1
|
Zhou K, Duan G, Liu Y, Peng B, Zhou X, Qin L, Liang L, Wei Y, Zhang Q, Li X, Qin H, Lai Y, Lu Y, Zhang Y, Huang J, Huang J, Ouyang Y, Bin B, Zhao M, Liu J, Yang J, Deng D. Persistent alterations in gray matter in COVID-19 patients experiencing sleep disturbances: a 3-month longitudinal study. Neural Regen Res 2025; 20:3013-3024. [PMID: 38934390 PMCID: PMC11826451 DOI: 10.4103/nrr.nrr-d-23-01651] [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: 10/05/2023] [Revised: 01/13/2024] [Accepted: 04/19/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00030/figure1/v/2024-11-26T163120Z/r/image-tiff Sleep disturbances are among the most prevalent neuropsychiatric symptoms in individuals who have recovered from severe acute respiratory syndrome coronavirus 2 infections. Previous studies have demonstrated abnormal brain structures in patients with sleep disturbances who have recovered from coronavirus disease 2019 (COVID-19). However, neuroimaging studies on sleep disturbances caused by COVID-19 are scarce, and existing studies have primarily focused on the long-term effects of the virus, with minimal acute phase data. As a result, little is known about the pathophysiology of sleep disturbances in the acute phase of COVID-19. To address this issue, we designed a longitudinal study to investigate whether alterations in brain structure occur during the acute phase of infection, and verified the results using 3-month follow-up data. A total of 26 COVID-19 patients with sleep disturbances (aged 51.5 ± 13.57 years, 8 women and 18 men), 27 COVID-19 patients without sleep disturbances (aged 47.33 ± 15.98 years, 9 women and 18 men), and 31 age- and gender-matched healthy controls (aged 49.19 ± 17.51 years, 9 women and 22 men) were included in this study. Eleven COVID-19 patients with sleep disturbances were included in a longitudinal analysis. We found that COVID-19 patients with sleep disturbances exhibited brain structural changes in almost all brain lobes. The cortical thicknesses of the left pars opercularis and left precuneus were significantly negatively correlated with Pittsburgh Sleep Quality Index scores. Additionally, we observed changes in the volume of the hippocampus and its subfield regions in COVID-19 patients compared with the healthy controls. The 3-month follow-up data revealed indices of altered cerebral structure (cortical thickness, cortical grey matter volume, and cortical surface area) in the frontal-parietal cortex compared with the baseline in COVID-19 patients with sleep disturbances. Our findings indicate that the sleep disturbances patients had altered morphology in the cortical and hippocampal structures during the acute phase of infection and persistent changes in cortical regions at 3 months post-infection. These data improve our understanding of the pathophysiology of sleep disturbances caused by COVID-19.
Collapse
Affiliation(s)
- Kaixuan Zhou
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Gaoxiong Duan
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Ying Liu
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Bei Peng
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaoyan Zhou
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lixia Qin
- Department of Sleep Medicine, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lingyan Liang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yichen Wei
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qingping Zhang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaocheng Li
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Haixia Qin
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yinqi Lai
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yian Lu
- Department of Sleep Medicine, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yan Zhang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiazhu Huang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jinli Huang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yinfei Ouyang
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Bolin Bin
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Mingming Zhao
- Department of Sleep Medicine, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jianrong Yang
- Guangxi Clinical Research Center for Sleep Medicine, the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Demao Deng
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Radiology, the People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
2
|
Wang J, Gao S, Cui Y, Liu XZ, Chen XX, Hang CH, Li W. Remote Organ Damage Induced by Stroke: Molecular Mechanisms and Comprehensive Interventions. Antioxid Redox Signal 2025. [PMID: 40170638 DOI: 10.1089/ars.2024.0720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Significance: Damage after stroke is not only limited to the brain but also often occurs in remote organs, including the heart, lung, liver, kidney, digestive tract, and spleen, which are frequently affected by complex pathophysiological changes. The organs in the human body are closely connected, and signals transmitted through various molecular substances could regulate the pathophysiological changes of remote organs. Recent Advances: The latest studies have shown that inflammatory response plays an important role in remote organ damage after stroke, and can aggravate remote organ damage by activating oxidative stress, sympathetic axis, and hypothalamic axis, and disturbing immunological homeostasis. Remote organ damage can also cause damage to the brain, aggravating inflammatory response and oxidative damage. Critical Issues: Therefore, an in-depth exploration of inflammatory and oxidative mechanisms and adopting corresponding comprehensive intervention strategies have become necessary to reduce damage to remote organs and promote brain protection. Future Directions: The comprehensive intervention strategy involves multifaceted treatment methods such as inflammation regulation, antioxidants, and neural stem cell differentiation. It provides a promising treatment alternative for the comprehensive recovery of stroke patients and an inspiration for future research and treatment. The various organs of the human body are interconnected at the molecular level. Only through comprehensive intervention at the molecular and organ levels can we save remote organ damage and protect the brain after stroke to the greatest extent. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Yue Cui
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xun-Zhi Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Xiang-Xin Chen
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Neurosurgical Institute, Nanjing University, Nanjing, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
3
|
Tcholadze G, Pantsulaia I, Ratiani L, Kopaleishvili L, Bolotashvili T, Jorbenadze A, Chikovani T. The Prognostic Value of Circulating Cytokines and Complete Blood Count-Based Inflammatory Markers in COVID-19 Patients With Atrial Fibrillation. Cardiol Res 2025; 16:153-160. [PMID: 40051670 PMCID: PMC11882233 DOI: 10.14740/cr2027] [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: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a high burden of cardiovascular disease, which has been worsened during the coronavirus disease 2019 (COVID-19) pandemic. The purpose of this study was to assess the association between clinical markers, especially interleukin-6 (IL-6) and other inflammatory biomarkers, and the severity of COVID-19 in patients with AF. Methods This retrospective cohort study categorized patients based on clinical presentations and laboratory results to investigate the prognostic significance of inflammatory markers in COVID-19 outcomes among those with AF. The study included 100 hospitalized COVID-19 patients aged between 40 to 80 years and was conducted at the Chapidze Hospital in Tbilisi, Georgia. Patients were then grouped by disease severity according to computed tomography (CT) scores, clinical symptoms, respiratory rate and oxygen saturation. Levels of IL-6 were obtained at three time points during hospitalization. A broad range of laboratory tests, including C-reactive protein (CRP), ferritin, and D-dimer, were also conducted. Results Patients with AF demonstrated significantly elevated levels of IL-6 (P = 0.024), CRP (P = 0.001), and ferritin (P < 0.001), suggesting a severe inflammatory response. D-dimer levels were also notably higher in the AF group (P < 0.005), indicating an increased risk of thrombotic complications. Oxygen saturation levels were significantly lower (P = 0.004) and CT scores higher in patients with AF. Furthermore, the length of hospitalization was longer among patients with AF (median duration significantly higher, P = 0.032), indicating a more severe disease course. Conclusions The proinflammatory markers such as IL-6 are independent predictive markers of COVID-19 severity in AF patients. Overall, it highlights urgent treatment approaches, such as available anti-inflammatory drugs, for COVID-19 patients with arrhythmias. Combining these biomarkers into clinical routines helps us better identify patients at risk and how to treat them.
Collapse
Affiliation(s)
- Giorgi Tcholadze
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ia Pantsulaia
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
- Vl. Bakhutashvili Institute of Medical Biotechnology, Tbilisi State Medical University, Tbilisi 0159, Georgia
| | | | | | | | | | - Tinatin Chikovani
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| |
Collapse
|
4
|
Frank T, Bruckmann P, Binneböse M, Wallis H, Elgner M, Junne F, Massag J, Mikolajczyk R, Schurr M, Giel K, Wedekind L, Kuhn J, Gündel H, Müller AM, Beckmann P, Lahmann C, Allwang C. [Psychosocial aspects of Long COVID - A psychotherapeutic treatment manual from the PsyLoCo project]. Psychother Psychosom Med Psychol 2025; 75:127-131. [PMID: 40179895 DOI: 10.1055/a-2535-0607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Long COVID describes long-term health sequelae after a SARS-CoV-2 infection. Despite various theoretical approaches, the pathogenesis of long COVID is not yet fully understood and the diagnostic criteria are vague. This makes the development of targeted, cause-specific therapies challenging. In addition to the symptoms, many patients suffer from various psychosocial burdens and a significantly reduced quality of life. In order to address this, psychosomatic or psychotherapeutic interventions should be used in addition to symptom-oriented treatment. Within the PsyLoCo project, a psychotherapeutic treatment manual was developed that is tailored to the specific needs of long COVID patients. In four modules, it addresses (1) coping and distress management, (2) physical complaints and pain, (3) chronic fatigue and affective symptoms and (4) social and working life. The manual was tested for feasibility and effectiveness in a multicentre, randomized controlled pilot study. The contents of this treatment manual are presented below.
Collapse
Affiliation(s)
- Tamara Frank
- Klinik für Psychosomatische Medizin und Psychotherapie, Klinikum rechts der Isar, School of Medicine and Health, Technische Universität München
| | - Paul Bruckmann
- Klinik für Psychosomatische Medizin und Psychotherapie, Klinikum rechts der Isar, School of Medicine and Health, Technische Universität München
| | - Marius Binneböse
- Universitätsklinik für Psychosomatische Medizin und Psychotherapie, Otto-von-Guericke-Universität Magdeburg
| | - Hannah Wallis
- Universitätsklinik für Psychosomatische Medizin und Psychotherapie, Otto-von-Guericke-Universität Magdeburg
| | - Melanie Elgner
- Universitätsklinik für Psychosomatische Medizin und Psychotherapie, Otto-von-Guericke-Universität Magdeburg
| | - Florian Junne
- Universitätsklinik für Psychosomatische Medizin und Psychotherapie, Otto-von-Guericke-Universität Magdeburg
| | - Janka Massag
- Institut für Medizinische Epidemiologie, Biometrie und Informatik, Profilzentrum Gesundheitswissenschaften, Martin-Luther-Universität Halle-Wittenberg, Halle
| | - Rafael Mikolajczyk
- Institut für Medizinische Epidemiologie, Biometrie und Informatik, Profilzentrum Gesundheitswissenschaften, Martin-Luther-Universität Halle-Wittenberg, Halle
| | - Marisa Schurr
- Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Tübingen
| | - Katrin Giel
- Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Tübingen
- German Center for Mental Health (DZPG), Standort Tübingen
| | - Lisa Wedekind
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Ulm
| | - Julia Kuhn
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Ulm
| | - Harald Gündel
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Ulm
| | - Anne-Maria Müller
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Freiburg
| | - Pauline Beckmann
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Freiburg
| | - Claas Lahmann
- Klinik für Psychosomatische Medizin und Psychotherapie, Universitätsklinikum Freiburg
| | - Christine Allwang
- Klinik für Psychosomatische Medizin und Psychotherapie, Klinikum rechts der Isar, School of Medicine and Health, Technische Universität München
| |
Collapse
|
5
|
Li K, Pan Y, Song X, Yang B, Wang H, Yang F, Liu Q, Lin X, Zhao S, Yuan Y, Zhang Z, Zhang B, Fan F, Ma D. Clinical characteristics and outcomes of acute myocardial infarction during the COVID-19 pandemic: a multicenter retrospective cohort study in Northern China. BMC Cardiovasc Disord 2025; 25:226. [PMID: 40148803 PMCID: PMC11948736 DOI: 10.1186/s12872-025-04686-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The impacts of COVID-19 on acute myocardial infarction (AMI) care were heterogeneous. The study aims to analyze the clinical characteristics and outcomes of AMI patients in China during different stages of the COVID-19 pandemic. METHODS This is a multicenter retrospective cohort study in Shanxi Province of northern China. Patients diagnosed with AMI during the zero-case, lockdown, and outbreak periods were included. Characteristics and outcomes were analyzed according to time periods and COVID-19 infection. The primary outcome was in-hospital mortality. Additional outcomes included reperfusion times, coronary angiographic measures, procedure or AMI-associated complications, arrhythmia, other adverse events, and left ventricular systolic dysfunction (LVSD). RESULTS The study included 1021 AMI patients, with 393, 250, and 378 from the zero-case, lockdown, and outbreak periods. No differences in in-hospital mortality or other adverse events were found by time periods. By infection status, 264 patients were COVID-positive, and 706 were COVID-negative. The COVID-positive ST-elevation myocardial infarction population had longer symptom-to-first medical contact (3.07 vs. 2.31, p = 0.026), pre-hospital time (4.58 vs. 3.67, p = 0.032), door-to-balloon (1.20 vs. 1.08, p = 0.046), and total ischemic time (5.80 vs. 4.70, p = 0.011). No differences in other in-hospital outcomes were found, except that multivariate logistic regression analysis demonstrated COVID-19 infection was correlated with increased risks of LVSD (OR 1.73, 95% CI 1.11-2.69, p = 0.015). CONCLUSIONS In-hospital mortality did not differ by time period or COVID-19 infection status. The COVID-positive AMI patients had longer reperfusion times and higher risks of LVSD. AMI treatments were impacted during the pandemic, and measures are warranted to minimize the reperfusion time.
Collapse
Affiliation(s)
- Kang Li
- Department of Cardiology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China.
- Department of Cardiology, Taiyuan Central Hospital, No.1 East Sandao Lane, Xinghualing District, Taiyuan, 030009, Shanxi, China.
| | - Yannan Pan
- Department of Cardiology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xiaojian Song
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030002, Shanxi, China
| | - Bin Yang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Huifeng Wang
- Department of Cardiology, Taigang General Hospital, Taiyuan, 030003, Shanxi, China
| | - Fan Yang
- Department of Cardiology, Taiyuan Central Hospital, No.1 East Sandao Lane, Xinghualing District, Taiyuan, 030009, Shanxi, China
| | - Quanbao Liu
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030002, Shanxi, China
| | - Xinhong Lin
- Department of Cardiology, Taiyuan Central Hospital, No.1 East Sandao Lane, Xinghualing District, Taiyuan, 030009, Shanxi, China
| | - Shuzhen Zhao
- Department of Cardiology, Taiyuan Central Hospital, No.1 East Sandao Lane, Xinghualing District, Taiyuan, 030009, Shanxi, China
| | - Yuqi Yuan
- Department of Cardiology, Jincheng People's Hospital, Jincheng, 048026, Shanxi, China
| | - Ze Zhang
- The Ninth School of clinical medicine, Shanxi Medical University, Taiyuan, 030009, Shanxi, China
| | - Bin Zhang
- The Ninth School of clinical medicine, Shanxi Medical University, Taiyuan, 030009, Shanxi, China
| | - Fangfang Fan
- Department of Cardiology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Dengfeng Ma
- Department of Cardiology, Taiyuan Central Hospital, No.1 East Sandao Lane, Xinghualing District, Taiyuan, 030009, Shanxi, China.
| |
Collapse
|
6
|
Lu C, Deng W, Qiao Z, Sun W, Xu W, Li T, Wang F. Effects of early-life air pollution exposure on childhood COVID-19 infection and sequelae in China. JOURNAL OF HAZARDOUS MATERIALS 2025; 491:137940. [PMID: 40107106 DOI: 10.1016/j.jhazmat.2025.137940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND While ambient air pollution has been associated with COVID-19 outcomes, the role of early-life exposure in childhood COVID-19 infection and sequelae remains unexplored. OBJECTIVES To assess the associations between early-life exposure to ambient air pollutants during and childhood COVID-19 infection and sequelae. METHODS This cross-sectional retrospective cohort study surveyed families with children aged 3-6 years in families across nine Chinese cities between December 2019 and May 2023. The primary outcomes were doctor-diagnosed childhood COVID-19 infection and sequelae. Individual exposure to PM2.5, PM2.5-10, PM10, SO2, NO2, CO, O3, and temperature were estimated. RESULTS Among 20,012 children from 60,036 participants, 5.81 % were diagnosed with COVID-19 infection, and 1.72 % had sequelae. Prenatal CO exposure was associated with higher infection risk (OR: 1.33; 95 % CI: 1.05-1.69 per IQR increase). SO2 exposure during the first trimester (OR: 3.02; 95 % CI: 1.20-7.61), second trimester (OR: 4.00; 95 % CI: 1.56-10.27) and third trimester (OR: 3.84; 95 % CI: 1.69-8.76) of pregnancy and the first year of life (OR: 8.43; 95 % CI: 1.80-39.48) was strongly associated with sequelae. Pre-existing allergies and coarser particulate matter (PM2.5-10 and PM10) amplified these associations. High relative humidity significantly increased the effect of exposure to NO2 during four-six months before pregnancy and the second trimester of pregnancy, as well as O3 exposure during the first year on childhood COVID-19 infection. CONCLUSIONS Early-life exposure to air pollutants and interactions with allergic conditions and coarser particles influence childhood COVID-19 risks.
