1
|
Mamani-Huanca M, Martínez S, López-López Á, López-Gonzálvez Á, Albóniga OE, Gradillas A, Barbas C, González-Ruiz V. CE-MS-Based Clinical Metabolomics of Human Plasma. Methods Mol Biol 2025; 2855:389-423. [PMID: 39354320 DOI: 10.1007/978-1-0716-4116-3_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Capillary electrophoresis coupled to mass spectrometry (CE-MS) has emerged as a powerful analytical technique with significant implications for clinical research and diagnostics. The integration of information from CE and MS strengthens confidence in the identification of compounds present in clinical samples. The ability of CE to separate molecules based on their electrophoretic mobility coupled to MS enables the accurate identification and quantification of analytes, even in complex biological matrices such as human plasma.Here, we present a detailed protocol for an untargeted metabolomics study using CE-MS and its application in a study on human plasma from patients suffering Long COVID syndrome. The protocol ranges from sample preparation to biological interpretation, detailing a workflow enabling the analysis of cationic and anionic compounds, metabolite identification, and data processing.
Collapse
Affiliation(s)
- Maricruz Mamani-Huanca
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Sara Martínez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Ángeles López-López
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Oihane E Albóniga
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Ana Gradillas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Víctor González-Ruiz
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain.
| |
Collapse
|
2
|
Wang Q, Zhou Y, Jing F, Feng Y, Ma J, Xue P, Dong Z. Effects of acute-phase COVID-19-related indicators on pulmonary fibrosis and follow-up evaluation. Eur J Med Res 2024; 29:585. [PMID: 39696619 DOI: 10.1186/s40001-024-02197-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Post-COVID-19 pulmonary fibrosis is a significant long-term respiratory morbidity affecting patients' respiratory health. This exploratory study aims to investigate the incidence, clinical characteristics, and acute-phase risk factors for pulmonary fibrosis in COVID-19 patients. Additionally, it evaluates pulmonary function and chest CT outcomes to provide clinical evidence for the early identification of high-risk patients and the prevention of post-COVID-19 pulmonary fibrosis. METHODS We retrospectively analyzed 595 patients hospitalized for COVID-19 from January 2022 to July 2023. Patients were divided into fibrosis and nonfibrosis groups on the basis of imaging changes. Baseline data, including demographics, disease severity, laboratory indicators, and chest imaging characteristics, were collected. Univariate and multivariate logistic regression analyses were performed to identify independent risk factors for pulmonary fibrosis. Pulmonary function and chest CT follow-ups were conducted for the fibrosis group. The data were processed via SPSS 26.0, with P < 0.05 considered statistically significant. RESULTS The incidence of pulmonary fibrosis was 4.37%, with 2.08% in moderate cases and 8.22% in severe cases. Significant differences were found between the fibrosis and nonfibrosis groups in sex; disease severity; NLR; ALB and LDH levels; and percentages of lung reticular lesions, consolidations, and GGOs (P < 0.05). Multivariate analysis revealed LDH (OR = 1.004, 95% CI 1.000-1.007, P = 0.035), ALB (OR = 0.871, 95% CI 0.778-0.974, P = 0.015), lung reticular lesion volume (OR = 1.116, 95% CI 1.040-1.199, P = 0.002), and lung consolidation volume (OR = 1.131, 95% CI 1.012-1.264, P = 0.030) as independent risk factors. The follow-up results revealed significant improvements in pulmonary function, specifically in the FVC%, FEV1%, and DLCO%, but not in the FEV1/FVC. Quantitative chest CT analysis revealed significant differences in lung reticular lesions, consolidation, and GGO volumes but no significant difference in honeycomb volume. CONCLUSIONS The incidence of pulmonary fibrosis post-COVID-19 increases with disease severity. LDH, ALB, lung reticular lesions, and consolidation volume are independent risk factors for patients with fibrosis.
Collapse
Affiliation(s)
- Qiong Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Respiratory Infection, ZhenHai Hospital of Traditional Chinese Medicine, Ningbo, 315200, China
| | - Ying Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Fangxue Jing
- Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Yingying Feng
- Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - JiangPo Ma
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China
- CiXi Biomedical Research Institute, WenZhou Medical University, Zhejiang, China
| | - Peng Xue
- Hainan University School of Mechanical and Electrical Engineering, Hainan, 570228, China
| | - Zhaoxing Dong
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
3
|
Gasparello J, Papi C, Marzaro G, Macone A, Zurlo M, Finotti A, Agostinelli E, Gambari R. Aged Garlic Extract (AGE) and Its Constituent S-Allyl-Cysteine (SAC) Inhibit the Expression of Pro-Inflammatory Genes Induced in Bronchial Epithelial IB3-1 Cells by Exposure to the SARS-CoV-2 Spike Protein and the BNT162b2 Vaccine. Molecules 2024; 29:5938. [PMID: 39770027 PMCID: PMC11677098 DOI: 10.3390/molecules29245938] [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: 10/31/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 01/05/2025] Open
Abstract
Garlic (Allium sativum L.) is a species of the onion family (Alliaceae) widely used as a food and a folk medicine. The objective of this study was to determine the effects of AGE (aged garlic extract) on pro-inflammatory genes relevant to COVID-19. To this aim, we treated bronchial epithelial IB3-1 cells with SARS-CoV-2 spike protein (S-protein) or with the COVID-19 BNT162b2 vaccine in the absence or in the presence of AGE. The results obtained demonstrated that AGE is a potent inhibitor of the S-protein-induced expression of the IL-1β, IL-6 and IL-8 genes. Bio-Plex analysis demonstrated that AGE reduced release of IL-6 and IL-8, which were highly induced by S-protein. No inhibition of cells' growth, toxicity and pro-apoptotic effects were found in AGE-treated cells. The effects of one of the major AGE constituents (S-allyl cysteine, SAC) were studied on the same experimental model systems. SAC was able to inhibit the S-protein-induced expression of IL-1β, IL-6 and IL-8 genes and extracellular release of IL-6 and IL-8, confirming that S-allyl-cysteine is one of the constituents of AGE that is responsible for inhibiting S-protein-induced pro-inflammatory genes. Docking experiments suggest that a possible mechanism of action of SAC is an interference with the activity of Toll-like receptors (TLRs), particularly TLR4, thereby inhibiting NF-κB- and NF-κB-regulated genes, such as IL-1β, IL-6 and IL-8 genes. These results suggest that both AGE and SAC deserve further experimental efforts to verify their effects on pro-inflammatory genes in SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Giovanni Marzaro
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy;
| | - Alberto Macone
- Department of Biochemical Sciences ‘A. Rossi Fanelli’, Sapienza University of Rome, 00185 Rome, Italy;
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, Via del Forte Tiburtino 98, 00159 Rome, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, 44121 Ferrara, Italy; (J.G.); (C.P.); (M.Z.); (A.F.)
