-2 infection. Purposeful upregulation of LL-37 could also serve as a preventative and therapeutic strategy for SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Karim M. Aloul
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - Josefine Eilsø Nielsen
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Erwin B. Defensor
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jennifer S. Lin
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - John A. Fortkort
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| | - Mehrdad Shamloo
- Department of Neurosurgery, School of Medicine, Stanford University, Stanford, CA, United States
| | - Jeffrey D. Cirillo
- Department of Microbial Pathogenesis and Immunology, Texas A&M College of Medicine, Bryan, TX, United States
| | - Adrian F. Gombart
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, United States
- The Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Annelise E. Barron
- Department of Bioengineering, Schools of Medicine and of Engineering, Stanford University, Stanford, CA, United States
| |
Collapse
|