Collapse
Affiliation(s)
- Chan Lu
- XiangYa School of Public Health, Central South University, Changsha 410013, China; FuRong Laboratory, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Low Carbon Healthy Building, Central South University, Changsha 410083, China.
| | - Wen Deng
- XiangYa School of Public Health, Central South University, Changsha 410013, China
| | - Zipeng Qiao
- XiangYa School of Public Health, Central South University, Changsha 410013, China
| | - Wenying Sun
- XiangYa School of Public Health, Central South University, Changsha 410013, China
| | - Wanxue Xu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300012, China; Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin 300012, China
| | - Ting Li
- Biomedical Engineering Institute, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, China
| | - Faming Wang
- Centre for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an 710054, China.
| |
Collapse
|
7
|
Tukaj S, Sitna M, Sitko K. The impact of the mRNA COVID-19 vaccine on the Th-like cytokine profile in individuals with no history of COVID-19: insights into autoimmunity targeting heat shock proteins. Front Immunol 2025; 16:1549739. [PMID: 40160814 PMCID: PMC11949786 DOI: 10.3389/fimmu.2025.1549739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Although some reports suggest that COVID-19 vaccination may exacerbate existing autoimmune diseases or trigger new-onset cases, a definitive causal relationship between the vaccines and these conditions has not been established. Several potential mechanisms have been proposed to explain this association, including: (i) molecular mimicry, which refers to a structural similarity between SARS-CoV-2 and human antigens; (ii) bystander activation, involving both B and T lymphocytes; and (iii) the effects of adjuvants. In this study, we investigated whether two doses of the mRNA COVID-19 vaccine influenced blood cytokine levels associated with major T helper cell populations, which are known to play a significant role in autoimmunity and revisited the role of the humoral autoimmune response directed against heat shock proteins (Hsps) in individuals with no history of COVID-19. While no significant differences were found in the levels of IFN-γ, IL-6, IL-22, IL-4, IL-8, IL-10, and IL-17A, between vaccinated and unvaccinated people, several positive correlations were observed between serum cytokine levels and circulating autoantibodies directed against self-Hsps exclusively in vaccinated individuals. These findings suggest that the mRNA COVID-19 vaccine does not impact cytokines involved in the pathogenesis of autoimmune diseases. Further research is required to evaluate the safety of COVID-19 vaccination in patients with autoimmune conditions, particularly those in whom anti-Hsps autoantibodies are suspected to contribute to disease development.
Collapse
Affiliation(s)
- Stefan Tukaj
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Gdańsk, Poland
| | | | | |
Collapse
|
8
|
Iroungou BA, Nze O A, M Kandet Y H, Longo-Pendy NM, Mezogho-Obame ND, Dikoumba AC, Mangouka GL. Interest of D-dimer level, severity of COVID-19 and cost of management in Gabon. World J Crit Care Med 2025; 14:100486. [DOI: 10.5492/wjccm.v14.i1.100486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is strongly associated with an increased risk of thrombotic events, including severe outcomes such as pulmonary embolism. Elevated D-dimer levels are a critical biomarker for assessing this risk. In Gabon, early implementation of anticoagulation therapy and D-dimer testing has been crucial in managing COVID-19. This study hypothesizes that elevated D-dimer levels are linked to increased COVID-19 severity.
AIM To determine the impact of D-dimer levels on COVID-19 severity and their role in guiding clinical decisions.
METHODS This retrospective study analyzed COVID-19 patients admitted to two hospitals in Gabon between March 2020 and December 2023. The study included patients with confirmed COVID-19 diagnoses and available D-dimer measurements at admission. Data on demographics, clinical outcomes, D-dimer levels, and healthcare costs were collected. COVID-19 severity was classified as non-severe (outpatients) or severe (inpatients). A multivariable logistic regression model was used to assess the relationship between D-dimer levels and disease severity, with adjusted odds ratios (OR) and 95%CI.
RESULTS A total of 3004 patients were included, with a mean age of 50.17 years, and the majority were female (53.43%). Elevated D-dimer levels were found in 65.81% of patients, and 57.21% of these experienced severe COVID-19. Univariate analysis showed that patients with elevated D-dimer levels had 3.33 times higher odds of severe COVID-19 (OR = 3.33, 95%CI: 2.84-3.92, P < 0.001), and this association remained significant in the multivariable analysis, adjusted for age, sex, and year of collection. The financial analysis revealed a substantial burden, particularly for uninsured patients.
CONCLUSION D-dimer predicts COVID-19 severity and guides treatment, but the high cost of anticoagulant therapy highlights the need for policies ensuring affordable access in resource-limited settings like Gabon.
Collapse
Affiliation(s)
- Berthe A Iroungou
- Unité Mixte de Recherche, Centre International de Recherches Médicales de Franceville et le Service de Santé Militaire, Libreville 20404, Estuaire, Gabon
| | - Arnaud Nze O
- AP-HP Health Economics, Research Unit, Htel Dieu Hospital, Paris 75004, Ile-de-France, France
| | - Helga M Kandet Y
- Laboratory, Hôpital D'Instruction des Armées D'Akanda, Libreville 20404, Estuaire, Gabon
| | - Neil-Michel Longo-Pendy
- Unité de Recherche en Écologie de la Santé, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville 769, Gabon
| | - Nina D Mezogho-Obame
- Laboratory, Hôpital D'Instruction des Armées Omar Bongo Ondimba, Libreville 20404, Estuaire, Gabon
| | - Annicet-Clotaire Dikoumba
- Unité Mixte de Recherche, Centre International de Recherches Médicales de Franceville et le Service de Santé Militaire, Libreville 20404, Estuaire, Gabon
| | - Guignali L Mangouka
- Department of Medecine Polyvalente, Hôpital D'Instruction des Armées Omar Bongo Ondimba, Libreville 20404, Estuaire, Gabon
| |
Collapse
|
9
|
Kim HJ, Suh JH, Kim MH, Choi MG, Chun EM. Broad-Spectrum Adverse Events of Special Interests Based on Immune Response Following COVID-19 Vaccination: A Large-Scale Population-Based Cohort Study. J Clin Med 2025; 14:1767. [PMID: 40095916 PMCID: PMC11900331 DOI: 10.3390/jcm14051767] [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: 12/24/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background/Objectives: Current studies on adverse events related to the COVID-19 vaccine have predominantly focused on severe, life-threatening side effects. However, numerous less severe but common adverse events (AEs) remain underreported and insufficiently investigated despite their potential impact. Methods: This population-based cohort study investigated the cumulative incidence rate (cIR) and risk of the broad-spectrum AEs of special interests (AESIs) based on immune response, including gynecological, dermatological, ophthalmological, otologic, and dental problems, following COVID-19 vaccination. Results: Among 4,203,887 individuals in Seoul, South Korea, the final analysis included 1,458,557 vaccinated subjects and 289,579 non-vaccinated subjects after the exclusion of underlying diseases. The cIR of AESIs for three months was significantly higher in vaccinated subjects than in non-vaccinated subjects, except for endometriosis. The vaccination significantly increased the risks of all the AESIs except for visual impairment. The risk of alopecia showed the highest HRs (HR [95% CI] = 2.40 [1.90-3.03]) among the AESIs following COVID-19 vaccination. Among the vaccinated subjects, heterologous vaccination was associated with the increased risk of most of the AESIs. Conclusions: Our findings suggest that clinicians should closely recognize and follow up on various COVID-19 vaccine-related AEs due to their unknown impact, even if they may not be serious at present.
Collapse
Affiliation(s)
- Hong Jin Kim
- Department of Orthopaedic Surgery, Kyung-in Regional Military Manpower Administration, Suwon 16440, Republic of Korea;
- Department of Orthopaedic Surgery, Inje University Sanggye Paik Hospital, College of Medicine, Inje University, Seoul 01757, Republic of Korea
| | - Jee Hyun Suh
- Department of Rehabilitation Medicine, College of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea;
| | - Min-Ho Kim
- Informatization Department, Ewha Womans University Seoul Hospital, Seoul 07804, Republic of Korea;
| | - Myeong Geun Choi
- Department of Internal Medicine, Division of Pulmonology and Critical Care Medicine, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea;
| | - Eun Mi Chun
- Department of Internal Medicine, Division of Pulmonology and Critical Care Medicine, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea;
| |
Collapse
|
10
|
Oblitas CM, Demelo-Rodríguez P, Barrera-López L, Galeano-Valle F, Rubio-Rivas M, Luque Del Pino J, Giner Galvañ V, Paredes-Ruíz D, Fernández-Madera Martínez R, Gericó Aseguinolaza M, Gómez-Huelgas R, Fernández FA, Torres Peña JD, Martín González JI, Méndez-Bailón M, Monge Monge D, Freire Castro SJ, Pastor Valverde C, Rodilla-Sala E, Guzmán García M, Rivas-Carmenado M, Gallo CM, Perea Ribis MA, Casas-Rojo JM, Millán Núñez-Cortés J. Impact of SARS-CoV-2 infection therapies on the risk of venous thromboembolism and cardiovascular events from the SEMI-COVID-19 Registry. Sci Rep 2025; 15:7722. [PMID: 40044746 PMCID: PMC11882944 DOI: 10.1038/s41598-025-90278-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
This study aimed to assess the impact of SARS-CoV-2 therapies on the risk of venous thromboembolism (VTE) and other cardiovascular events. A retrospective, multicenter, observational study included hospitalized patients in Spain due to acute SARS-CoV-2 infection from March 2020 to March 2022. A total of 184,324 hospitalized COVID-19 patients were included, with a mean age of 67.5 (± 16) years of whom 58.4% were male. Among the comorbidities, arterial hypertension was the most common, affecting 52.5% (9618 patients), followed by dyslipidemia in 39.5% (7237 patients), diabetes mellitus in 23.7% (1748 patients), and atrial fibrillation in 10.6% (1948 patients). The overall mortality rate was 17.4% (3183 patients) and 9.9% (1819 patients) required admission to an intensive care unit. Cardiovascular events occurred in 4.08% (748 patients), with VTE occurring in 2.78% (510 patients), myocardial infarction in 0.75% (137 patients), and ischemic stroke in 0.55% (101 patients). Among therapies, beta-lactams were used in 66.7% (12,228 patients), systemic corticosteroids in 56.9% (10,424 patients), and tocilizumab in 11.6% (2128 patients). Multivariate analysis revealed an independent association between VTE and the use of tocilizumab (adjusted OR 2.07; p < 0.01), corticosteroids (adjusted OR 1.44; p = 0.02), and macrolides (adjusted OR 0.58; p < 0.01). None of the therapies were associated with the risk of myocardial infarction or ischemic stroke. In this large national cohort, tocilizumab and corticosteroids exhibited an independent association for the risk of VTE, but not for myocardial infarction or ischemic stroke.
Collapse
Affiliation(s)
- Crhistian-Mario Oblitas
- Internal Medicine Department, Hospital Clínico de Santiago, Santiago de Compostela, Spain.
- Sanitary Research Institute of Santiago, Santiago de Compostela, Spain.
- School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
- General University Hospital Gregorio Marañón, C/ Doctor Esquerdo, 46, 28007, Madrid, Spain.
| | - Pablo Demelo-Rodríguez
- School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Internal Medicine Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Lucía Barrera-López
- Internal Medicine Department, Hospital Clínico de Santiago, Santiago de Compostela, Spain
- Sanitary Research Institute of Santiago, Santiago de Compostela, Spain
| | - Francisco Galeano-Valle
- School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Internal Medicine Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Manuel Rubio-Rivas
- Internal Medicine Department, Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Vicente Giner Galvañ
- Internal Medicine Department, Hospital Universitario San Juan de Alicante, Alicante, Spain
| | - Diana Paredes-Ruíz
- Internal Medicine Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Ricardo Gómez-Huelgas
- Internal Medicine Department, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | | | | | | | - Daniel Monge Monge
- Internal Medicine Department, Hospital Complejo Asistencial de Segovia, Segovia, Spain
| | | | - Cruz Pastor Valverde
- Internal Medicine Department, Hospital Universitario Infanta Cristina, Parla, Spain
| | | | | | - María Rivas-Carmenado
- Internal Medicine Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | - José-Manuel Casas-Rojo
- School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Internal Medicine Department, Hospital Universitario Infanta Cristina, Parla, Spain
- Sanitary Research Institute Puerta de Hierro-Segovia de Arana, Madrid, Spain
| | - Jesús Millán Núñez-Cortés
- School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Internal Medicine Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| |
Collapse
|
11
|
Yılmaz M, Mirzaoğlu Ç. Retrospective Cohort Study: Severe COVID-19 Leads to Permanent Blunted Heart Rate Turbulence. Diagnostics (Basel) 2025; 15:621. [PMID: 40075869 PMCID: PMC11899457 DOI: 10.3390/diagnostics15050621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Heart rate turbulence (HRT) is a non-invasive technique that can be used to evaluate autonomic nervous system (ANS) function and cardiac arrhythmia. The objective of this study is to investigate whether COVID-19 can lead to long-term blunted HRT following recovery. Methods: This retrospective cohort study included 253 individuals with a confirmed history of COVID-19, referred to as the recovered COVID-19 group, along with 315 healthy participants who had no history of the virus. The recovered COVID-19 group was categorized into three subgroups based on their chest CT severity scores. The HRT analyses were obtained from a 24-h electrocardiography-Holter recording. Results: This study revealed that the HRT onset value was elevated in the recovered COVID-19 group, while the HRT slope value showed a significant decrease when compared to the control group. Correlation analyses indicated a positive relationship between the chest CT severity score and HRT onset, whereas a negative correlation was observed between the chest CT severity score and HRT slope. Regression analyses identified recovery from severe COVID-19, chest CT severity score, hypertension (HT), and smoking as independent predictors of both abnormal HRT onset and the existence of an abnormal HRT slope. Conclusions: Individuals who have recovered from severe COVID-19 are expected to encounter a permanent blunting of HRT, which is regarded as a significant indicator of an increased risk of ventricular arrhythmias and impaired autonomic nervous system (ANS) function. Recovered severe COVID-19 individuals should be carefully evaluated for HRT with 24-h ECG-Holter.