| |
Collapse
|
4
|
Reyes Z, Stovall MC, Punyamurthula S, Longo M, Maraganore D, Solch-Ottaiano RJ. The impact of gut microbiome and diet on post-acute sequelae of SARS-CoV-2 infection. J Neurol Sci 2024; 467:123295. [PMID: 39550783 DOI: 10.1016/j.jns.2024.123295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024]
Abstract
Long COVID, also known as Post COVID-19 condition by the World Health Organization or Post-Acute Sequelae of SARS-CoV-2 infection (PASC), is defined as the development of symptoms such as post-exertional malaise, dysgeusia, and partial or full anosmia three months after initial SARS-CoV-2 infection. The multisystem effects of PASC make it difficult to distinguish from its mimickers. Further, a comprehensive evaluation of the gut microbiome, nutrition, and PASC has yet to be studied. The gut-brain axis describes bidirectional immune, neural, endocrine, and humoral modulatory interactions between the gut microbiome and brain function. We explore recent studies that support an association between alterations in gut microbiome diversity and the severity of acute-phase COVID-19, and how these may be affected by diets rich in antioxidants and fiber. The Mediterranean Diet (MeDi) has demonstrated promising neuroprotective effects through its anti-inflammatory processes. Further, diets rich in fiber increase gut diversity and increase the amount of short-chain fatty acids (SCFAs) within the body-both shown to protect from acute COVID-19 complications. Long-term changes to the gut microbiome persist after acute infection and may increase susceptibility to PASC. This study builds on existing knowledge of determinants of PASC and highlights a relationship between nutrition, gut microbiome, acute-phase COVID-19, and, subsequently, PASC susceptibility.
Collapse
Affiliation(s)
- Zabrina Reyes
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America
| | - Mary Catherine Stovall
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America
| | - Sanjana Punyamurthula
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America
| | - Michele Longo
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America
| | - Demetrius Maraganore
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America; Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, United States of America; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, United States of America
| | - Rebecca J Solch-Ottaiano
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, United States of America; Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, United States of America; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, United States of America.
| |
Collapse
|
5
|
Mamalelala TT, Karmen-Tuohy S, Chimbwete L, Mokone DJ, Shapiro R, Young C, Schwanke Khilji S. Perceptions of prevalence and management of post-acute sequelae of SARS-CoV-2 (PASC) infection among healthcare workers in Kweneng District, Botswana: Report of a district-wide survey. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003865. [PMID: 39602378 PMCID: PMC11602055 DOI: 10.1371/journal.pgph.0003865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/06/2024] [Indexed: 11/29/2024]
Abstract
Over 9.5 million confirmed cases of COVID-19 infection have been recorded in Africa. The syndrome of post-acute sequelae of SARS-CoV-2 infection (PASC) affects an estimated 32% to 87% of COVID patients globally. Data regarding prevalence and impact of PASC in Botswana are limited. This study used a cross-sectional survey design to query healthcare workers in Kweneng District, Botswana about perceived PASC prevalence, duration, symptoms, impact, and management strategies. The survey was disseminated to participants via pre-existing WhatsApp groups and paper copy. Descriptive statistics were used to analyse quantitative data, including demographic data. 72 respondents consented and completed the survey, from an estimated 650 staff meeting eligibility criteria; 63% were female and 36% were male. The majority (90%) were nurses, with doctors and "other" accounting for 6% and 4% of respondents, respectively; no administrators responded. Over half (72%) worked at primary care facilities and 28% worked in hospitals. Nearly all (93%) indicated seeing patients with PASC on a weekly basis, though the majority (61%) identified these patients as comprising <10% of total patients. The most frequently reported PASC symptom was persistent cough (64%), followed by shortness of breath (54%) and fatigue (49%). A substantial minority of respondents were unsure how to manage common PASC symptoms, with 29% and 36% indicating uncertainty regarding management of persistent cough and fatigue, respectively. Findings indicate that PASC symptoms are frequently encountered in clinical practice in Botswana with significant overlap with acute COVID-19, influenza-like illnesses, and tuberculosis, likely placing increased burden on existing health system processes. Providers reported uncertainty in managing presumed PASC, and current practice patterns may contribute to unintended adverse effects. Clear clinical algorithms for PASC screening, diagnosis, and management should be developed and disseminated in Botswana to mitigate the effects of PASC symptoms and improve the quality of life of COVID-19 survivors.
Collapse
Affiliation(s)
- Tebogo T. Mamalelala
- Faculty of Health Sciences, School of Nursing, University of Botswana, Gaborone, Botswana
| | - Savannah Karmen-Tuohy
- NYU Grossman School of Medicine, New York University, New York, New York, United States of America
| | - Lettie Chimbwete
- Department of Health Sciences, University of Pretoria, Pretoria, South Africa
| | | | - Roger Shapiro
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Botswana-Harvard Health Partnership, Gaborone, Botswana
| | - Claire Young
- NYU Grossman School of Medicine, New York University, New York, New York, United States of America
| | - Sara Schwanke Khilji
- Botswana-Harvard Health Partnership, Gaborone, Botswana
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
6
|
Goniewicz K. Confronting the Ongoing Challenges of COVID-19: A Comprehensive Approach for Health Care Systems. Disaster Med Public Health Prep 2024; 18:e214. [PMID: 39463310 DOI: 10.1017/dmp.2024.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
|
7
|
Grach SL, Dudenkov DV, Pollack B, Fairweather D, Aakre CA, Munipalli B, Croghan IT, Mueller MR, Overgaard JD, Bruno KA, Collins NM, Li Z, Hurt RT, Tal MC, Ganesh R, Knight DTR. Overlapping conditions in Long COVID at a multisite academic center. Front Neurol 2024; 15:1482917. [PMID: 39524912 PMCID: PMC11543549 DOI: 10.3389/fneur.2024.1482917] [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: 08/19/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Background Many patients experience persistent symptoms after COVID-19, a syndrome referred to as Long COVID (LC). The goal of this study was to identify novel new or worsening comorbidities self-reported in patients with LC. Methods Patients diagnosed with LC (n = 732) at the Mayo Long COVID Care Clinic in Rochester, Minnesota and Jacksonville, Florida were sent questionnaires to assess the development of new or worsening comorbidities following COVID-19 compared to patients with SARS-CoV-2 that did not develop LC (controls). Both groups were also asked questions screening for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), generalized joint hypermobility (GJH) and orthostatic intolerance. 247 people with LC (33.7%) and 40 controls (50%) responded to the surveys. Results In this study LC patients averaged 53 years of age and were predominantly White (95%) women (75%). The greatest prevalence of new or worsening comorbidities following SARS-CoV-2 infection in patients with LC vs. controls reported in this study were pain (94.4% vs. 0%, p < 0.001), neurological (92.4% vs. 15.4%, p < 0.001), sleep (82.8% vs. 5.3%, p < 0.001), skin (69.8% vs. 0%, p < 0.001), and genitourinary (60.6% vs. 25.0%, p = 0.029) issues. 58% of LC patients screened positive for ME/CFS vs. 0% of controls (p < 0.001), 27% positive for GJH compared to 10% of controls (p = 0.026), and a positive average score of 4.0 on orthostatic intolerance vs. 0 (p < 0.001). The majority of LC patients with ME/CFS were women (77%). Conclusion We found that comorbidities across 12 surveyed categories were increased in patients following SARS-CoV-2 infection. Our data also support the overlap of LC with ME/CFS, GJH, and orthostatic intolerance. We discuss the pathophysiologic, research, and clinical implications of identifying these conditions with LC.
Collapse
Affiliation(s)
- Stephanie L. Grach
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Daniel V. Dudenkov
- Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Beth Pollack
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - DeLisa Fairweather
- Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
- Department of Immunology, Mayo Clinic, Jacksonville, FL, United States
| | - Chris A. Aakre
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Bala Munipalli
- Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Ivana T. Croghan
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Division of Quantitative Health Sciences, Rochester, MN, United States
| | - Michael R. Mueller
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Joshua D. Overgaard
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Katelyn A. Bruno
- Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Cardiovascular Medicine, University of Florida, Gainesville, FL, United States
| | - Nerissa M. Collins
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Zhuo Li
- Department of Biostatistics, Mayo Clinic, Jacksonville, FL, United States
| | - Ryan T. Hurt
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Michal C. Tal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ravindra Ganesh
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Dacre T. R. Knight
- Department of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
8
|
Ganesh R, Munipalli B. Long COVID and hypermobility spectrum disorders have shared pathophysiology. Front Neurol 2024; 15:1455498. [PMID: 39301475 PMCID: PMC11410636 DOI: 10.3389/fneur.2024.1455498] [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: 06/27/2024] [Accepted: 08/02/2024] [Indexed: 09/22/2024] Open
Abstract
Hypermobility spectrum disorders (HSD) and hypermobile Ehlers-Danlos syndrome (hEDS) are the most common joint hypermobility conditions encountered by physicians, with hypermobile and classical EDS accounting for >90% of all cases. Hypermobility has been detected in up to 30-57% of patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), fibromyalgia, postural orthostatic tachycardia syndrome (POTS), and long COVID (LC) compared to the general population. Extrapulmonary symptoms, including musculoskeletal pain, dysautonomia disorders, cognitive disorders, and fatigue, are seen in both LC and HSD. Additionally, ME/CFS has overlapping symptoms with those seen in HSD. Mast cell activation and degranulation occurring in both LC and ME/CFS may result in hyperinflammation and damage to connective tissue in these patients, thereby inducing hypermobility. Persistent inflammation may result in the development or worsening of HSD. Hence, screening for hypermobility and other related conditions including fibromyalgia, POTS, ME/CFS, chronic pain conditions, joint pain, and myalgia is essential for individuals experiencing LC. Pharmacological treatments should be symptom-focused and geared to a patient's presentation. Paced exercise, massage, yoga, and meditation may also provide benefits.
Collapse
Affiliation(s)
- Ravindra Ganesh
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Bala Munipalli
- Division of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
9
|
Setiabudi D, Azhali BA, Tirtosudiro MA, Ramadhan MH, Rinaldhi M, Nataprawira HM. Long COVID or Post-Acute Sequelae of COVID-19 (PASC) in Children and Adolescents. Clin Med Res 2024; 22:131-137. [PMID: 39438144 PMCID: PMC11495663 DOI: 10.3121/cmr.2024.1858] [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: 06/17/2023] [Revised: 08/11/2024] [Accepted: 08/19/2024] [Indexed: 10/25/2024]
Abstract
Introduction: Few studies of children with long COVID (post-COVID-19 condition) or post-acute sequelae of SARS CoV-2 (PASC) have been reported. Those terms describe symptoms that persist for weeks or months or as new symptoms that develop after SARS-CoV-2 infection. This condition might be found to various degrees in the severity of COVID-19. This study aimed to describe long COVID in confirmed SARS-CoV-2-infected children.Design: An observational cross-sectional study.Setting: Tertiary care hospital between January and November 2021.Participants: Children, age 5-17 years, with virologically confirmed COVID?19.Methods: This study was conducted by completing an electronic form after informed consent was obtained. The subject's characteristics and parent's or guardian's phone numbers were retrieved from the pediatric COVID-19 registry. Parents were contacted to complete a structured electronic questionnaire about the long COVID symptoms noticed in their children. Descriptive statistics were displayed in percentages and median.Results: Parent contact numbers were documented in 125/135 children who fulfilled the study criteria. There were 61 parents (48.8%) who gave consent, while the rest either did not respond or refused. There were 16 children reported as deceased primarily due to chronic renal disease and leukemia. Of the 45 children enrolled, the median (IQR) age was 11 years (5.3-17.6); 51.1% were female; and 75.6% had comorbidities. Two-thirds developed long COVID symptoms, most frequently in the age 5-9 years group, and mostly fatigue (45.2%), decreased appetite (38.7%), and muscle aches (32.3%). All patients with moderate to severe and more than half asymptomatic to mild COVID-19 developed long COVID.Conclusion: Most children had long COVID symptoms similar to adults despite being otherwise asymptomatic or having mild COVID-19.