Collapse
Affiliation(s)
- Mücahid Yılmaz
- Department of Cardiology, Elazığ Fethi Sekin City Hospital, University of Health Sciences, 23280 Elazığ, Turkey;
| | | |
Collapse
|
12
|
Kalibatas V, Kaseliene S, Kalediene R, Mesceriakova O, Sauliune S. Perceptions of healthcare access among Lithuanians aged 65 and over during the COVID-19 pandemic. Front Public Health 2025; 13:1504049. [PMID: 40104119 PMCID: PMC11913820 DOI: 10.3389/fpubh.2025.1504049] [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: 09/30/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Aim This study investigates the perceived accessibility of healthcare services among older adults in Lithuania during the COVID-19 pandemic. The study is significant as it sheds light on geographical, organizational, and financial healthcare access issues encountered by the older population. Methods Conducted in January 2024, the study involved an anonymous questionnaire survey of 1,503 Lithuanian residents aged 65 and older. Results The most frequently utilized healthcare services were consultations with a general practitioner (75.4%) 22.0% of respondents reported not receiving any healthcare services. 53.5% respondents were satisfied with travel time to specialists. Common challenges included difficulties in getting appointments with specialists (53.9%) and dentists (36.2%). Financial barriers led to unmet healthcare needs: 12.6% of the respondents did not receive needed services, 12.8% did not undergo recommended tests, and 14.2% did not purchase prescribed medications. Healthcare services were less accessible to elders with lower education, lower incomes, and those who self-rated health poorly (p < 0.05). Conclusion Most respondents received the healthcare they needed during the pandemic and rated geographical access positively. However, some problems in organizational and financial access were disclosed. The observed social gradient indicates that socioeconomic factors significantly influence healthcare access, potentially increasing vulnerability among certain groups.
Collapse
Affiliation(s)
- Vytenis Kalibatas
- Department of Health Management, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Snieguole Kaseliene
- Department of Health Management, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ramune Kalediene
- Department of Health Management, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Olga Mesceriakova
- Department of Health Management, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Skirmante Sauliune
- Department of Health Management, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
13
|
Zhao X, Zhang Y, Luo B. Ferroptosis, from the virus point of view: opportunities and challenges. Crit Rev Microbiol 2025; 51:246-263. [PMID: 38588443 DOI: 10.1080/1040841x.2024.2340643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 12/21/2023] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
Ferroptosis is a new type of cell death, which is mainly dependent on the formation and accumulation of reactive oxygen species and lipid peroxides mediated by iron. It is distinct from other forms of regulation of cell death in morphology, immunology, biochemistry, and molecular biology. Various cell death mechanisms have been observed in many viral infections, and virus-induced cell death has long been considered as a double-edged sword that can inhibit or aggravate viral infections. However, understanding of the role of ferroptosis in various viral infections is limited. Special attention will be paid to the mechanisms of ferroptosis in mediating viral infection and antiviral treatment associated with ferroptosis. In this paper, we outlined the mechanism of ferroptosis. Additionally, this paper also review research on ferroptosis from the perspective of the virus, discussed the research status of ferroptosis in virus infection and classified and summarized research on the interaction between viral infections and ferroptosis.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Bing Luo
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
14
|
Koceva H, Amiratashani M, Akbarimoghaddam P, Hoffmann B, Zhurgenbayeva G, Gresnigt MS, Marcelino VR, Eggeling C, Figge MT, Amorim MJ, Mosig AS. Deciphering respiratory viral infections by harnessing organ-on-chip technology to explore the gut-lung axis. Open Biol 2025; 15:240231. [PMID: 40037530 PMCID: PMC11879621 DOI: 10.1098/rsob.240231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
The lung microbiome has recently gained attention for potentially affecting respiratory viral infections, including influenza A virus, respiratory syncytial virus (RSV) and SARS-CoV-2. We will discuss the complexities of the lung microenvironment in the context of viral infections and the use of organ-on-chip (OoC) models in replicating the respiratory tract milieu to aid in understanding the role of temporary microbial colonization. Leveraging the innovative capabilities of OoC, particularly through integrating gut and lung models, opens new avenues to understand the mechanisms linking inter-organ crosstalk and respiratory infections. We will discuss technical aspects of OoC lung models, ranging from the selection of cell substrates for extracellular matrix mimicry, mechanical strain, breathing mechanisms and air-liquid interface to the integration of immune cells and use of microscopy tools for algorithm-based image analysis and systems biology to study viral infection in vitro. OoC offers exciting new options to study viral infections across host species and to investigate human cellular physiology at a personalized level. This review bridges the gap between complex biological phenomena and the technical prowess of OoC models, providing a comprehensive roadmap for researchers in the field.
Collapse
Affiliation(s)
- Hristina Koceva
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Mona Amiratashani
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Parastoo Akbarimoghaddam
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Bianca Hoffmann
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Gaukhar Zhurgenbayeva
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Leibniz Institute of Photonic Technologies e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Jena, Germany
| | - Mark S. Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Vanessa Rossetto Marcelino
- Melbourne Integrative Genomics, School of BioSciences, University of Melbourne, Parkville, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Australia
| | - Christian Eggeling
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Leibniz Institute of Photonic Technologies e.V., Member of the Leibniz Centre for Photonics in Infection Research (LPI), Jena, Germany
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
- Jena Center for Soft Matter, Jena, Germany
| | - Marc Thilo Figge
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Maria-João Amorim
- Católica Biomédical Research Centre, Católica Medical School, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Alexander S. Mosig
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
- Jena Center for Soft Matter, Jena, Germany
- Center of Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
15
|
Velásquez García HA, Wong S, Jeong D, Binka M, Naveed Z, Wilton J, Hawkins NM, Janjua NZ. Risk of Major Adverse Cardiovascular Events After SARS-CoV-2 Infection in British Columbia: A Population-Based Study. Am J Med 2025; 138:524-531.e34. [PMID: 38670520 DOI: 10.1016/j.amjmed.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND COVID-19 is associated with increased risk of post-acute cardiovascular outcomes. Population-based evidence for long periods of observation is still limited. METHODS This population-based cohort study was conducted using data (2020-2021) from the British Columbia COVID-19 Cohort. The exposure of interest was severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, identified through reverse transcription-polymerase chain reaction (RT-PCR) assay. Individuals who tested positive (exposed) on RT-PCR were matched to negative controls (unexposed) on sex, age, and RT-PCR collection date in a 1:4 ratio. Outcomes of interest were incident major adverse cardiovascular events and acute myocardial infarction, identified more than 30 days after RT-PCR collection date. The association between SARS-CoV-2 infection and cardiovascular risk was assessed through multivariable survival models. Population attributable fractions were computed from Cox models. RESULTS We included 649,320 individuals: 129,864 exposed and 519,456 unexposed. The median duration of follow-up was 260 days; 1,786 events (0.34%) took place among the unexposed, and 702 (0.54%) in the exposed. The risk of major adverse cardiovascular events was higher in the exposed (adjusted hazard ratio [aHR] 1.34; 95% confidence interval [CI], 1.22-1.46), with greater risk observed in those who were hospitalized (aHR 3.81; 95% CI, 3.12-4.65) or required intensive care unit admission (aHR 6.25; 95% CI, 4.59-8.52) compared with the unexposed group. The fraction of cardiovascular events attributable to SARS-CoV-2 was 7.04% (95% CI, 4.67-9.41%). Comparable results were observed for acute myocardial infarction. CONCLUSIONS SARS-CoV-2 infection was associated with higher cardiovascular risk, with graded increase across the acute COVID-19 severity, contributing to 7% of incident major adverse cardiovascular events. These findings suggest that long-term monitoring of cardiovascular risk is required in COVID-19 survivors.
Collapse
Affiliation(s)
- Héctor Alexander Velásquez García
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Stanley Wong
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada
| | - Dahn Jeong
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Mawuena Binka
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Zaeema Naveed
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - James Wilton
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada
| | - Nathaniel Mark Hawkins
- Division of Cardiology, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada; Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Naveed Zafar Janjua
- Data and Analytic Services, British Columbia Centre for Disease Control, Vancouver, British Columbia V5Z 4R4, Canada; School of Population and Public Health, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada; Centre for Advancing Health Outcomes, St. Paul's Hospital, Vancouver, British Columbia V6Z 1Y6, Canada.
| |
Collapse
|
16
|
Roubinian NH, Greene J, Spencer BR, Bravo M, Bruhn R, Saa P, Stone M, Custer B, Kleinman S, Liu VX, Norris PJ, Busch MP. Blood donor SARS-CoV-2 infection or vaccination and adverse outcomes in plasma and platelet transfusion recipients. Transfusion 2025; 65:485-495. [PMID: 40012124 DOI: 10.1111/trf.18159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/08/2025] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Despite data supporting the safety of SARS-CoV-2 vaccination, concerns regarding the receipt of blood products from donors previously infected or vaccinated against SARS-CoV-2 persist. We assessed whether transfusions of plasma or platelet products from donors with prior SARS-CoV-2 infection or vaccination were associated with adverse outcomes in patients without COVID-19. METHODS We linked donor SARS-CoV-2 spike and nucleocapsid antibody data and vaccination history to blood products transfused between June 1, 2020 and March 31, 2022. We used logistic regression, adjusting for demographics and comorbidities, to calculate odds ratios and 95% confidence intervals (CI) for posttransfusion thrombosis, increased respiratory requirement, and hospital mortality. Outcomes were assessed as per transfused unit from previously infected or vaccinated donors compared to units from uninfected or unvaccinated donors. RESULTS Among 8715 hospitalizations of 7773 transfusion recipients linked to donor SARS-CoV-2 antibody data, there were 251 thromboses, 700 hospitalizations with increased respiratory requirements, and 1443 deaths. Among 15,167 transfused plasma units, 4993 and 1106 were from vaccinated donors and previously infected donors, respectively. Among 19,295 transfused platelet units, 8530 and 1368 were from vaccinated and previously infected donors, respectively. There were no associations between the transfusion of blood products from vaccinated or previously infected donors and thrombosis, increased respiratory requirements, or hospital mortality (all CI including 1). Nor were there associations between the receipt of blood products from recently infected or vaccinated donors or high SARS-CoV-2 antibody titers and adverse outcomes. DISCUSSION Donor SARS-Cov-2 infection and vaccination were not associated with adverse patient outcomes and do not need to be considered in blood allocation.
Collapse
Affiliation(s)
- Nareg H Roubinian
- Kaiser Permanente Northern California Division of Research, Pleasanton, California, USA
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - John Greene
- Kaiser Permanente Northern California Division of Research, Pleasanton, California, USA
| | - Bryan R Spencer
- American Red Cross, Scientific Affairs, Dedham, Massachusetts, USA
| | | | - Roberta Bruhn
- Vitalant Research Institute, San Francisco, California, USA
| | - Paula Saa
- American Red Cross, Scientific Affairs, Derwood, Maryland, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Brian Custer
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Steve Kleinman
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Vincent X Liu
- Kaiser Permanente Northern California Division of Research, Pleasanton, California, USA
| | - Philip J Norris
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Michael P Busch
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
17
|
Gambini F, Arbon D, Nickl P, Zatecka V, Fedosieieva O, Labaj J, Novosadova V, Trylcova J, Weber J, Prochazka J, Balounova J, Sedlacek R. New mouse model for inducible hACE2 expression enables to dissect SARS-CoV-2 pathology beyond the respiratory system. Mamm Genome 2025:10.1007/s00335-025-10115-1. [PMID: 39985688 DOI: 10.1007/s00335-025-10115-1] [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: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection is not limited to the respiratory tract as receptors, including the angiotensin-converting enzyme 2 (ACE2), are expressed across many tissues. This study employed a new conditional mouse model, Rosa26creERT2/chACE2, which expresses human ACE2 (hACE2) across multiple organs, to investigate the effects of SARS-CoV-2 infection beyond the respiratory system. This strain demonstrated susceptibility to SARS-CoV-2 infection in a dose and sex-dependent manner, showing that infected male mice exhibited more severe disease outcomes, including significant weight loss, pronounced lung pathology and dysfunction, and increased mortality, compared to females. In contrast to intratracheal infection, intranasal virus administration facilitated viral spread to the brain, thereby underscoring the nasal route's role in the pathogenesis of neurological manifestations. Intranasal infection also led to increased innate immune system activation as compared to intratracheal virus administration, even though both routes activated the adaptive immune response. This model provides a valuable tool to study SARS-CoV-2 in individual tissues or use a multisystemic approach, and it also advances possibilities for preclinical evaluation of antiviral therapies and vaccine strategies.
Collapse
Affiliation(s)
- Federica Gambini
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, 142 20, Czech Republic
| | - Dominik Arbon
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Petr Nickl
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Vaclav Zatecka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Olha Fedosieieva
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Juraj Labaj
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Vendula Novosadova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Jana Trylcova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 166 10, Czech Republic
| | - Jan Weber
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 166 10, Czech Republic
| | - Jan Prochazka
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, 142 20, Czech Republic
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Jana Balounova
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic.
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, Prague, 142 20, Czech Republic.
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prumyslova 595, Vestec, 252 50, Czech Republic.
| |
Collapse
|
18
|
Dong K, Tang J, Xu C, Gui W, Tian J, Chun B, Li D, Wang L. The effects of blood flow restriction combined with endurance training on athletes' aerobic capacity, lower limb muscle strength, anaerobic power and sports performance: a meta-analysis. BMC Sports Sci Med Rehabil 2025; 17:24. [PMID: 39987129 PMCID: PMC11847382 DOI: 10.1186/s13102-025-01072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/05/2025] [Indexed: 02/24/2025]
Abstract
OBJECTIVE To evaluate the effects of blood flow restriction (BFR) combined with endurance training on aerobic capacity, lower limb muscle strength, anaerobic power, and sports performance to supply effective scientific guidance for training. Two reviewers independently screened the literature, extracted data, and assessed the risk of bias of the included studies. We searched PubMed, Medline, Cochrane, SPORTDiscus and Web of Science databases up to 28 October 2024. Two reviewers independently screened the literature, extracted data, and assessed the risk of bias of the included studies. We calculated the effect size using standardized mean difference values and the random effects model. The results showed a medium effect size on maximal oxygen uptake (V̇O2max), a large effect size on lower limb muscle strength, a small effect size on anaerobic power and sports performance. In conclusion, while BFR training during endurance training had a significant positive effect on lower limb muscle strength and moderate improvement in V̇O2max, its impact on anaerobic power and sports performance was relatively small. These findings suggest that BFR training may be effective for enhancing muscle strength and aerobic capacity, but its benefits on anaerobic power and sport-specific performance may be limited. Therefore, it is important to carefully design BFR training programs to target specific outcomes.
Collapse
Affiliation(s)
- Kuan Dong
- School of Physical Education, Central China Normal University, Wuhan, China
| | - Jing Tang
- School of Electrical and Electronic EngineeringHuBei University of Technology, Wuhan, China
| | - Chengli Xu
- School of Physical Education, Central China Normal University, Wuhan, China
| | - Wenliang Gui
- School of Physical Education, Central China Normal University, Wuhan, China
| | - Jing Tian
- School of Physical Education, Central China Normal University, Wuhan, China.
| | - Buongo Chun
- Graduate School of Physical Education, Myongji University, Yongin, Republic of Korea
| | - Dong Li
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
- School of Physical Education and Health, Zhaoqing University, Zhaoqing, China
| | - Liqing Wang
- School of Physical Education, Central China Normal University, Wuhan, China
| |
Collapse
|
19
|
Kohler AK, Richter S, Schmid M, Zimmermann H, Winterer H, Schneider S, Kollum M. Three-Year Follow-Up of COVID-19 Cases in District of Constance, Germany. A Prospective, Controlled Cohort Study (FSC19-KN). J Clin Med 2025; 14:1439. [PMID: 40094912 PMCID: PMC11900586 DOI: 10.3390/jcm14051439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Background and Objectives: Long-term sequalae of viral diseases, especially after infections with SARS-CoV-2 (COVID-19), can induce multi-organ involvement, as around 65 million people worldwide report persistent symptoms that go far beyond the acute course. Studies indicate that early virus variants pose a higher risk of developing post-COVID-19 conditions. The primary aim of this study was to investigate the possible long-term effects based on the hospitalization rates and associated clinical events in patients infected with SARS-CoV-2 over an observational period of three years after the initial infection. Secondarily, an investigation of health-related quality of life and functional status was performed. Methods and Materials: The study presented was designed as a prospective, controlled cohort study to follow up on COVID-19 cases in the district of Konstanz, Germany (FSC19-KN). The positive group included subjects who had a primary infection with SARS-CoV-2 between March and December 2020. The control group included subjects who did not have a SARS-CoV-2 infection, as evidenced by a negative antibody test. As the primary endpoint, hospitalization rates and respective related admission diagnosis during the observational period of three years from January 2021 until July 2024 were analyzed. The health-related quality of life and functional outcomes were measured by the SF-36 questionnaire and Post-COVID-19 Functional Status (PCFS) as the secondary endpoint. Results: During the three years of observation after inclusion in the study, the hospitalization rate did not differ significantly between the two groups of initially infected and non-infected subjects (cumulative events, verum group 57 to control group 45, OR 1.24, CI 0.83; 1.85, p = 0.30). However, the health-related quality of life, measured by SF-36 sub scores of the SARS-CoV-2-positive subjects, achieved significantly lower results, except for the dimension 'energy and fatigue', in which subjects of the verum group still achieved significantly lower scores. Conclusions: Mild COVID-19 cases have no significant impact on hospitalization rates during an observational period of three years after initial infection. Yet, SARS-CoV-2-positive subjects reported a reduced health-related quality of life and functional outcomes. Ultimately, only the sub score quality 'energy and fatigue' still registered significant differences between both cohorts at the end of the three-year observational period.