Collapse
Affiliation(s)
- Djatnika Setiabudi
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Buti Azfiani Azhali
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Akbar Tirtosudiro
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Monika Hasna Ramadhan
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhamad Rinaldhi
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Heda Melinda Nataprawira
- Department of Child Health, Hasan Sadikin General Hospital / Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
10
|
Ghorra N, Popotas A, Besse-Hammer T, Rogiers A, Corazza F, Nagant C. Cytokine Profile in Patients with Postacute Sequelae of COVID-19. Viral Immunol 2024; 37:346-354. [PMID: 39172652 DOI: 10.1089/vim.2024.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
The enduring impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its disease manifestation, COVID-19, on public health remains significant. Postacute sequelae of SARS-CoV-2 infection (PASC) affect a considerable number of patients, impairing their quality of life. While the role of the cytokine storm in acute COVID-19 is well established, its contribution to the pathophysiology of PASC is not fully understood. This study aimed to analyze the cytokine profile of patients with PASC following in vitro stimulation of Toll-like receptor (TLR) pathways, comparing them with a healthy control group. From October 2020 till March 2021, Brugmann University Hospital's clinical research unit included patients with PASC in the study. Whole blood samples were collected from 50 patients and 25 healthy volunteers. After in vitro stimulation under five different conditions, cytokine levels were measured using a multiplex method. Significantly decreased cytokine levels were observed in patients with PASC compared with healthy volunteers, particularly after TLR4 (interleukin [IL]-1α, IL-1β, IL-2, IL-10, interferon (IFN)α, IFNγ, IFNω, and tumor necrosis factor (TNF)α) and TLR7/8 (IL-1α, IL-1β, IFNα, IFNω, IFNγ, and TNFα) pathway stimulation. Principal component analysis identified two distinct clusters, suggesting a likely dysregulation of immunity involving TLR4 and TLR7/8 pathways in patients with PASC. Our study suggests that TLR4 and TLR7/8 pathways play a role in the pathophysiology of PASC. Continuous basal activation of immunity could explain the high basal concentrations of cytokines described in these patients and the decreased amplitude of response of these signaling pathways following specific stimulation.
Collapse
Affiliation(s)
- Nathalie Ghorra
- Department of Immunology, LHUB-ULB (Laboratoire Hospitalier Universitaire de Bruxelles), Brussels, Belgium
| | - Alexandros Popotas
- Translational Research Unit, Hôpital Universitaire Des Enfants Reine Fabiola, Bruxelles, Belgique
- Translational Research Unit, Université Libre de Bruxelles, CHU Brugmann, Brussels, Belgium
| | - Tatiana Besse-Hammer
- Department of Clinical Research, Brugmann University Hospital, Brussels, Belgium
- Department of Neurology, Brugmann University Hospital, Brussels, Belgium
| | - Anne Rogiers
- Department of Clinical Research, Brugmann University Hospital, Brussels, Belgium
- Department of Neurology, Brugmann University Hospital, Brussels, Belgium
| | - Francis Corazza
- Department of Immunology, LHUB-ULB (Laboratoire Hospitalier Universitaire de Bruxelles), Brussels, Belgium
- Translational Research Unit, Hôpital Universitaire Des Enfants Reine Fabiola, Bruxelles, Belgique
| | - Carole Nagant
- Department of Immunology, LHUB-ULB (Laboratoire Hospitalier Universitaire de Bruxelles), Brussels, Belgium
| |
Collapse
|
11
|
Sprague Martinez L, Sharma N, John J, Battaglia TA, Linas BP, Clark CR, Hudson LB, Lobb R, Betz G, Ojala O'Neill SO, Lima A, Doty R, Rahman S, Bassett IV. Long COVID impacts: the voices and views of diverse Black and Latinx residents in Massachusetts. BMC Public Health 2024; 24:2265. [PMID: 39169314 PMCID: PMC11337633 DOI: 10.1186/s12889-024-19567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE To understand how Long COVID is impacting the health and social conditions of the Black and Latinx communities. BACKGROUND Emerging research on Long COVID has identified three distinct characteristics, including multi-organ damage, persistent symptoms, and post-hospitalization complications. Given Black and Latinx communities experienced significantly higher COVID rates in the first phase of the pandemic they may be disproportionately impacted by Long COVID. METHODS Eleven focus groups were conducted in four languages with diverse Black and Latinx individuals (n = 99) experiencing prolonged symptoms of COVID-19 or caring for family members with prolonged COVID-19 symptoms. Data was analyzed thematically. RESULTS Most participants in non-English language groups reported they were unfamiliar with the diagnosis of long COVID, despite experiencing symptoms. Long COVID impacts spanned financial and housing stability to physical and mental health impacts. Participants reported challenging encounters with health care providers, a lack of support managing symptoms and difficulty performing activities of daily living including work. CONCLUSIONS There is a need for multilingual, accessible information about Long COVID symptoms, improved outreach and healthcare delivery, and increased ease of enrollment in long-term disability and economic support programs.
Collapse
Affiliation(s)
- Linda Sprague Martinez
- Health Disparities Institute, UConn Health, 241 Main Street, Hartford, CT, 06106, USA.
- School of Medicine, University of Connecticut, 263 Farmington Avenue, Farmington, CT, USA.
| | | | - Janice John
- Cambridge Health Alliance, Cambridge, MA, USA
| | - Tracy A Battaglia
- Boston University School of Medicine, Boston Medical Center, Boston University Clinical and Translational Science Institute, Boston, MA, USA
| | - Benjamin P Linas
- Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | | | - Linda B Hudson
- Tufts University School of Public Health and Community Medicine, Boston, MA, USA
| | - Rebecca Lobb
- Boston University Clinical and Translational Science Institute, Boston, MA, USA
| | - Gillian Betz
- Health Disparities Institute, UConn Health, 241 Main Street, Hartford, CT, 06106, USA
| | | | - Angelo Lima
- Archipelago Strategies Group, Boston, MA, USA
| | - Ross Doty
- Archipelago Strategies Group, Boston, MA, USA
| | | | | |
Collapse
|
12
|
Mukkawar RV, Reddy H, Rathod N, Kumar S, Acharya S. The Long-Term Cardiovascular Impact of COVID-19: Pathophysiology, Clinical Manifestations, and Management. Cureus 2024; 16:e66554. [PMID: 39258051 PMCID: PMC11384648 DOI: 10.7759/cureus.66554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has resulted in a substantial global health crisis, with effects extending far beyond the acute phase of infection. This review aims to provide a comprehensive overview of the long-term cardiovascular impact of COVID-19, focusing on the pathophysiology, clinical manifestations, diagnostic approaches, management strategies, and future research directions. SARS-CoV-2 induces cardiovascular complications through mechanisms such as inflammation, endothelial dysfunction, and direct myocardial injury, leading to conditions like myocarditis, heart failure, arrhythmias, and thromboembolic events. These long-term effects, collectively called "long COVID" or post-acute sequelae of SARS-CoV-2 infection (PASC), present significant challenges for healthcare systems and patient management. Diagnostic approaches include imaging techniques and laboratory tests to identify and monitor cardiovascular complications. Management strategies emphasize a holistic approach, incorporating pharmacological treatments and lifestyle modifications. Special attention is required for vulnerable populations, including those with pre-existing cardiovascular conditions. Ongoing research is essential to understand the full spectrum of long-term cardiovascular impacts and to develop effective treatments. This review highlights the critical need for continued vigilance, multidisciplinary care, and research to address the cardiovascular sequelae of COVID-19 and improve long-term health outcomes for survivors.