Collapse
Affiliation(s)
- Ann-Kathrin Kohler
- Hegau Bodensee Klinikum Singen, Gesundheitsverbund Landkreis Konstanz, Virchow Str. 10, 78224 Singen, Germany; (A.-K.K.); (S.R.); (M.S.); (H.Z.)
| | - Stephan Richter
- Hegau Bodensee Klinikum Singen, Gesundheitsverbund Landkreis Konstanz, Virchow Str. 10, 78224 Singen, Germany; (A.-K.K.); (S.R.); (M.S.); (H.Z.)
| | - Michael Schmid
- Hegau Bodensee Klinikum Singen, Gesundheitsverbund Landkreis Konstanz, Virchow Str. 10, 78224 Singen, Germany; (A.-K.K.); (S.R.); (M.S.); (H.Z.)
| | - Heidi Zimmermann
- Hegau Bodensee Klinikum Singen, Gesundheitsverbund Landkreis Konstanz, Virchow Str. 10, 78224 Singen, Germany; (A.-K.K.); (S.R.); (M.S.); (H.Z.)
| | - Hannes Winterer
- Landratsamt Konstanz, Amt für Gesundheit und Versorgung—Gesundheitsamt, Schellestraße 15, 78315 Radolfzell, Germany;
| | - Steffen Schneider
- Institut für Herzinfarktforschung, Bremserstr. 79, 67063 Ludwigshafen, Germany;
| | - Marc Kollum
- Hegau Bodensee Klinikum Singen, Gesundheitsverbund Landkreis Konstanz, Virchow Str. 10, 78224 Singen, Germany; (A.-K.K.); (S.R.); (M.S.); (H.Z.)
| |
Collapse
|
20
|
Dinoi G, Togo MV, Guida P, Deruvo C, Samarelli F, Imbrici P, Nicolotti O, De Luca A, Mastroianni F, Liantonio A, Altomare CD. Retrospective Clinical Investigation into the Association Between Abnormal Blood Clotting, Oral Anticoagulant Therapy, and Medium-Term Mortality in a Cohort of COVID-19 Patients. Biomedicines 2025; 13:535. [PMID: 40149514 PMCID: PMC11940371 DOI: 10.3390/biomedicines13030535] [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: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: People affected by COVID-19 are exposed to abnormal clotting and endothelial dysfunction, which may trigger thromboembolic events. This study aimed at retrospectively investigating whether oral anticoagulant therapy (OAT), encompassing either direct oral anticoagulants (DOACs), mainly apixaban, or the vitamin K antagonist (VKA) warfarin, could have impacted medium-term mortality in a cohort of SARS-CoV-2 patients. Methods: Among 1238 COVID-19 patients, hospitalized from 17 March 2020 to 15 June 2021, 247 survivors and 247 deceased within 90 days from hospitalization were matched 1:1 based on age, sex, and intensive care unit (ICU) admission within three days. Conditional logistic regression was used to estimate associations by means of odds ratio (OR) with a 95% confidence interval (CI). Results: A univariate regression analysis suggested that OAT, no differently from subcutaneous low-molecular-weight heparins (LMWHs) during hospitalization, has no significant impact (p value > 0.05) on medium-term mortality. A multivariate analysis, limited to baseline variables (i.e., comorbidities and pharmacotherapies at hospital admission) showing significant association (p < 0.05) to mortality in a univariate analysis, revealed that, compared to patients living at 90 days from hospitalization, deceased patients had cancer histories (OR 1.75, CI 1.06-2.90, p = 0.029) or suffered from asthma (OR 2.25, CI 1.13-4.47, p = 0.021). In contrast, heart failure (HF), atrial fibrillation (AF), arteriopathy, chronic obstructive pulmonary disease (COPD), and kidney failure (KF), which, in a univariate analysis, were found to be associated with the endpoint (p < 0.05), lost significance in a multivariate analysis. Therapy at admission with aldosterone antagonists also appeared to be associated with medium-term mortality (OR 2.49, CI 1.52-4.08, p < 0.001); whereas, vitamin D supplementation during hospitalization appeared to be beneficial. Although not conclusive, a search into the Eudravigilance database, combined with consulting a digital predictive platform (PLATO, polypharmacology platform prediction), suggested potential off-target activities, which might contribute to increasing the severity of SARS-CoV-2 infection. Conclusions: This retrospective clinical study furnished evidences of the impact of OAT, comorbidities and other pharmacological treatments on COVID-19 clinical course.
Collapse
Affiliation(s)
- Giorgia Dinoi
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Maria Vittoria Togo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Pietro Guida
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Caterina Deruvo
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Francesco Samarelli
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Paola Imbrici
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Orazio Nicolotti
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Franco Mastroianni
- Department of Internal Medicine, F. Miulli General Hospital, 70021 Bari, Italy; (P.G.); (F.M.)
| | - Antonella Liantonio
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| | - Cosimo Damiano Altomare
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (G.D.); (M.V.T.); (C.D.); (F.S.); (P.I.); (O.N.); (A.D.L.)
| |
Collapse
|
21
|
Kupriyanova Y, Yurchenko I, Bobrov P, Bartels F, Wierichs S, Jonuscheit M, Korzekwa B, Prystupa K, Schön M, Mendez D, Trenkamp S, Burkart V, Wagner R, Schrauwen-Hinderling V, Roden M. Alterations of hepatic lipid content following COVID-19 in persons with type 2 diabetes. BMJ Open Diabetes Res Care 2025; 13:e004727. [PMID: 39965871 PMCID: PMC11836859 DOI: 10.1136/bmjdrc-2024-004727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION The study aimed to assess the effect of COVID-19 on hepatic lipid (HL) content, fibrosis risk, and adiposity in persons with type 2 diabetes. RESEARCH DESIGN AND METHODS Participants with type 2 diabetes with a history of mild COVID-19 (n=15, age 58±12 years, body mass index 30.9±5.2 kg/m2) were examined before (baseline) and 1 year (12±2 months) after (follow-up) recovery from COVID-19. Investigations for changes in metabolic risk comprised clinical examination, fasting blood sampling and MR-based measurements. Potential changes were corrected with the time course of the respective parameters in a group of participants who did not contract COVID-19 over the same time course (n=14, 61±6 years, 30.0±4.6 kg/m2). RESULTS COVID-19 resulted in a relative increase in HL content of 56% (95% CI 18%, 106%; p=0.04) measured as proton density fat fraction (HL-PDFF), corrected for the time course in the absence of COVID-19. While no changes in hepatic stiffness and volume, intramyocellular lipids, whole-body, subcutaneous and visceral adipose tissue volumes as well as homeostatic model assessment of insulin resistance and beta-cell function were observed. CONCLUSIONS History of COVID-19 in persons with type 2 diabetes is associated with higher HL-PDFF after 1 year following recovery from infection. TRIAL REGISTRATION NUMBER NCT01055093.
Collapse
Affiliation(s)
- Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Frederik Bartels
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Stefan Wierichs
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Marc Jonuscheit
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Benedict Korzekwa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Dania Mendez
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Sandra Trenkamp
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Vera Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Borges FFDR, Braga ACB, Viana BS, Valente J, Bemfica JM, Sant’Anna T, Goulart CDL, Almeida-Val F, Arêas GPT. Functional Capacity Impairment in Long COVID After 17 Months of Severe Acute Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:276. [PMID: 40003501 PMCID: PMC11855803 DOI: 10.3390/ijerph22020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/03/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Long COVID represents a significant challenge in understanding the prolonged impact of the disease. Despite its increasing recognition, detailed insights into the long-term cardiopulmonary consequences remain sparse. This study aimed to evaluate the functional capacity of individuals with persistent symptoms after severe COVID-19 infection compared to control individuals without symptomatic COVID or mild COVID after 17 months. This is a case-control study assessing 34 individuals divided into two groups regarding functional capacity by distance in a 6-min walk test (D6MWT) associated with gas analysis, spirometry, respiratory muscle strength, and quality of life. During the 6 MWT, an important lower heart rate (HR) was observed for the COVID group (106 ± 10 bpm, difference mean: 21.3; p < 0.001), with greater exertional perception (Borg dyspnea: 4.5 [2.0-9.0], p < 0.001 and Borg fatigue: 4.0 [2.0-7.0], p = 0.01), a significant decrease in the distance covered (416 ± 94 m, difference mean: 107; p = 0.002), and a low value of O2 uptake (V˙O2) (11 ± 5.0 mL/(kg min), difference mean: 8.3; p = 0.005) and minute ventilation (22 ± 8 L/min, difference mean: 18.6; p = 0.002), in addition to very low quality of life scores. Regression analysis showed a significant association between D6MWT and Borg fatigue and Borg dyspnea at rest (p = 0.003; p = 0.009). V˙O2 and HR were also significantly associated with the outcomes of the D6MWT (p = 0.04 and p = 0.004, respectively). In conclusion, individuals who have severe COVID-19 and persist with symptoms have low functional capacity, low V˙O2, low HR behavior, and low quality of life.
Collapse
Affiliation(s)
| | | | | | - Jefferson Valente
- Tropical Medicine Graduation Program, Universidade Estadual do Amazonas, Manaus 3578, Brazil; (J.V.)
| | - João Marcos Bemfica
- Tropical Medicine Graduation Program, Universidade Estadual do Amazonas, Manaus 3578, Brazil; (J.V.)
| | - Thaís Sant’Anna
- Physical Therapy Department, Universidade Federal do Amazonas, Manaus 6200, Brazil; (A.C.B.B.); (T.S.)
| | - Cássia da Luz Goulart
- Physical Therapy Department, Universidade de Brasília, Campus Ceilandia, Brasília 72220-275, Brazil;
| | - Fernando Almeida-Val
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil;
| | | |
Collapse
|
23
|
Hatakeyama J, Nakamura K, Aso S, Kawauchi A, Fujitani S, Oshima T, Kato H, Ota K, Kamijo H, Asahi T, Muto Y, Hori M, Iba A, Hosozawa M, Iso H. Effects of Long COVID in Patients with Severe Coronavirus Disease 2019 on Long-Term Functional Impairments: A Post Hoc Analysis Focusing on Patients Admitted to the ICU in the COVID-19 Recovery Study II. Healthcare (Basel) 2025; 13:394. [PMID: 39997269 PMCID: PMC11855593 DOI: 10.3390/healthcare13040394] [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: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Background/Objectives: This study investigated the prevalence of functional impairments and the effects of long COVID on long-term functional impairments in patients with severe COVID-19. Methods: We conducted a nationwide multicenter cohort study in collaboration with nine hospitals, collecting data using self-administered questionnaires from participants aged 20 years or older who were diagnosed with COVID-19, admitted to the intensive care unit (ICU) between April 2021 and September 2021, and discharged alive. Questionnaires regarding daily life, sequela, and functional impairments were mailed to patients in August 2022. The effects of long COVID on functional impairments were examined using a multivariate logistic regression analysis. Results: The survey was completed by 220 patients, with a mean of 416 days after discharge. Among respondents, 20.5% had physical impairments (n = 45), 35.0% had mental disorders (n = 77), and 42.7% had either (n = 94). Furthermore, 77.7% had long COVID (171/220), and the most common symptom was dyspnea (40.0%). The multivariate analysis showed that fatigue/malaise, upper respiratory tract symptoms, myalgia, muscle weakness, decreased concentration, sleep disorder, brain fog, and dizziness were risk factors for functional impairments at one year. Conclusions: Many patients with severe COVID-19 admitted to the ICU still suffered from post-intensive care syndrome even after one year, which manifested in combination with direct symptoms of the original disease, such as long COVID.
Collapse
Affiliation(s)
- Junji Hatakeyama
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki 569-8686, Osaka, Japan;
| | - Kensuke Nakamura
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, 2-1-1 Jonan-cho, Hitachi 317-0077, Ibaraki, Japan
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Kanagawa, Japan
| | - Shotaro Aso
- Department of Health Services Research, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku 113-0033, Tokyo, Japan;
| | - Akira Kawauchi
- Department of Critical Care and Emergency Medicine, Japanese Red Cross Maebashi Hospital, 389-1 Asakura-machi, Maebashi 371-0811, Gunma, Japan;
| | - Shigeki Fujitani
- Department of Emergency Medicine and Critical Care Medicine, St. Marianna University, 2-16-1 Sugao, Miyamae-ku, Kawasaki 216-8511, Kanagawa, Japan;
| | - Taku Oshima
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8677, Chiba, Japan;
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Kanagawa, Japan;
| | - Kohei Ota
- Department of Emergency and Critical Care Medicine, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Hiroshima, Japan;
| | - Hiroshi Kamijo
- Department of Emergency and Critical Care Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Nagano, Japan;
| | - Tomohiro Asahi
- Department of Cardiology, Naha City Hospital, 2-31-1 Furujima, Naha 902-8511, Okinawa, Japan;
| | - Yoko Muto
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku 162-8655, Tokyo, Japan; (Y.M.); (M.H.); (A.I.); (M.H.); (H.I.)
| | - Miyuki Hori
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku 162-8655, Tokyo, Japan; (Y.M.); (M.H.); (A.I.); (M.H.); (H.I.)
| | - Arisa Iba
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku 162-8655, Tokyo, Japan; (Y.M.); (M.H.); (A.I.); (M.H.); (H.I.)
| | - Mariko Hosozawa
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku 162-8655, Tokyo, Japan; (Y.M.); (M.H.); (A.I.); (M.H.); (H.I.)
| | - Hiroyasu Iso
- Institute for Global Health Policy Research, Bureau of International Health Cooperation, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku 162-8655, Tokyo, Japan; (Y.M.); (M.H.); (A.I.); (M.H.); (H.I.)
| |
Collapse
|
24
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
25
|
Oelsner EC, Krishnaswamy A, Rustamov R, Balte PP, Ali T, Allen NB, Andrews HF, Anugu P, Arynchyn A, Bateman LA, Cai J, Chang H, Chen L, Elkind MSV, Floyd JS, Gabriel KP, Gharib SA, Gutierrez JD, Stukovsky KH, Howard VJ, Isasi CR, Jager L, Jin L, Judd SE, Kanaya AM, Kandula NR, Kelly MR, Khan SS, Kucharska-Newton A, Lee JS, Levitan EB, Lewis CE, Make BJ, Malloy K, Manly JJ, Mauger D, Min YI, Murabito JM, Murphy CG, Norwood AF, O’Connor GT, Ortega VE, Patel AA, Pirzada A, Regan EA, Ring KB, Rosamond WD, Schwartz DA, Shikany JM, Sotres-Alvarez D, Tarlton C, Tse J, Meneses EMU, Vankineni M, Wenzel SE, Woodruff PG, Xanthakis V, Yang JH, Zakai NA, Zhang Y, Post WS. Classifying COVID-19 hospitalizations in epidemiology cohort studies: The C4R study. PLoS One 2025; 20:e0316198. [PMID: 39928595 PMCID: PMC11809881 DOI: 10.1371/journal.pone.0316198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/08/2024] [Indexed: 02/12/2025] Open
Abstract
RATIONALE Robust COVID-19 outcomes classification is important for ongoing epidemiology research on acute and post-acute COVID-19 conditions. Protocolized medical record review is an established method to validate endpoints for clinical trials and cardiovascular epidemiology cohorts; however, a protocol to adjudicate hospitalizations for COVID-19 among epidemiology cohorts was lacking. OBJECTIVES We developed a protocol to ascertain and adjudicate hospitalized COVID-19 across a meta-cohort of 14 US prospective cohort studies. This report describes the first three years of protocol implementation (October 1, 2020-October 1, 2023) and evaluates its repeatability and performance compared to classification by administrative codes. METHODS The protocol was adapted from cohort approaches to clinical cardiovascular events ascertainment and adjudication. Potential COVID-19 hospitalizations and deaths were identified by self-/proxy-report and, in some cases, active surveillance. Medical records were requested from hospitals and adjudicated for COVID-19 outcomes by clinically trained personnel according to a standardized rubric. Inter-rater agreement was assessed. The sensitivity and specificity of discharge diagnosis codes was compared to adjudicated diagnoses. MEASUREMENTS AND MAIN RESULTS The study obtained medical records for 1,167 potential COVID-19 hospitalizations, which underwent protocolized adjudication. Adjudication confirmed COVID-19 infection was present for 1,030 (88%) events, of which COVID-19 was not the cause of hospitalization for 78 (8%). Of 952 hospitalizations determined by adjudicators to be caused by COVID-19, 319 (34%) participants were critically ill and 210 (22%) died. Pneumonia was confirmed in 822 (86%) and acute kidney injury in 350 (37%); other cardiovascular and thrombotic complications were rare (2-5%). Interrater reliability among adjudicators was high (kappa = 0.85-1.00) except for myocardial infarction (kappa = 0.60). Compared to adjudication, sensitivity of discharge diagnosis codes was higher for pneumonia (84%) and pulmonary embolism (81%) than for other complications (48-70%). CONCLUSIONS Protocolized adjudication confirmed four out of five COVID-19 hospitalizations in a US meta-cohort and confirmed cases of pneumonia, pulmonary embolism, and other conditions that were not indicated by discharge diagnosis codes. These results highlight the importance of validating health outcomes for use in research on COVID-19 and post-COVID-19 conditions, and some limitations of claims-based data.
Collapse
Affiliation(s)
- Elizabeth C. Oelsner
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Akshaya Krishnaswamy
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Rafail Rustamov
- Department of Medicine, Nassau University Medical Center, East Meadow, New York, United States of America
| | - Pallavi P. Balte
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Tauqeer Ali
- Center for American Indian Health Research, Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Norrina B. Allen
- Center for Epidemiology and Population Health, Department of Preventive Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Howard F. Andrews
- Data Coordinating Center and Biostatistics Department, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Pramod Anugu
- Department of Medicine University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Alexander Arynchyn
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lori A. Bateman
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Harry Chang
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Lucas Chen
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Mitchell S. V. Elkind
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, United States of America
| | - James S. Floyd
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington, United States of America
- Division of General Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kelley Pettee Gabriel
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Sina A. Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jose D. Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Karen Hinckley Stukovsky
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington, United States of America
| | - Virginia J. Howard
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Carmen R. Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Lauren Jager
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington, United States of America
| | - Ling Jin
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Suzanne E. Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Alka M. Kanaya
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Namratha R. Kandula
- Center for Epidemiology and Population Health, Department of Preventive Medicine, Northwestern University, Chicago, Illinois, United States of America
- Division of General Internal Medicine, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Maureen R. Kelly
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Sadiya S. Khan
- Division of Cardiology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Anna Kucharska-Newton
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Joyce S. Lee
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Emily B. Levitan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Cora E. Lewis
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Barry J. Make
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Kimberly Malloy
- Center for American Indian Health Research, Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jennifer J. Manly
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - David Mauger
- Division of Biostatistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, State College, Pennsylvania, United States of America
| | - Yuan-I Min
- Department of Medicine University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Joanne M. Murabito
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Charles G. Murphy
- Division of Pulmonary Allergy, and Critical Care Medicine, Department of Medicine, Columbia University Medical Center, New York, New York, United States of America
| | - Arnita F. Norwood
- Department of Medicine University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - George T. O’Connor
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Victor E. Ortega
- Division of Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Department of Medicine, Mayo Clinic Scottsdale, Scottsdale, Arizona, United States of America
| | - Ashmi A. Patel
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Amber Pirzada
- Institute for Minority Health Research, University of Illinois Chicago, Chicago, Illinois, United States of America
| | - Elizabeth A. Regan
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Kimberly B. Ring
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Wayne D. Rosamond
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David A. Schwartz
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - James M. Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Daniela Sotres-Alvarez
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Cheryl Tarlton
- Center for American Indian Health Research, Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Janis Tse
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Elman M. Urbina Meneses
- Division of Pulmonary and Critical Care, Graduate School of Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Maya Vankineni
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, School of Public Heath, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Vanessa Xanthakis
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, and Department of Biostatistics, School of Public Health, Boston University, Boston, Massachusetts, United States of America
| | - Ji Hyun Yang
- NHLBI’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts, United States of America
| | - Neil A. Zakai
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont, United States of America
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Ying Zhang
- Center for American Indian Health Research, Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
26
|
Brahimi N, Croitoru D, Saidoune F, Zabihi H, Gilliet M, Piguet V. From Viral Infection to Skin Affliction: Unveiling Mechanisms of Cutaneous Manifestations in COVID-19 and Post-COVID Conditions. J Invest Dermatol 2025; 145:257-265. [PMID: 39665720 DOI: 10.1016/j.jid.2024.03.047] [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: 04/23/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 12/13/2024]
Abstract
COVID-19 skin manifestations are multifaceted, ranging from urticaria, morbilliform or papulovesicular rash, livedoid purpuric lesions, and to pseudochilblains (also called COVID toes). Recent insights into the mechanism of these manifestations have highlighted that morbilliform, papulovesicular, and livedoid/purpuric rashes are related to virus-induced endothelial cell damage and linked to moderate-to-severe disease, whereas pseudochilblains are related to an exaggerated IFN-1 production by plasmacytoid dendritic cells in protected individuals. In this paper, we will review the clinical and physiopathological features of cutaneous COVID-19 manifestations in relation to the direct viral cytopathic effects and dysregulated IFN-1 responses. We will also review the emerging insights into post-COVID conditions (also termed long COVID) and how they may be implicated in the persistence of COVID-19-associated skin diseases.
Collapse
Affiliation(s)
- Nesrine Brahimi
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Women's College Hospital, Toronto, Canada
| | - David Croitoru
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Women's College Hospital, Toronto, Canada
| | - Fanny Saidoune
- Department of Dermatology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Haleh Zabihi
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Michel Gilliet
- Department of Dermatology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland.
| | - Vincent Piguet
- Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Dermatology, Women's College Hospital, Toronto, Canada.
| |
Collapse
|
27
|
Hounye AH, Pan X, Zhao Y, Cao C, Wang J, Venunye AM, Xiong L, Chai X, Hou M. Significance of supervision sampling in control of communicable respiratory disease simulated by a new model during different stages of the disease. Sci Rep 2025; 15:3787. [PMID: 39885197 PMCID: PMC11782622 DOI: 10.1038/s41598-025-86739-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) interventions in interrupting transmission have paid heavy losses politically and economically. The Chinese government has replaced scaling up testing with monitoring focus groups and randomly supervising sampling, encouraging scientific research on the COVID-19 transmission curve to be confirmed by constructing epidemiological models, which include statistical models, computer simulations, mathematical illustrations of the pathogen and its effects, and several other methodologies. Although predicting and forecasting the propagation of COVID-19 are valuable, they nevertheless present an enormous challenge. This paper emphasis on pandemic simulation models by introduced respiratory-specific transmission to extend and complement the classical Susceptible-Exposed-(Asymptomatic)-Infected-Recovered SE(A)IR model to assess the significance of the COVID-19 transmission control features to provide an explanation of the rationale for the government policy. A novel epidemiological model is developed using mean-field theory. Utilizing the SE(A)IR extended framework, which is a suitable method for describing the progression of epidemics over actual or genuine landscapes, we have developed a novel model named SEIAPUFR. This model effectively detects the connections between various stages of infection. Subsequently, we formulated eight ordinary differential equations that precisely depict the population's temporal development inside each segment. Furthermore, we calibrated the transmission and clearance rates by considering the impact of various control strategies on the epidemiological dynamics, which we used to project the future course of COVID-19. Based on these parameter values, our emphasis was on determining the criteria for stabilizing the disease-free equilibrium (DEF). We also developed model parameters that are appropriate for COVID-19 outbreaks, taking into account varied population sizes. Ultimately, we conducted simulations and predictions for other prominent cities in China, such as Wuhan, Shanghai, Guangzhou, and Shenzhen, that have recently been affected by the COVID-19 outbreak. By integrating different control measures, respiratory-specific modeling, and disease supervision sampling into an expanded SEI (A) R epidemic model, we found that supervision sampling can improve early warning of viral activity levels and superspreading events, and explained the significance of containments in controlling COVID-19 transmission and the rationality of policy by the influence of different containment measures on the transmission rate. These results indicate that the control measures during the pandemic interrupted the transmission chain mainly by inhibiting respiratory transmission, and the proportion of supervision sampling should be proportional to the transmission rate, especially only aimed at preventing a resurgence of SARS-CoV-2 transmission in low-prevalence areas. Furthermore, The incidence hazard of Males and Females was 1.39(1.23-1.58), and 1.43(1.26-1.63), respectively. Our investigation found that the ratio of peak sampling is directly related to the transmission rate, and both decrease when control measures are implemented. Consequently, the control measures during the pandemic interrupted the transmission chain mainly by inhibiting respiratory transmission. Reasonable and effective interventions during the early stage can flatten the transmission curve, which will slow the momentum of the outbreak to reduce medical pressure.
Collapse
Affiliation(s)
- Alphonse Houssou Hounye
- General Surgery Department of Second Xiangya Hospital, Central South University Changsha, 139 Renmin Road, Changsha, Hunan, 410011, China
| | - Xiaogao Pan
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, China
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Yuqi Zhao
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Cong Cao
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Jiaoju Wang
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| | - Abidi Mimi Venunye
- General Surgery Department of Second Xiangya Hospital, Central South University Changsha, 139 Renmin Road, Changsha, Hunan, 410011, China
| | - Li Xiong
- General Surgery Department of Second Xiangya Hospital, Central South University Changsha, 139 Renmin Road, Changsha, Hunan, 410011, China.
| | - Xiangping Chai
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, China.
- Emergency Medicine and Difficult Diseases Institute, Second Xiangya Hospital, Central South University, Changsha, 139 Renmin Road, Changsha, 410011, Hunan, China.
| | - Muzhou Hou
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China.
| |
Collapse
|
28
|
de Souza T, Rosa AS, Constantino-Teles P, Ferreira VNS, Archanjo BS, Soares CAG, Picciani PHS, Allão Cassaro RA, Miranda MD, Poneti G. Silver Nanoparticles-Functionalized Textile against SARS-CoV-2: Antiviral Activity of the Capping Oleylamine Molecule. ACS APPLIED MATERIALS & INTERFACES 2025; 17:5710-5718. [PMID: 39807796 PMCID: PMC11788990 DOI: 10.1021/acsami.4c15289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
COVID-19 disease, triggered by SARS-CoV-2 virus infection, has led to more than 7.0 million deaths worldwide, with a significant fraction of recovered infected people reporting postviral symptoms. Smart surfaces functionalized with nanoparticles are a powerful tool to inactivate the virus and prevent the further spreading of the disease. Literature reports usually focus on the role of nanomaterial composition and size dispersion in evaluating their efficacy against SARS-CoV-2. Here, the anti-SARS-CoV-2 activity of oleylamine (OAm) used as a capping agent of silver nanoparticles is quantified for the first time. Spherical hydrophobic nanoparticles with 8 ± 2 nm diameter were prepared and characterized by Fourier transform infrared, dynamic light scattering, and transmission electron microscopy techniques. Biological assays showed that microgram amounts of nanoparticles, deposited on nonwoven textile obtained from surgical masks, efficiently inactivated up to 99.6(2)% of the virus with just 2 min of exposure. The virucidal activity of the corresponding amount of free OAm has been determined as well, reaching up to 67(1)% of activity for an exposure time of 10 min. Inductively coupled plasma optical emission spectrometry results pointed out a low leaching out of the nanoparticles in contact with water or culture medium. All in all, these results propose the capping molecules as an important chemical variable to be taken into account in the design of fast, efficient, and long-lasting anti-SARS-CoV-2 coatings.
Collapse
Affiliation(s)
| | - Alice S. Rosa
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Pamella Constantino-Teles
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Vivian Neuza S. Ferreira
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
| | - Braulio S. Archanjo
- Materials
Metrology Division, National Institute of
Metrology, Quality, and Technology, Duque de Caxias, Rio de Janeiro 25250-020, Brazil
| | - Carlos A. G. Soares
- Departamento
de Genética, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-617, Brazil
| | - Paulo H. S. Picciani
- Instituto
de Macromoléculas Professora Eloisa Mano, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Rafael A. Allão Cassaro
- Instituto
de Química, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-909, Brazil
| | - Milene Dias Miranda
- Laboratory
of Morphology and Virus Morphogenesis, Oswaldo
Cruz Institute, Fiocruz,
Avenida Brasil, Rio de Janeiro 21041-250, Brazil
- Programa
de pós-graduação em Biologia Celular e Molecular,
Instituto Oswaldo Cruz, Fundação
Oswaldo Cruz, Rio de
Janeiro 21041-250, Brazil
| | - Giordano Poneti
- Instituto
de Química, Universidade Federal
do Rio de Janeiro, Rio de
Janeiro 21941-909, Brazil
- Dipartimento
di Scienze Ecologiche e Biologiche, Università
degli Studi della Tuscia, Largo dell’Università, Viterbo 01100, Italy
| |
Collapse
|
29
|
Alicia LB, María Ángeles OG, Desirée MG, Maximino R, Marilina GA. Utility of Protein Markers in COVID-19 Patients. Int J Mol Sci 2025; 26:653. [PMID: 39859366 PMCID: PMC11766239 DOI: 10.3390/ijms26020653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has been a challenge at the healthcare level not only in the early stages of the pandemic, but also in the subsequent appearance of long-term COVID-19. Several investigations have attempted to identify proteomic biomarkers in an attempt to improve clinical care, guide treatment and predict possible patient outcomes. Proteins such as C-reactive protein (CRP) or interleukin 6 (IL-6) are clear markers of severe disease, but many others have been proposed that could help in risk stratification and in the prediction of specific complications. This review aims to bring together the most relevant studies in this regard, providing information to identify the most notable biomarkers in relation to COVID-19 found to date.