Collapse
Affiliation(s)
- Rushi V Mukkawar
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Harshitha Reddy
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Nishant Rathod
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sourya Acharya
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
13
|
Manuelpillai B, Zendt M, Chang-Rabley E, Ricotta EE. Stuck in pandemic uncertainty: a review of the persistent effects of COVID-19 infection in immune-deficient people. Clin Microbiol Infect 2024; 30:1007-1011. [PMID: 38552795 PMCID: PMC11254561 DOI: 10.1016/j.cmi.2024.03.027] [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: 01/17/2024] [Revised: 03/04/2024] [Accepted: 03/23/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND People who are immune-deficient/disordered (IDP) are underrepresented in COVID-19 studies. Specifically, there is limited research on post-SARS-CoV-2 infection outcomes, including viral persistence and long-term sequelae in these populations. OBJECTIVES This review aimed to examine the published literature on the occurrence of persistent SARS-CoV-2 positivity, relapse, reinfections, variant coinfection, and post-acute sequelae of COVID-19 in IDP. Although the available literature largely centred on those with secondary immunodeficiencies, studies on people with inborn errors of immunity are also included. SOURCES PubMed was searched using medical subject headings terms to identify relevant articles from the last 4 years. Articles on primary and secondary immunodeficiencies were chosen, and a special emphasis was placed on including articles that studied people with inborn errors of immunity. The absence of extensive cohort studies including these individuals has limited most articles in this review to case reports, whereas the articles focusing on secondary immunodeficiencies include larger cohort, case-control, and cross-sectional studies. Articles focusing solely on HIV/AIDS were excluded. CONTENT Scientific literature suggests that IDP of any age are more likely to experience persistent SARS-CoV-2 infections. Although adult IDP exhibits a higher rate of post-acute sequelae of COVID-19, milder COVID-19 infections in children may reduce their risk of experiencing post-acute sequelae of COVID-19. Reinfections and coinfections may occur at a slightly higher rate in IDP than in the general population. IMPLICATIONS Although IDP experience increased viral persistence and inter-host evolution, it is unlikely that enough evidence can be generated at the population-level to support or refute the hypothesis that infections in IDP are significantly more likely to result in variants of concern than infections in the general population. Additional research on the relationship between viral persistence and the rate of long-term sequelae in IDP could inform the understanding of the immune response to SARS-CoV-2 in IDP and the general population.
Collapse
Affiliation(s)
- Bevin Manuelpillai
- Rollins School of Public Health, Emory University, Atlanta, GA, USA; Epidemiology and Data Management Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mackenzie Zendt
- Epidemiology and Data Management Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emma Chang-Rabley
- Epidemiology and Data Management Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emily E Ricotta
- Epidemiology and Data Management Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
14
|
Li H, Wan L, Liu M, Ma E, Huang L, Yang Y, Li Q, Fang Y, Li J, Han B, Zhang C, Sun L, Hou X, Li H, Sun M, Qian S, Duan X, Zhao R, Yang X, Chen Y, Wu S, Zhang X, Zhang Y, Cheng G, Chen G, Gao Q, Xu J, Hou L, Wei C, Zhong H. SARS-CoV-2 spike-induced syncytia are senescent and contribute to exacerbated heart failure. PLoS Pathog 2024; 20:e1012291. [PMID: 39102426 PMCID: PMC11326701 DOI: 10.1371/journal.ppat.1012291] [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: 10/18/2023] [Revised: 08/15/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
SARS-CoV-2 spike protein (SARS-2-S) induced cell-cell fusion in uninfected cells may occur in long COVID-19 syndrome, as circulating SARS-2-S or extracellular vesicles containing SARS-2-S (S-EVs) were found to be prevalent in post-acute sequelae of COVID-19 (PASC) for up to 12 months after diagnosis. Although isolated recombinant SARS-2-S protein has been shown to increase the SASP in senescent ACE2-expressing cells, the direct linkage of SARS-2-S syncytia with senescence in the absence of virus infection and the degree to which SARS-2-S syncytia affect pathology in the setting of cardiac dysfunction are unknown. Here, we found that the senescent outcome of SARS-2-S induced syncytia exacerbated heart failure progression. We first demonstrated that syncytium formation in cells expressing SARS-2-S delivered by DNA plasmid or LNP-mRNA exhibits a senescence-like phenotype. Extracellular vesicles containing SARS-2-S (S-EVs) also confer a potent ability to form senescent syncytia without de novo synthesis of SARS-2-S. However, it is important to note that currently approved COVID-19 mRNA vaccines do not induce syncytium formation or cellular senescence. Mechanistically, SARS-2-S syncytia provoke the formation of functional MAVS aggregates, which regulate the senescence fate of SARS-2-S syncytia by TNFα. We further demonstrate that senescent SARS-2-S syncytia exhibit shrinked morphology, leading to the activation of WNK1 and impaired cardiac metabolism. In pre-existing heart failure mice, the WNK1 inhibitor WNK463, anti-syncytial drug niclosamide, and senolytic dasatinib protect the heart from exacerbated heart failure triggered by SARS-2-S. Our findings thus suggest a potential mechanism for COVID-19-mediated cardiac pathology and recommend the application of WNK1 inhibitor for therapy especially in individuals with post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Huilong Li
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, China
| | - Muyi Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Qihong Li
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Bingqing Han
- Beijing Institute of Biotechnology, Beijing, China
| | - Chang Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Lijuan Sun
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Xufeng Hou
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingyu Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sichong Qian
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuhui Zhang
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | | | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Gengye Chen
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi Gao
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Junjie Xu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
15
|