Collapse
Affiliation(s)
- López-Biedma Alicia
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
| | - Onieva-García María Ángeles
- Preventive Medicine and Public Health Unit, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Preventive Medicine and Public Health Research Group, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Martín-García Desirée
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Redondo Maximino
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - García-Aranda Marilina
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
| |
Collapse
|
30
|
Li H, Lin H, Fan T, Huang L, Zhou L, Tian X, Zhao R, Zhang Y, Yang X, Wan L, Zhong H, Jiang N, Wei C, Chen W, Hou L. Exosomes derived from syncytia induced by SARS-2-S promote the proliferation and metastasis of hepatocellular carcinoma cells. Front Cell Infect Microbiol 2025; 14:1415356. [PMID: 39844837 PMCID: PMC11750861 DOI: 10.3389/fcimb.2024.1415356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/29/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) is characterized by fever, fatigue, dry cough, dyspnea, mild pneumonia and acute lung injury (ALI), which can lead to acute respiratory distress syndrome (ARDS), and SARS-CoV-2 can accelerate tumor progression. However, the molecular mechanism for the increased mortality in cancer patients infected with COVID-19 is unclear. Methods Colony formation and wound healing assays were performed on Huh-7 cells cocultured with syncytia. Exosomes were purified from the cell supernatant and verified by nanoparticle tracking analysis (NTA), Western blot (WB) analysis and scanning electron microscopy (SEM). Differentially expressed proteins in syncytia-derived exosomes (Syn-Exos) and their functions was analyzed by Proteomic sequencing. Syn-Exo-mediated promotion of hepatocellular carcinoma cells was measured by CCK-8 and Transwell migration assays. The mechanism by which Syn-Exos promote tumor growth was analyzed by Western blotting. A patient-derived xenotransplantation (PDX) mouse model was constructed to evaluate the pathological role of the SARS-CoV-2 spike protein (SARS-2-S). The number of syncytia in the tumor tissue sections was determined by immunofluorescence analysis. Results Syncytium formation promoted the proliferation and migration of hepatocellular carcinoma cells. Proteomic sequencing revealed that proteins that regulate cell proliferation and metastasis in Syn-Exos were significantly upregulated. Syn-Exos promote the proliferation and migration of hepatocellular carcinoma cells. Animal experiments showed that a pseudotyped lentivirus bearing SARS-2-S (SARS-2-Spp) promoted tumor development in PDX mice. More syncytia were found in tumor tissue from SARS-2-Spp mice than from VSV-Gpp mice. Conclusions Syn-Exos induced by SARS-2-S can promote the proliferation and metastasis of hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Huilong Li
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Haotian Lin
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Tinghui Fan
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Linfei Huang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Li Zhou
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaoyu Tian
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Ruzhou Zhao
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Yanhong Zhang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Luming Wan
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Nan Jiang
- Department of Pharmacy, Medical Supplies Center of People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Congwen Wei
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Wei Chen
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Genetic engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
31
|
D'Orsi G, Palladini M, Mazza MG, Rovere-Querini P, Scalabrini A, Benedetti F. A novel analysis of interoceptive underpinnings of anxious psychopathology in COVID-19 survivors. Behav Brain Res 2025; 476:115275. [PMID: 39332641 DOI: 10.1016/j.bbr.2024.115275] [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: 06/14/2024] [Revised: 08/16/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
INTRODUCTION SARS-CoV-2 affects brain, body, and their interchange. We investigated interoceptive mechanisms in COVID-19 survivors focusing on their potential link with psychopathology and inflammatory biomarkers. METHODS We assessed interoceptive accuracy (IAc) and time-perceiving (TA) skills of 57 COVID-19 survivors one month after hospital discharge through, respectively, a heartbeats perception task and a time duration task. Each participant was assessed about his interoceptive awareness (IAw) through Multidimensional Assessment of Interoceptive Awareness questionnaire (MAIA) and then, screened for post-traumatic (Impact of Events Scale - IES-R), anxious (State-Trait Anxiety Inventory - STAI-Y1) and depressive (Zung Self-Rating Depression Scale - ZSDS; Beck Depression Inventory - BDI-13) symptoms. Biomarkers of inflammation (platelet count, PC; mean platelet volume, MPV and systemic immune-inflammation index, SII) were obtained in a subsample of 40 survivors by a blood sampling conducted at admission and discharge time from the hospital. Correlational, GLM, GLMZ, and mediation analyses were performed. RESULTS IAc did not correlate with TA confirming the reliability of interoceptive measure. IAc positively predicts MAIA's Trusting subscale and negatively predicts anxious psychopathology which fully mediates the effect of IAc on Trusting.PC at hospital admission predicts anxiety at one month after recovery. Again, a higher decrease of SII during hospitalization predicts higher IAc skill and lower anxiety state at one month. The link between SII change and anxiety is fully mediated by IAc. CONCLUSIONS Our results unveil a potential key role of interoception and brain-body interchange in the exacerbation and maintenance of anxiety psychopathology in COVID-19 survivors.
Collapse
Affiliation(s)
- Greta D'Orsi
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Mariagrazia Palladini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| | - Mario Gennaro Mazza
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele, Milano, Italy; Unit of Innate Immunity and Tissue Remodeling, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Andrea Scalabrini
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; Department of Human and Social Sciences University of Bergamo, Bergamo, Italy.
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
32
|
Alberto RPJ, Benjamin GN, Elizabeth RMJ, Alberto CDL, Eliseo PDB. Understanding COVID-19-related Acute Renal Injury in Elderly Individuals: Preexisting Systemic Inflammation before COVID-19 (SIC). Endocr Metab Immune Disord Drug Targets 2025; 25:300-309. [PMID: 38919086 DOI: 10.2174/0118715303312433240611093855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024]
Abstract
In this study, we examined preexisting systemic inflammation before COVID-19 (SIC), as assessed through C-reactive protein (CRP) levels, to gain insights into the origins of acute kidney injury (AKI) in adults with comorbidities affected by COVID-19. Although aging is not categorized as a disease, it is characterized by chronic inflammation, and older individuals typically exhibit higher circulating levels of inflammatory molecules, particularly CRP, compared to younger individuals. Conversely, elevated CRP concentrations in older adults have been linked with the development of comorbidities. Simultaneously, these comorbidities contribute to the production of inflammatory molecules, including CRP. Consequently, older adults with comorbidities have higher CRP concentrations than their counterparts without comorbidities or those with fewer comorbidities. Given that CRP levels are correlated with the development and severity of AKI in non-COVID-19 patients, we hypothesized that individuals with greater SIC are more likely to develop AKI during SARS-CoV-2 infection than those with less SIC.
Collapse
Affiliation(s)
- Ruiz-Pacheco Juan Alberto
- Investigador por México-CONAHCYT, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, México
| | - Gomez-Navarro Benjamin
- Servicio de Nefrología y Trasplantes, Hospital Country 2000, Guadalajara, Jalisco, México
| | | | - Castillo-Díaz Luis Alberto
- Departamento de Medicina y Ciencias de la Salud, Facultad Interdiciplinaria de Ciencias Biológicas y de la Salud, Universidad de Sonora, Hermosillo, México
| | - Portilla-de Buen Eliseo
- División de Investigación Quirúrgica, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara, Jalisco, México
| |
Collapse
|
33
|
Veronesi G, De Matteis S, Silibello C, Giusti EM, Ageno W, Ferrario MM. Interactive Effects of Long-term Exposure to Air Pollutants on SARS-CoV-2 Infection and Severity: A Northern Italian Population-based Cohort Study. Epidemiology 2025; 36:11-19. [PMID: 39316827 PMCID: PMC11594552 DOI: 10.1097/ede.0000000000001792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND We examined interactions, to our knowledge not yet explored, between long-term exposures to particulate matter (PM 10 ) with nitrogen dioxide (NO 2 ) and ozone (O 3 ) on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity and severity. METHODS We followed 709,864 adult residents of Varese Province from 1 February 2020 until the first positive test, COVID-19 hospitalization, or death, up to 31 December 2020. We estimated residential annual means of PM 10 , NO 2 , and O 3 in 2019 from chemical transport and random-forest models. We estimated the interactive effects of pollutants with urbanicity on SARS-CoV-2 infectivity, hospitalization, and mortality endpoints using Cox regression models adjusted for socio-demographic factors and comorbidities, and additional cases due to interactions using Poisson models. RESULTS In total 41,065 individuals were infected, 5203 were hospitalized and 1543 died from COVID-19 during follow-up. Mean PM 10 was 1.6 times higher and NO 2 2.6 times higher than WHO limits, with wide gradients between urban and nonurban areas. PM 10 and NO 2 were positively associated with SARS-CoV-2 infectivity and mortality, and PM 10 with hospitalizations in urban areas. Interaction analyses estimated that the effect of PM 10 (per 3.5 µg/m 3 ) on infectivity was strongest in urban areas [hazard ratio (HR) = 1.12; 95% CI =1.09, 1.16], corresponding to 854 additional cases per 100,000 person-years, and in areas at high NO 2 co-exposure (HR = 1.15; 1.08, 1.22). At higher levels of PM 10 co-exposure, the protective association of O 3 reversed (HR =1.32, 1.17, 1.49), yielding 278 additional cases per µg/m 3 increase in O 3 . We estimated similar interactive effects for severity endpoints. CONCLUSIONS We estimate that interactive effects between pollutants exacerbated the burden of the SARS-CoV-2 pandemic in urban areas.
Collapse
Affiliation(s)
- Giovanni Veronesi
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine, University of Insubria, Varese, Italy
| | - Sara De Matteis
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Emanuele M. Giusti
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine, University of Insubria, Varese, Italy
| | - Walter Ageno
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine, University of Insubria, Varese, Italy
| | - Marco M. Ferrario
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine, University of Insubria, Varese, Italy
| |
Collapse
|
34
|
Xu QQ, Yu DD, Fan XD, Cui HR, Dai QQ, Zhong XY, Zhang XY, Zhao C, You LZ, Shang HC. Chinese Medicine for Treatment of COVID-19: A Review of Potential Pharmacological Components and Mechanisms. Chin J Integr Med 2025; 31:83-95. [PMID: 38958885 DOI: 10.1007/s11655-024-3909-z] [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] [Accepted: 08/02/2023] [Indexed: 07/04/2024]
Abstract
Coronavirus disease 2019 (COVID-19) is an acute infectious respiratory disease that has been prevalent since December 2019. Chinese medicine (CM) has demonstrated its unique advantages in the fight against COVID-19 in the areas of disease prevention, improvement of clinical symptoms, and control of disease progression. This review summarized the relevant material components of CM in the treatment of COVID-19 by searching the relevant literature and reports on CM in the treatment of COVID-19 and combining with the physiological and pathological characteristics of the novel coronavirus. On the basis of sorting out experimental methods in vivo and in vitro, the mechanism of herb action was further clarified in terms of inhibiting virus invasion and replication and improving related complications. The aim of the article is to explore the strengths and characteristics of CM in the treatment of COVID-19, and to provide a basis for the research and scientific, standardized treatment of COVID-19 with CM.
Collapse
Affiliation(s)
- Qian-Qian Xu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Dong-Dong Yu
- The Geriatrics Center, First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, 230031, China
| | - Xiao-Dan Fan
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - He-Rong Cui
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Qian-Qian Dai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xiao-Ying Zhong
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, 51006, China
| | - Xin-Yi Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Chen Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liang-Zhen You
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Hong-Cai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| |
Collapse
|
35
|
Sun J, Deng X, Huang J, He G, Huang S. Data mining of adverse drug event signals with Nirmatrelvir/Ritonavir from FAERS. PLoS One 2024; 19:e0316573. [PMID: 39739861 DOI: 10.1371/journal.pone.0316573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/12/2024] [Indexed: 01/02/2025] Open
Abstract
Nirmatrelvir/Ritonavir, acting as an effective agent against COVID-19, has achieved considerable results in clinical studies in terms of drug efficacy. However, there is little research about its medication safety. Based on the FDA adverse event reporting system (FAERS) database, this study aims to mine the adverse reaction signals of the latest major recommended drug Nirmatrelvir/Ritonavir for the antiviral treatment of COVID-19, so as to provide a basis for safe and rational drug use. The reporting odds ratio (ROR) was used to explore the adverse event report data of all COVID-19 emergency use authorization (EUA) products in the FAERS database with the deadline of third quarter of 2023. In the analysis, 135427 adverse drug event (ADE) reports were found, and 35250 ADEs were reported with Nirmatrelvir/Ritonavir as the primary suspected drug, which was involved in multiple system. There was a high signal intensity of dysgeusia (ROR = 72.98), diarrhea (ROR = 3.03) and headache (ROR = 1.25), which was compatible with the adverse reactions recorded in the manual for Nirmatrelvir/Ritonavir. In addition, it was suggested that Nirmatrelvir/Ritonavir might cause pale-colored stools (ROR = 45.53), chromaturia (ROR = 3.07), yellow skin (ROR = 3.62), tongue coating (ROR = 35.55) and other new adverse reactions (not included in the instructions manual for Nirmatrelvir/Ritonavir). The ADEs of Nirmatrelvir/Ritonavir that are not in the instructions and are highly relevant in the real world are supplemented, prompting clinical attention to the ADEs of the drug, and providing a theoretical basis for the safe and effective application of the drug.
Collapse
Affiliation(s)
- Ji Sun
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The First Hospital of Changsha, Changsha, PR China
| | - Xuanyu Deng
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The First Hospital of Changsha, Changsha, PR China
| | - Juanjuan Huang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The First Hospital of Changsha, Changsha, PR China
| | - Gefei He
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The First Hospital of Changsha, Changsha, PR China
| | - Shiqiong Huang
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The First Hospital of Changsha, Changsha, PR China
| |
Collapse
|
36
|
Saric L, Domazet Bugarin J, Dosenovic S. Vitamin D Supplementation in Critically Ill-Narrative Review. Nutrients 2024; 17:156. [PMID: 39796590 PMCID: PMC11723408 DOI: 10.3390/nu17010156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Studies have shown a high prevalence of vitamin D deficiency in critically ill patients, and these patients are at higher risk for pneumonia and have increased incidence of sepsis and mortality. In this study, we reviewed available literature from randomized controlled trials (RCTs) on vitamin D supplementation in critically ill patients and summarized the evidence in this narrative review. METHODS Randomized controlled trials that included vitamin D supplementation as an intervention were eligible for inclusion. No limits were set regarding vitamin D dosage or route of administration, as well as for primary and secondary outcomes. A search was conducted in MEDLINE via PubMed for eligible RCTs. References from systematic reviews (SRs) and meta-analyses (MAs) were screened, and Clinicaltrials.gov was searched for ongoing studies. RESULTS A total of 21 RCTs involving 3166 patients were analyzed. There was a large heterogeneity in terms of patients' characteristics and inclusion criteria. Only six studies included patients with vitamin D levels < 50 nmol/L. Regarding clinically important outcomes, most of the studies did not show differences between the intervention and control group in terms of mortality, intensive care unit (ICU) or hospital length of stay (LoS). CONCLUSIONS There is great variability in trial designs regarding the selection of patients, dosage, dosing intervals and routes of administration of vitamin D supplements. Better study designs are mandatory for future clinical research, with measuring and reporting basal vitamin D levels before randomization. Since variability in supplementation regimes limits the possibility of data synthesis, standardized protocols for vitamin D supplementation should be used in clinical trial settings.
Collapse
Affiliation(s)
- Lenko Saric
- Department of Anesthesiology, Reanimatology and Intensive Care, University Hospital Split, 21000 Split, Croatia; (J.D.B.); (S.D.)