Resendes NM, Bradley J, Tang F, Hammel IS, Ruiz JG. The association of non-severe COVID-19 infection and progression to frailty among robust older veterans. J Nutr Health Aging 2024; 28:100296. [PMID: 38901116 DOI: 10.1016/j.jnha.2024.100296] [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: 04/01/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Studies have shown that frailty was increased in hospitalized COVID-19 patients. However, it is not clear whether non-severe COVID-19 increases the risk for pre-frailty and frailty development. Our study aimed to determine the risk of developing frailty and pre-frailty in robust veterans who contracted non-severe COVID-19. METHODS We conducted a retrospective cohort study to assess the association of SARS-CoV-2 infection with the development of pre-frailty and frailty status among robust U.S. veterans using VA COVID-19 Shared Data Resource. We included patients 55 years and older who had at least one SARS-CoV-2 testing between March 15, 2020, and November 30, 2020, had been active patients in the past 12 months, and had a VA frailty index of zero (robust status) at the time of testing. Cox proportional hazard model was used to assess the association between COVID-19 infection and developing frailty or pre-frailty and frailty. We also assessed the association by patients' age groups, sex, and race. FINDINGS We identified 82070 veterans mean age 68.3 ± 7.8, 74738 (91.1%) male, 53899 (65.7%) white, 7557 (9.2%) with mild COVID-19 infection. Over the follow up period of 36 months, testing positive for COVID-19 was associated with a 66% increase in the hazard of becoming frail (adjusted HR = 1.66, 95%CI: 1.32-2.08), and a 68% increase in the hazard of becoming pre-frail (adjusted HR = 1.68, 95%CI: 1.45-1.94). Among male patients, mild COVID-19 infection was associated with a 54% increase in the hazard of becoming frail (adjusted HR = 1.54, 95% CI: 1.21-1.96), while among female patients there was a 330% increase (adjusted HR = 4.30, 95% CI: 2.13-8.64). CONCLUSIONS AND RELEVANCE Non-severe COVID-19 infection that occurred in robust older adults increased the risk of developing frailty. Further multi-center prospective cohort studies evaluating the mechanism of action and clinical trials of treatment options for post-COVID frailty are indicated in Veterans to support clinical care.
Collapse
Affiliation(s)
- Natasha M Resendes
- Miami VA (Veterans Administration) Healthcare System Geriatric Research, Education and Clinical Center (GRECC), Miami, Florida, USA; Dept. of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Jerry Bradley
- Dept. of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Fei Tang
- Miami VA (Veterans Administration) Healthcare System Geriatric Research, Education and Clinical Center (GRECC), Miami, Florida, USA
| | - Iriana S Hammel
- Miami VA (Veterans Administration) Healthcare System Geriatric Research, Education and Clinical Center (GRECC), Miami, Florida, USA; Dept. of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jorge G Ruiz
- Memorial Healthcare System, Hollywood, Florida, USA; Dept of Medicine, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, Florida, USA
| |
Collapse
|
16
|
Tarcha R, Khalayli N, Kudsi M. Sicca syndrome as complication of COVID-19 infection: A case report. Clin Case Rep 2024; 12:e9177. [PMID: 39021490 PMCID: PMC11252430 DOI: 10.1002/ccr3.9177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Several reports of suspected oral and ocular manifestations of coronavirus disease 2019 (COVID-19) has prompted investigations into ocular signs, symptoms, and transmission (5).11.2% of patients with COV19 infection had ocular symptoms, including ocular pain, conjunctivitis, dry eye, and floaters, meanwhile, many studies had documented oral symptoms such as dry mouth and dysgeusia in these patients. Our case reported a 39-year-old male, presented with symptoms of dry mouth and dry eye lasting more than 3 months. The patient had recovered from (PCR-confirmed) COVID-19 which lasted 10 days, 4 months ago. The physical examination was normal. Ocular findings include conjunctival hyperemia and superficial punctate keratitis. The anti-nuclear antibody (ANA) was weekly positive at 1/80. Schirmer test considered positive. He continued on 200 mg/day of hydroxychloroquine, along with tear drops until now with remission. Sicca symptoms may be a sequel of COVID-19 infection, and physicians should be aware of this sequel. The sequela of this infection is not understood, with limited data in the literature. Future prospective cohort studies are needed to reveal the impact of these features on oral health.
Collapse
Affiliation(s)
- Raghad Tarcha
- Department of Rheumatology, Faculty of MedicineDamascus UniversityDamascusSyria
| | - Naram Khalayli
- Department of Psychiatry, Faculty of MedicineDamascus UniversityDamascusSyria
| | | |
Collapse
|
17
|
Baumert BO, Wang H, Samy S, Park SK, Lam CN, Dunn K, Pinto-Pacheco B, Walker D, Landero J, Conti D, Chatzi L, Hu H, Goodrich JA. Environmental pollutant risk factors for worse COVID-19 related clinical outcomes in predominately hispanic and latino populations. ENVIRONMENTAL RESEARCH 2024; 252:119072. [PMID: 38729411 PMCID: PMC11198996 DOI: 10.1016/j.envres.2024.119072] [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: 01/19/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Per- and poly-fluorinated compounds (PFAS) and heavy metals constitute two classes of environmental exposures with known immunotoxicant effects. In this pilot study, we aimed to evaluate the impact of exposure to heavy metals and PFAS on COVID-19 severity. We hypothesized that elevated plasma-PFAS concentrations and urinary heavy metal concentrations would be associated with increased odds of ICU admission in COVID-19 hospitalized individuals. METHODS Using the University of Southern California Clinical Translational Sciences Institute (SC-CTSI) biorepository of hospitalized COVID-19 patients, urinary concentrations of 15 heavy metals and urinary creatinine were measured in n = 101 patients and plasma concentrations of 13 PFAS were measured in n = 126 patients. COVID-19 severity was determined based on whether a patient was admitted to the ICU during hospitalization. Associations of metals and PFAS with ICU admission were assessed using logistic regression models, controlling for age, sex, ethnicity, smoking status, and for metals, urinary dilution. RESULTS The average age of patients was 55 ± 14.2 years. Among SC-CTSI participants with urinary measurement of heavy metals and blood measures of PFAS, 54.5% (n = 61) and 54.8% (n = 80) were admitted to the ICU, respectively. For heavy metals, we observed higher levels of Cd, Cr, and Cu in ICU patients. The strongest associations were with Cadmium (Cd). After accounting for covariates, each 1 SD increase in Cd resulted in a 2.00 (95% CI: 1.10-3.60; p = 0.03) times higher odds of admission to the ICU. When including only Hispanic or Latino participants, the effect estimates between cadmium and ICU admission remained similar. Results for PFAS were less consistent, with perfluorodecanesulfonic acid (PFDS) exhibiting a positive but non-significant association with ICU admission (Odds ratio, 95% CI: 1.50, 0.97-2.20) and perfluorodecanoic acid (PFDA) exhibiting a negative association with ICU admission (0.53, 0.31-0.88). CONCLUSIONS This study supports the hypothesis that environmental exposures may impact COVID-19 severity.
Collapse
Affiliation(s)
- Brittney O Baumert
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hongxu Wang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Shar Samy
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, United States
| | - Sung Kyun Park
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Chun Nok Lam
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kathryn Dunn
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Brismar Pinto-Pacheco
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Douglas Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Julio Landero
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - David Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Leda Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Howard Hu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Jesse A Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
18
|
Barboza-Solis C, Fantin R, Hildesheim A, Pfeiffer R, Porras C, Butt J, Waterboer T, Raventós H, Abdelnour A, Aparicio A, Loria V, Prevots DR, Gail MH, Herrero R. COVID-19 and long-term impact on symptoms and Health-Related Quality of Life in Costa Rica: the RESPIRA cohort study. BMC Infect Dis 2024; 24:557. [PMID: 38834971 DOI: 10.1186/s12879-024-09450-6] [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: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Evidence continues to accumulate regarding the potential long-term health consequences of COVID-19 in the population. To distinguish between COVID-19-related symptoms and health limitations from those caused by other conditions, it is essential to compare cases with community controls using prospective data ensuring case-control status. The RESPIRA study addresses this need by investigating the lasting impact of COVID-19 on Health-related Quality of Life (HRQoL) and symptomatology in a population-based cohort in Costa Rica, thereby providing a robust framework for controlling HRQoL and symptoms. METHODS The study comprised 641 PCR-confirmed, unvaccinated cases of COVID-19 and 947 matched population-based controls. Infection was confirmed using antibody tests on enrollment serum samples and symptoms were monitored monthly for 6 months post-enrolment. Administered at the 6-month visit (occurring between 6- and 2-months post-diagnosis for cases and 6 months after enrollment for controls), HRQoL and Self-Perceived Health Change were assessed using the SF-36, while brain fog, using three items from the Mental Health Inventory (MHI). Regression models were utilized to analyze SF-36, MHI scores, and Self-Perceived Health Change, adjusted for case/control status, severity (mild case, moderate case, hospitalized) and additional independent variables. Sensitivity analyses confirmed the robustness of the findings. RESULTS Cases showed significantly higher prevalences of joint pain, chest tightness, and skin manifestations, that stabilized at higher frequencies from the fourth month post-diagnosis onwards (2.0%, 1.2%, and 0.8% respectively) compared to controls (0.9%, 0.4%, 0.2% respectively). Cases also exhibited significantly lower HRQoL than controls across all dimensions in the fully adjusted model, with a 12.4 percentage-point difference [95%CI: 9.4-14.6], in self-reported health compared to one year prior. Cases reported 8.0% [95%CI: 4.2, 11.5] more physical limitations, 7.3% [95%CI: 3.5, 10.5] increased lack of vitality, and 6.0% [95%CI: 2.4, 9.0] more brain fog compared to controls with similar characteristics. Undiagnosed cases detected with antibody tests among controls had HRQoL comparable to antibody negative controls. Differences were more pronounced in individuals with moderate or severe disease and among women. CONCLUSIONS PCR-confirmed unvaccinated cases experienced prolonged HRQoL reductions 6 months to 2 years after diagnosis, this was particularly the case in severe cases and among women. Mildly symptomatic cases showed no significant long-term sequelae.
Collapse
Affiliation(s)
| | - Romain Fantin
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Allan Hildesheim
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Ruth Pfeiffer
- Biostatistics Branch, Division of Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Julia Butt
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henriette Raventós
- Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica
- Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
| | | | - Amada Aparicio
- Caja Costarricense de Seguro Social, San José, Costa Rica
| | - Viviana Loria
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - D Rebecca Prevots
- Epidemiology and Population Studies Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Mitchell H Gail
- Biostatistics Branch, Division of Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive RM 7-E138, MSC 9780, Bethesda, MD, 20892, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| |
Collapse
|
19
|
Elfessi ZZ, Gardner J, Gordon HS, Rubinstein I. Long COVID in Women Veterans Residing in Underserved, Socioeconomically Disadvantaged Neighborhoods of Chicago. Popul Health Manag 2024; 27:227-230. [PMID: 38607581 DOI: 10.1089/pop.2024.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Affiliation(s)
- Zane Z Elfessi
- Emergency Medicine, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
- Research Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
| | - Jessica Gardner
- Research Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
| | - Howard S Gordon
- Medical Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
- Research Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
| | - Israel Rubinstein
- Medical Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
- Research Services, Jesse Brown VA Medical Center and University of Illinois Colleges of Pharmacy and Medicine in Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Stapleton T, Norris L, Clancy K, O'Gorman A, Bannan C, Kent B, Conlon P, Nadajaran P, Kerr C, Connolly D. Outcomes of a Fatigue Management Intervention for People With Post COVID-19 Condition. Am J Phys Med Rehabil 2024; 103:410-417. [PMID: 38014889 DOI: 10.1097/phm.0000000000002368] [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: 11/29/2023]
Abstract
OBJECTIVE Fatigue is identified as one of the most prevalent and persistent problems reported by people with post COVID-19 condition that negatively impacts on everyday living and resumption of pre-COVID-19 lifestyle. A pilot occupational therapy fatigue management intervention was designed for patients presenting with post COVID-19 condition fatigue. DESIGN A retrospective analysis was carried out after the delivery of the fatigue management intervention. Self-reported measures of fatigue, well-being, and health status were taken at baseline and repeated at 2 wks after intervention. Baseline and postintervention scores were compared using nonparametric analysis. RESULTS Sixty participants (73% female), median age 50.5 yrs (range, 17-74), 93% reporting symptoms persisting for 12 wks or longer, completed the fatigue management intervention. All participants reported moderate to severe fatigue impacting on everyday activity at baseline. The greatest impact of fatigue was on engagement in leisure and work activity. Statistically significant improvement in fatigue ( P < 0.001), well-being ( P < 0.001), and health status ( P < 0.001) were noted after the intervention. CONCLUSIONS Findings indicate the potential of occupational therapy fatigue management interventions to enable self-management strategies and reduce the negative impact of fatigue among people with post COVID-19 condition.