- University Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Josipa Domazet Bugarin
- Department of Anesthesiology, Reanimatology and Intensive Care, University Hospital Split, 21000 Split, Croatia; (J.D.B.); (S.D.)
| | - Svjetlana Dosenovic
- Department of Anesthesiology, Reanimatology and Intensive Care, University Hospital Split, 21000 Split, Croatia; (J.D.B.); (S.D.)
| |
Collapse
|
37
|
Wang F, Han H, Wang C, Wang J, Peng Y, Chen Y, He Y, Deng Z, Li F, Rong Y, Wang D, Liu W, Chen H, Zhang Z. SARS-CoV-2 membrane protein induces neurodegeneration via affecting Golgi-mitochondria interaction. Transl Neurodegener 2024; 13:68. [PMID: 39726060 DOI: 10.1186/s40035-024-00458-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Neurological complications are a significant concern of Coronavirus Disease 2019 (COVID-19). However, the pathogenic mechanism of neurological symptoms associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is poorly understood. METHODS We used Drosophila as a model to systematically analyze SARS-CoV-2 genes encoding structural and accessory proteins and identified the membrane protein (M) that disrupted mitochondrial functions in vivo. The M protein was stereotaxically injected to further assess its effects in the brains of wild-type (WT) and 5 × FAD mice. Omics technologies, including RNA sequencing and interactome analysis, were performed to explore the mechanisms of the effects of M protein both in vitro and in vivo. RESULTS Systematic analysis of SARS-CoV-2 structural and accessory proteins in Drosophila identified that the M protein induces mitochondrial fragmentation and dysfunction, leading to reduced ATP production, ROS overproduction, and eventually cell death in the indirect flight muscles. In WT mice, M caused hippocampal atrophy, neural apoptosis, glial activation, and mitochondrial damage. These changes were further aggravated in 5 × FAD mice. M was localized to the Golgi apparatus and genetically interacted with four wheel drive (FWD, a Drosophila homolog of mammalian PI4KIIIβ) to regulate Golgi functions in flies. Fwd RNAi, but not PI4KIIIα RNAi, reversed the M-induced Golgi abnormality, mitochondrial fragmentation, and ATP reduction. Inhibition of PI4KIIIβ activity suppressed the M-induced neuronal cell death. Therefore, M induced mitochondrial fragmentation and apoptosis likely through disruption of Golgi-derived PI(4)P-containing vesicles. CONCLUSIONS M disturbs the distribution and function of Golgi, leading to mitochondrial abnormality and eventually neurodegeneration via a PI4KIIIβ-mediated mechanism. This study reveals a potential mechanism for COVID-19 neurological symptoms and opens a new avenue for development of therapeutic strategies targeting SARS-CoV-2 M or mitochondria.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Hailong Han
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Caifang Wang
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Jingfei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Yanni Peng
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Ye Chen
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yaohui He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Zhouyang Deng
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Fang Li
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yikang Rong
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Danling Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Wen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Zhuohua Zhang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China.
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
38
|
Guerrerio AL, Mateja A, MacCarrick G, Fintzi J, Brittain E, Frischmeyer-Guerrerio PA, Dietz HC. Cardiovascular complications in vascular connective tissue disorders after COVID-19 infection and vaccination. PLoS One 2024; 19:e0315499. [PMID: 39705273 DOI: 10.1371/journal.pone.0315499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/26/2024] [Indexed: 12/22/2024] Open
Abstract
BACKGROUND COVID-19 infection and vaccination have been reported to confer an elevated risk for cardiovascular events (CVE). We sought to determine whether individuals with an underlying vascular connective tissue disorder including Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), or vascular Ehlers Danlos syndrome (vEDS) are at increased risk for cardiac events after COVID-19 infection or vaccination. METHODS 325 respondents self-reported data through a cross-sectional, web-based survey available from 22 November 2021, through 15 March 2022 regarding COVID-19 illness and vaccinations, the occurrence of any CVE, and adverse events following vaccination. The data were analyzed using a Cox proportional hazards model with time varying indicators for COVID-19 illness/vaccination in the preceding 30 days. RESULTS COVID-19 illness was significantly associated with an increased rate of a new abnormal heart rhythm 30 days following infection. No other CVEs were reported in the 90 days after COVID-19 illness. We did not find evidence of an increased rate of any CVE in the 30 days following any COVID-19 vaccination dose. CONCLUSION In respondents with MFS, LDS, or vEDS, we uncovered no evidence of an increase in CVEs in the 30 days following COVID-19 illness, with the possible exception of dysrhythmia. In light of the absence of a substantial increase in self-reported CVEs in the 30 days following COVID-19 vaccination, these data are in keeping with the recommendation from the Marfan Foundation Professional Advisory Board that all eligible persons be vaccinated for COVID-19.
Collapse
Affiliation(s)
- Anthony L Guerrerio
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research. Frederick, Maryland, United States of America
| | - Gretchen MacCarrick
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Erica Brittain
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Pamela A Frischmeyer-Guerrerio
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
39
|
Gutowski M, Klimkiewicz J, Rustecki B, Michałowski A, Skalec T, Lubas A. Peripheral and Organ Perfusion's Role in Prognosis of Disease Severity and Mortality in Severe COVID-19 Patients: Prospective Cohort Study. J Clin Med 2024; 13:7520. [PMID: 39768443 PMCID: PMC11728247 DOI: 10.3390/jcm13247520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Severe COVID-19 is associated with a generalized inflammatory response leading to peripheral and organ perfusion disorders. Objectives: This study aimed to evaluate the usefulness of peripheral and organ perfusion assessments in the prediction of prognosis and mortality in patients with severe COVID-19. Patients and Methods: In the first 48 h of hospitalization, peripheral perfusion (saturation, Finger Infrared Thermography-FIT; Capillary Refill Time-CRT), and the color Doppler renal cortex perfusion (RCP) were estimated in a group of 102 severe COVID-19 patients. Results: In total, 40 patients experienced deterioration and required intensification of oxygen treatment, and 24 finally died. In comparison with a stable course of the disease, patients with deterioration had initially higher WBC, CRP, AST, LDH, and CRT, but a lower oxygenation ratio and RCP. Deceased patients were older, had higher CRP, LDH, and CRT, but lower hemoglobin, oxygenation ratio, and RCP compared to survivors. In the multivariable regression analysis from perfusion parameters, only RCP and CRT were independently linked with deterioration (OR 0.002, p < 0.001; OR 1.825, p = 0.003, respectively) and death (OR 0.001, p = 0.004; OR 1.910, p = 0.003, respectively). Conclusions: Initial assessment of peripheral and organ perfusion can be helpful in identifying hospitalized severe COVID-19 patients with a higher risk of deterioration and death. Capillary Refill Time and Renal Cortical Perfusion were prognostic markers of deterioration or death. On the other hand, Finger Infrared Thermography and saturation were not statistically significant in predicting patient outcome. An RCP cut-off value below 0.127 and 0.112 [cm/s] and a Capillary Refill Time longer than 3.25 and 4.25 [s] indicate the risk of deterioration or death, respectively.
Collapse
Affiliation(s)
- Mateusz Gutowski
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Jakub Klimkiewicz
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Bartosz Rustecki
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Andrzej Michałowski
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Tomasz Skalec
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Arkadiusz Lubas
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland
| |
Collapse
|
40
|
Patrascu R, Dumitru CS, Laza R, Besliu RS, Gug M, Zara F, Laitin SMD. The Role of Age and Comorbidity Interactions in COVID-19 Mortality: Insights from Cardiac and Pulmonary Conditions. J Clin Med 2024; 13:7510. [PMID: 39768431 PMCID: PMC11677844 DOI: 10.3390/jcm13247510] [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: 11/14/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Understanding the interactions between age and comorbidities is crucial for assessing COVID-19 mortality, particularly in patients with cardiac and pulmonary conditions. This study investigates the relationship between comorbidities and mortality outcomes in a cohort of hospitalized COVID-19 patients, emphasizing the interplay of age, cardiac, and pulmonary conditions. Methods: We analyzed a cohort of 3005 patients hospitalized with COVID-19 between 2020 and 2022. Key variables included age, comorbidities (diabetes, cardiac, pulmonary, and neoplasms), and clinical outcomes. Chi-square tests and logistic regression models were used to assess the association between comorbidities and mortality. Stratified analyses by age, diabetes, and pulmonary conditions were conducted to explore interaction effects. Additionally, interaction terms were included in multivariable logistic regression models to evaluate the combined impact of age, comorbidities, and mortality. Results: Cardiac conditions such as hypertension, ischemic cardiopathy, and myocardial infarction showed significant protective effects against mortality in younger patients and in those without pulmonary conditions (p < 0.001). However, these protective effects were diminished in older patients and those with pulmonary comorbidities. Age was found to be a significant modifier of the relationship between cardiac conditions and mortality, with a stronger protective effect observed in patients under the median age (p < 0.001). Pulmonary comorbidities significantly increased the risk of mortality, particularly when co-occurring with cardiac conditions (p < 0.001). Diabetes did not significantly modify the relationship between cardiac conditions and mortality. Conclusions: The findings highlight the complex interactions between age, cardiac conditions, and pulmonary conditions in predicting COVID-19 mortality. Younger patients with cardiac comorbidities show a protective effect against mortality, while pulmonary conditions increase mortality risk, especially in older patients. These insights suggest that individualized risk assessments incorporating age and comorbidities are essential for managing COVID-19 outcomes.
Collapse
Affiliation(s)
- Raul Patrascu
- Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Cristina Stefania Dumitru
- Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Ruxandra Laza
- Infectious Diseases University Clinic, Department XIII, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
- Clinical Hospital of Infectious Diseases and Pneumology “Dr. Victor Babes”, 300310 Timisoara, Romania;
| | - Razvan Sebastian Besliu
- Epidemiology Clinic, ‘Pius Brinzeu’ Emergency Clinical County Hospital Timisoara, Liviu Rebreanu Boulevard No. 156, 300723 Timisoara, Romania;
| | - Miruna Gug
- Discipline of Genetics, Department of Microscopic Morphology, Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Flavia Zara
- Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Pathology, Emergency City Hospital, 300254 Timisoara, Romania
| | - Sorina Maria Denisa Laitin
- Clinical Hospital of Infectious Diseases and Pneumology “Dr. Victor Babes”, 300310 Timisoara, Romania;
- Epidemiology University Clinic, Department XIII, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| |
Collapse
|
41
|
Li Y, Lin Y, Yi Y, Zhu N, Cui X, Li X. COVID-19 Vaccination and Transient Increase in CD4/CD8 Cell Counts in People with HIV: Evidence from China. Vaccines (Basel) 2024; 12:1365. [PMID: 39772028 PMCID: PMC11680300 DOI: 10.3390/vaccines12121365] [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: 10/17/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: Accumulating evidence has confirmed the efficacy and safety of COVID-19 vaccines against SARS-CoV-2 infection. However, the effect of COVID-19 vaccination on immuno-virological parameters in people with HIV (PWH) is uncertain. Methods: A total of 372 PWH treated at Beijing Ditan Hospital were included. Unvaccinated PWH were matched 1:3 with vaccinated PWH using a propensity score matching algorithm. Differences in immuno-virological markers between the matched groups were analyzed. The Wilcoxon signed rank test was used to test for changes in CD4 and CD8 counts and HIV viral load over two months around vaccination. In addition, we investigated the long-term changes in HIV-related markers in different vaccination dose groups and in the entire vaccinated population. Results: Vaccinated PWH had a higher CD4/CD8 ratio (0.64 (0.49, 0.78) vs. 0.80 (0.56, 1.03), p = 0.037) than unvaccinated PWH within a two-month window after the third dose. There were 337 PWH who received COVID-19 vaccination, and 73.9% (n = 249) received three doses of vaccine. We observed a transient increase in CD4 count and CD4/CD8 ratio within a two-month window after vaccination, especially after the second dose (CD4 count: 583.5 (428.5, 706.8) vs. 618.0 (452.0, 744.0), p = 0.018; CD4/CD8 ratio: 0.70 (0.50, 0.91) vs. 0.71 (0.53, 0.96), p < 0.001)) and the third dose (CD4 count: 575.5 (435.5, 717.0) vs. 577.5 (440.8, 754.8), p = 0.001; CD4/CD8 ratio: 0.70 (0.52, 0.93) vs. 0.79 (0.53, 1.00), p < 0.001)). Recent CD4 counts and CD4/CD8 ratios were lower than after COVID-19 but remained higher than before COVID-19 in vaccinated PWH. In addition, COVID-19 vaccination had no negative effect on HIV viral load. Conclusions: A transient increase in CD4 count and CD4/CD8 ratio was observed after COVID-19 vaccination. However, the enhanced cellular immune response induced by vaccination may diminish over time and return to normal levels. There is no adverse effect of vaccination on HIV viral load.
Collapse
Affiliation(s)
- Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yingying Lin
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| | - Yunyun Yi
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing 100853, China;
| | - Na Zhu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xinyu Cui
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| |
Collapse
|
42
|
Blasco A, Royuela A, García-Gómez S, Gómez-Lozano N, Sánchez-Arjona A, de la Fuente J, Anel J, Sánchez-Galarraga I, Pérez-Redondo M, González E, Silva L. Association of SARS-CoV-2 immunoserology and vaccination status with myocardial infarction severity and outcome. Vaccine 2024; 42:126305. [PMID: 39244425 DOI: 10.1016/j.vaccine.2024.126305] [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: 05/04/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND The COVID-19 pandemic adversely affected the severity and prognosis of patients with acute myocardial infarction (MI) caused by atherothrombosis (type 1 MI). The effect, if any, of COVID-19 vaccination and natural SARS-CoV2 serologic immunity in these patients is unclear. Our aim was to analyze the association between the severity and outcome of patients with type 1 MI and their previous SARS-CoV2 vaccination and serostatus. METHODS A single-center retrospective cohort study conducted between March 1, 2020 and March 1, 2023. Clinical and follow-up information was collected from medical records and patients. Total antibodies (IgM, IgA, IgG) to nucleocapsid (N) antigens were measured by ECLIA (electrochemiluminescence-based immunoassay) to test the immune response to natural infection. If positive, IgM and IgG antibodies to spike (S) surface antigens were measured by CLIA to test the immune response to vaccine or natural infection. Multivariable logistic regression analysis was performed, adjusting for age, sex, hypertension, diabetes, and dyslipidemia. RESULTS Total sample of 949 patients, 656 with ST-segment elevation MI (STEMI) and 293 with non-ST-segment elevation MI (NSTEMI). Mean age was 64 (SD 13) years, 80 % men. Pre-admission vaccination status was: ≥ 1 dose, 53 % of patients; complete vaccination, 49 %; first booster dose, 25 %. The majority (84 %) of vaccines administered were mRNA-based. Six months after MI, 92 (9.7 %) patients had a major adverse cardiac event (MACE) and 50 died; 11 % of patients had severe heart failure or cardiogenic shock (Killip III-IV) after STEMI. Vaccinated patients with STEMI and positive serology (Pos/Vax group) had a higher risk of Killip III-IV on admission: OR 2.63 (1.27-5.44), p = 0.010. SARS-CoV-2 S-specific IgG titers were highest in this group (median > 2080 AU/mL, [IQR 1560- >2080] vs 91 [32-198] in the unvaccinated group). In the overall sample, a higher incidence of 6-month MACE was not demonstrated (OR 1.89 [0.98-3.61], p = 0.055). CONCLUSIONS The combination of vaccination and natural SARS-CoV2 infection was associated with the development of severe heart failure and cardiogenic shock in patients with STEMI, possibly related to an increased serological response.
Collapse
Affiliation(s)
- Ana Blasco
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain; Research Ethics Committee, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain.
| | - Ana Royuela
- Biostatistics Unit, Instituto de Investigación Puerta de Hierro-Segovia de Arana, Madrid, Spain; Center for Biomedical Research in Epidemiology and Public Health Network (CIBERESP), Madrid, Spain
| | - Sergio García-Gómez
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Natalia Gómez-Lozano
- Immunology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Alberto Sánchez-Arjona
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge de la Fuente
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Jorge Anel
- Microbiology Department, Serology Section, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Marina Pérez-Redondo
- Intensive Care Unit, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Elisa González
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Lorenzo Silva
- Cardiology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| |
Collapse
|
43
|
Khan MW, Ahmad M, Qudrat S, Afridi F, Khan NA, Afridi Z, Fahad, Azeem T, Ikram J. Vagal nerve stimulation for the management of long COVID symptoms. INFECTIOUS MEDICINE 2024; 3:100149. [PMID: 39678231 PMCID: PMC11638592 DOI: 10.1016/j.imj.2024.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
This review investigates the therapeutic potential of vagal nerve stimulation (VNS) in managing long COVID, a condition marked by persistent symptoms following acute SARS-CoV-2 infection. Long COVID manifests as ongoing fatigue, cognitive impairment, and autonomic dysfunction, hypothesized to arise from sustained inflammatory and neurological dysregulation. The vagus nerve, central to modulating systemic inflammation and autonomic homeostasis, represents a promising therapeutic target for symptom alleviation through VNS. A comprehensive literature search was conducted across PubMed, Scopus, and Web of Science to identify studies evaluating VNS in the context of long COVID. Preliminary evidence from small-scale pilot studies suggests VNS may attenuate systemic inflammation through activation of the cholinergic anti-inflammatory pathway (CAP), thus restoring autonomic balance and ameliorating symptoms such as fatigue, cognitive dysfunction, and anxiety. In targeting the inflammatory cascade that underlies both acute COVID-19 pathophysiology and its prolonged sequelae, VNS holds potential as an innovative intervention for persistent post-viral symptoms. While these initial findings indicate promise, current data remain limited in scope and robustness, underscoring the need for larger, controlled trials to validate the efficacy and mechanisms of VNS in long COVID management. Establishing a clearer understanding of VNS's impact on inflammation and autonomic regulation in this context is crucial to inform clinical guidelines and therapeutic strategies for long COVID, potentially offering a targeted approach for mitigating this disabling condition.
Collapse
Affiliation(s)
- Malik W.Z. Khan
- Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Muhammad Ahmad
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Salma Qudrat
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Fatma Afridi
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Najia Ali Khan
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Zain Afridi
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Fahad
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Touba Azeem
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Jibran Ikram
- Department of Cardiology, Cleveland Clinic Foundation, Ohio 44195, USA
| |
Collapse
|
44
|
Alswaina NF, Alsowinea A, Alhabeeb YK, Aljurbua A, Alghelfes A, Aljabaan B, Alshetwi HS. Ocular Symptoms Among COVID-19 Positive Patients in Saudi Arabia: A Cross-Sectional Survey. Cureus 2024; 16:e76671. [PMID: 39807460 PMCID: PMC11728207 DOI: 10.7759/cureus.76671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Background Coronavirus disease (COVID-19), a widespread viral illness, has been linked to a range of respiratory and other systemic symptoms. Along with the respiratory symptoms caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2), many extrapulmonary manifestations have also been reported. This study was conducted to report the ocular manifestations of COVID-19 in confirmed cases from the Qassim region, of Saudi Arabia. Methods In this retrospective survey-based study, an electronic survey was distributed via social media to individuals who reported a positive COVID-19 test. Demographic data, medical and ocular history, and data about their COVID-19 infection and ocular symptoms were collected. Results A total of 200 survey responses were included (35% male and 65% female, age 30.3 years). At least one ocular symptom was reported by 41 (20.5%) participants. Light sensitivity (8.5%), blurred vision (7.5%), redness (7.0%), and eye pain (7.0%) were the most common ocular symptoms. Conclusion Among the study participants, ocular symptoms were reported by more than one-fifth of the COVID-19 patients. These ocular symptoms were mostly mild. Further research is required to fully understand the association between COVID-19 and ocular manifestations.
Collapse
Affiliation(s)
- Nayef F Alswaina
- Department of Ophthalmology, College of Medicine, Qassim University, Buraydah, SAU
| | | | | | | | | | - Bedr Aljabaan
- College of Medicine, Qassim University, Buraydah, SAU
| | | |
Collapse
|
45
|
Bianconi V, Mannarino MR, Figorilli F, Ricciutelli F, De Carlo S, Zullo V, Corba M, Sahebkar A, Greco A, Lombardini R, Paltriccia R, Pirro M. Treatment with proton pump inhibitors is associated with secondary bacterial infections and sepsis in patients with COVID-19: a retrospective analysis of their joint impact on in-hospital prognosis. Ann Med 2024; 56:2399761. [PMID: 39475004 PMCID: PMC11616760 DOI: 10.1080/07853890.2024.2399761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/29/2024] [Accepted: 04/27/2024] [Indexed: 11/10/2024] Open
Abstract
Introduction and objectives. Secondary bacterial infections (SBIs) contribute to worse in-hospital outcomes in patients with Coronavirus disease 2019 (COVID-19). Treatment with proton pump inhibitors (PPIs) is associated with an increased risk of bacterial infections in different clinical settings. However, the association between PPI treatment prior to hospital admission and the occurrence of either SBIs or secondary bacterial sepsis (SBS) as well as their joint impact on clinical outcomes of patients hospitalized for COVID-19 are not clarified. Patients and methods. We retrospectively analyzed preadmission PPI use, in-hospital occurrence of SBIs and SBS, and in-hospital outcomes of a cohort of patients hospitalized for COVID-19. Results. Among 1087 patients, 447 (41%) were on PPI treatment prior to hospital admission. During the hospital stay, 197 (18%) and 223 (20%) patients were diagnosed with SBIs and SBS, respectively. The composite endpoint of intensive care unit (ICU) admission/in-hospital death was met by 214 (20%) patients. Preadmission PPI treatment was independently associated with up to a 2.1-fold and 1.7-fold increased risk of SBIs and SBS, respectively. The occurrence of SBS was independently associated with up to a 2.2-fold increased risk of ICU admission/in-hospital death. A significant preadmission PPI treatment x SBS interaction emerged in predicting ICU admission/in-hospital death (F = 5.221, pinteraction = 0.023). Conclusions. PPI treatment prior to hospital admission for COVID-19 is associated with an increased risk of SBIs and SBS. In addition, it interacts with SBS in predicting in-hospital prognosis. An appropriate use of PPIs may attenuate the risk of adverse clinical outcomes during hospitalization for COVID-19.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimo R. Mannarino
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filippo Figorilli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federica Ricciutelli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefania De Carlo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Zullo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Martina Corba
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alessia Greco
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Lombardini
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Paltriccia
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
46
|
Kamdar A, Sykes R, Thomson CR, Mangion K, Ang D, Lee MAW, Van Agtmael T, Berry C. Vascular fibrosis and extracellular matrix remodelling in post-COVID 19 conditions. INFECTIOUS MEDICINE 2024; 3:100147. [PMID: 39649442 PMCID: PMC11621938 DOI: 10.1016/j.imj.2024.100147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/02/2024] [Accepted: 09/23/2024] [Indexed: 12/10/2024]
Abstract
Causal associations between viral infections and acute myocardial injury are not fully understood, with mechanisms potentially involving direct cardiovascular involvement or systemic inflammation. This review explores plausible mechanisms of vascular fibrosis in patients with post-COVID-19 syndrome, focusing on extracellular matrix remodelling. Despite global attention, significant mechanistic or translational breakthroughs in the management of post-viral syndromes remain limited. No effective pharmacological or non-pharmacological interventions are currently available for patients experiencing persistent symptoms following COVID-19 infection. The substantial expansion of scientific knowledge resulting from collaborative efforts by medical experts, scientists, and government organisations in undertaking COVID-19 research could inform treatment strategies for other post-viral syndromes and respiratory illnesses. There is a critical need for clinical trials to evaluate potential therapeutic candidates, providing evidence to guide treatment decisions for post-COVID-19 syndromes.
Collapse
Affiliation(s)
- Anna Kamdar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Cameron R. Thomson
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| | - Daniel Ang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
| | - Michelle AW Lee
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Tom Van Agtmael
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow G81 4DY, UK
- Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow G51 4TF, UK
| |
Collapse
|
47
|
Aldiabat M, Aleyadeh W, Muzammil T, Adewuyi K, Alahmad M, Jabri A, Alhuneafat L, Kilani Y, Alsakarneh S, Bilal M. Rates, Risk Factors, and Outcomes of Nonvariceal Upper Gastrointestinal Bleeding in Patients Hospitalized for COVID-19 in the United States. Curr Med Sci 2024; 44:1202-1209. [PMID: 39673580 DOI: 10.1007/s11596-024-2838-6] [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: 06/15/2023] [Accepted: 12/11/2023] [Indexed: 12/16/2024]
Abstract
OBJECTIVE This study aimed to investigate the incidence and predictors of non-variceal upper gastrointestinal bleeding (NVUGIB) in hospitalized patients with coronavirus disease 2019 (COVID-19), as well as the inpatient outcomes associated with this complication. METHODS This was an analysis of the National Inpatient Sample Database from January to December 2020. Adult COVID-19 patients were categorized into two groups based on NVUGIB development during hospitalization. Multivariate logistic analysis was performed to identify predictors and outcomes associated with NVUGIB in hospitalized COVID-19 patients in the US, after adjusting for age, sex, race, and Charlson Comorbidity Index (CCI) score, using Stata/BE 17.0. RESULTS Among 1 050 045 hospitalized patients, 1.87% developed NVUGIB. Asian Americans had the highest risk, followed by Native Americans, Hispanics, and African Americans, with odds ratios (ORs) of 1.70, 1.59, 1.40, and 1.14, respectively. Patients with higher CCI scores were also at greater risk (with ORs of 1.47, 2.09, and 3.45 for CCI scores of 1, 2, and 3, respectively). COVID-19 patients with NVUGIB had a higher risk of inpatient mortality (OR=3.84), acute kidney injury (OR=3.12), hypovolemic shock (OR=13.7), blood transfusion (OR=7.02), and in-hospital cardiac arrest (OR=4.02). CONCLUSION NVUGIB occurred in 1.87% of hospitalized COVID-19 patients and was associated with a threefold increase in mortality. Further research is necessary to identify strategies for reducing its incidence in COVID-19 patients with multiple risk factors.
Collapse
Affiliation(s)
- Mohammad Aldiabat
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, 02115, USA.
| | - Wesam Aleyadeh
- Department of Medicine, Cleveland Clinic Akron General, Akron, 44307, USA
| | - Taimur Muzammil
- Department of Medicine, Allegheny Health Network, Pittsburgh, 15212, USA
| | - Kemi Adewuyi
- Department of Medicine, Allegheny Health Network, Pittsburgh, 15212, USA
| | - Majd Alahmad
- Department of Medicine, University of Pittsburgh, Pittsburgh, 15261, USA
| | - Ahmad Jabri
- Heart and Vascular Center, MetroHealth Medical Center, Cleveland, 44109, USA
| | - Laith Alhuneafat
- Department of Medicine, Allegheny Health Network, Pittsburgh, 15212, USA
| | - Yassine Kilani
- Department of Medicine, Lincoln Medical Center/Weil Cornell Medicine, Bronx, 10451, USA
| | - Saqr Alsakarneh
- Department of Medicine, University of Missouri-Kansas City, Kansas City, 64108, USA
| | - Mohammad Bilal
- Department of Medicine, University of Minnesota/Minneapolis VA Medical Center, Minneapolis, 55417, USA
| |
Collapse
|
48
|
Teng L, Chang G, Song X, Zhang M, Han Y, Chang W, Shen Z. Construction and validation of a risk model of proteinuria in patients with omicron COVID-19: retrospective cohort study. Ren Fail 2024; 46:2365979. [PMID: 39108141 PMCID: PMC11308959 DOI: 10.1080/0886022x.2024.2365979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND To explore the risk factors of proteinuria in Omicron variant patients and to construct and verify the risk predictive model. METHODS 1091 Omicron patients who were hospitalized from August 2022 to November 2022 at Tianjin First Central Hospital were defined as the derivation cohort. 306 Omicron patients who were hospitalized from January 2022 to March 2022 at the same hospital were defined as the validation cohort. The risk factors of proteinuria in derivation cohort were screened by univariate and multivariate logistic regression analysis, and proteinuria predicting scoring system was constructed and the receiver operating characteristic(ROC)curve was drawn to test the prediction ability. The proteinuria risk model was externally validated in validation cohort. RESULTS 7 factors including comorbidities, blood urea nitrogen (BUN), serum sodium (Na), uric acid (UA), C reactive protein (CRP) and vaccine dosages were included to construct a risk predictive model. The score ranged from -5 to 16. The area under the ROC curve(AUC) of the model was 0.8326(95% CI 0.7816 to 0.8835, p < 0.0001). Similarly to that observed in derivation cohort, the AUC is 0.833(95% CI 0.7808 to 0.9002, p < 0.0001), which verified good prediction ability and diagnostic accuracy in validation cohort. CONCLUSIONS The risk model of proteinuria after Omicron infection had better assessing efficiency which could provide reference for clinical prediction of the risk of proteinuria in Omicron patients.
Collapse
Affiliation(s)
- Lanbo Teng
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ge Chang
- Department of Clinical Medicine, Tianjin Medical University, Tianjin, China
| | - Xinyuan Song
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Miaomiao Zhang
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yingying Han
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenxiu Chang
- Department of Nephrology, Tianjin First Central Hospital, Nankai University, Tianjin, China
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zhongyang Shen
- National Health Commission (NHC) Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China
| |
Collapse
|
49
|
Mormile R, Mormile C, Picone C. Alzheimer's disease following COVID-19: a two player match? Infection 2024; 52:2547-2549. [PMID: 39143436 DOI: 10.1007/s15010-024-02368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
Common pathways may underlie the association between COVID-19 and risk for Alzheimer's disease (AD). We conjecture that severe COVID-19 may contribute to AD onset in predisposed individuals through aberrant MDSCs expression and increased IL-6 expression levels leading to immunosuppression in inflamed brains. Research studies are needed to gain empirical evidence to strengthen the hypothesis of the involvement of MDSCs and IL-6 in the formation of AD following COVID-19 infection and possibly vaccination enabling a more in-depth understanding of the role of immunosuppression in the onset of neurodegenerative diseases at any age. Identifying why those who get severe COVID-19 are more likely to develop AD may offer a novel therapeutic approach to delay or prevent cognitive decline.
Collapse
Affiliation(s)
- Raffaella Mormile
- Division of Pediatrics and Neonatology, Moscati Hospital, Via A. Gramsci, Aversa, 81031, Italy.
| | | | - Carmine Picone
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli", Naples, Italy
- Department of Medicine and Health Science, Vincenzo Tiberio University of Molise, Campobasso, Italy
| |
Collapse
|
50
|
Muayad J, Laylani NA, Davila-Siliezar P, Li HK, Lee AG. COVID-19-Associated Outer Retinopathy and Bilateral Optic Disc Edema. J Neuroophthalmol 2024; 44:e602-e604. [PMID: 38117569 DOI: 10.1097/wno.0000000000002069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Affiliation(s)
- Jawad Muayad
- Texas A and M School of Medicine (JM), College Station, Texas; Department of Ophthalmology (NAL, PDS, HKL, AGL), Blanton Eye Institute, Houston Methodist Hospital, Houston, Texas; Community Retina Group (HKL), Houston, Texas; The Houston Methodist Research Institute (HKL, AGL), Houston Methodist Hospital, Houston, Texas; Departments of Ophthalmology (AGL), Neurology, and Neurosurgery, Weill Cornell Medicine, New York, New York; Department of Ophthalmology (AGL), University of Texas Medical Branch, Galveston, Texas; Department of Ophthalmology (AGL), University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Ophthalmology (AGL), Texas A and M School of Medicine, Texas; Department of Ophthalmology (AGL), the University of Iowa Hospitals and Clinics, Iowa City, Iowa; and Department of Ophthalmology (AGL), Baylor College of Medicine, Houston, Texas
| | | | | | | | | |
Collapse
|