Collapse
Affiliation(s)
- Tadhg Stapleton
- From the Discipline of Occupational Therapy, School of Medicine, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland Trinity College Dublin, Dublin, Ireland (TS, DC); Department of Occupational Therapy, St James's Hospital, James's Street, Dublin, Ireland (LN, KC, AO'G); Department of Infectious Diseases, St James's Hospital, Dublin, Ireland, Dublin (CB, PC, CK, PN); Department of Clinical Medicine, School of Medicine, Trinity College, Dublin, Ireland (CB, BK); and Respiratory Medicine, St James's Hospital, Dublin, Ireland (BK, PN)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Qiao H, Deng X, Qiu L, Qu Y, Chiu Y, Chen F, Xia S, Muenzel C, Ge T, Zhang Z, Song P, Bonnin A, Zhao Z, Yuan W. SARS-CoV-2 induces blood-brain barrier and choroid plexus barrier impairments and vascular inflammation in mice. J Med Virol 2024; 96:e29671. [PMID: 38747003 PMCID: PMC11446308 DOI: 10.1002/jmv.29671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 07/18/2024]
Abstract
The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.
Collapse
Affiliation(s)
- Haowen Qiao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Xiangxue Deng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Lingxi Qiu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yafei Qu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yuanpu Chiu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Feixiang Chen
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Cheyene Muenzel
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Tenghuan Ge
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zixin Zhang
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Pengfei Song
- Department of Electrical and Computer Engineering, the Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana–Champaign, Urbana, IL, USA
| | - Alexandre Bonnin
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| |
Collapse
|
22
|
Hsiao YY, Elliott TR, Jaramillo J, Douglas ME, Powers MB, Warren AM. The Fatigue and Altered Cognition Scale among SARS-CoV-2 Survivors: Psychometric Properties and Item Correlations with Depression and Anxiety Symptoms. J Clin Med 2024; 13:2186. [PMID: 38673462 PMCID: PMC11050894 DOI: 10.3390/jcm13082186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/17/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objectives: This study examined the psychometric properties of the Fatigue and Altered Cognition Scale (FACs) among adult COVID-19 survivors and its unique ability to assess symptomology not accounted for by measures of depression and anxiety. Methods: COVID-19 survivors completed an online survey that included the FACs, a measure of brain fog and central fatigue with 20 items rated on a digital-analog scale. Useable data from 559 participants were analyzed to test the two-factor structure of the FACs, test for measurement invariance by sex and device was used to complete the survey (hand-held, computer), and item correlations with symptoms of depression and anxiety were examined. Results: The two-factor structure of the FACs replicated, supporting the separate assessments of brain fog and fatigue, χ2(164) = 1028.363, p < 0.001, CFI = 0.934, TLI = 0.923, RMSEA = 0.097, SRMR = 0.053. The FACs exhibited invariance at the scalar level, indicating item and factor integrity regardless of sex and device type. Using a correlation > 0.70 as a criterion (i.e., indicating more than 50% shared variance between two items), items on the FACs (assessing fatigue and lack of energy) were highly correlated with feeling tired or having little energy on the depression measure. No other items correlated with any anxiety symptom larger than 0.70. Conclusions: The FACs appears to be a psychometrically sound and efficient measure for use with COVID-19 survivors, assessing symptoms of brain fog and central fatigue that are not attributable to symptoms assessed by established measures of depression and anxiety.
Collapse
Affiliation(s)
- Yu-Yu Hsiao
- Department of Individual, Family and Community Education, University of New Mexico, Albuquerque, NM 87131, USA; (Y.-Y.H.); (J.J.)
| | - Timothy R. Elliott
- Department of Educational Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Julie Jaramillo
- Department of Individual, Family and Community Education, University of New Mexico, Albuquerque, NM 87131, USA; (Y.-Y.H.); (J.J.)
| | - Megan E. Douglas
- Baylor Scott & White Research Institute, Dallas, TX 75246, USA; (M.E.D.); (M.B.P.); (A.M.W.)
| | - Mark B. Powers
- Baylor Scott & White Research Institute, Dallas, TX 75246, USA; (M.E.D.); (M.B.P.); (A.M.W.)
| | - Ann Marie Warren
- Baylor Scott & White Research Institute, Dallas, TX 75246, USA; (M.E.D.); (M.B.P.); (A.M.W.)
| |
Collapse
|
23
|
Ren J, Gao Q, Zhou X, Chen L, Guo W, Feng K, Huang T, Cai YD. Identification of key gene expression associated with quality of life after recovery from COVID-19. Med Biol Eng Comput 2024; 62:1031-1048. [PMID: 38123886 DOI: 10.1007/s11517-023-02988-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Post-acute sequelae of COVID-19 (PASC) is a persistent complication of severe acute respiratory syndrome coronavirus 2 infection that includes symptoms, such as fatigue, cognitive impairment, and respiratory distress. These symptoms severely affect the quality of life of patients after their recovery from COVID-19. In this study, a group of machine learning algorithms analyzed the whole blood RNA-seq data from patients with different PASC levels. The purpose of this analysis was to identify the gene markers associated with PASC and the special expression patterns for different PASC levels. By comparing the quality of life of patients after the acute phase of COVID-19 and before the disease, samples in the dataset were divided into three groups, namely, "Better," "The Same," and "Worse." Each patient was represented by the expression levels of 58,929 genes. The machine learning-based workflow included six feature-ranking algorithms, incremental feature selection (IFS), and four classification algorithms. The feature ranking algorithms were in charge of assessing feature importance, whereas IFS with classification algorithms were used to extract essential genes and to construct efficient classifiers and classification rules. The expression of top genes in the results was associated with the immune response to viral infection, which is supported by the published literature. For example, patients with low CCDC18 expression and high CPED1 expression had good quality of life, whereas those with low CDC16 expression had poor quality of life.
Collapse
Affiliation(s)
- JingXin Ren
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Qian Gao
- Department of Pharmacy, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - XianChao Zhou
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Wei Guo
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, 200030, China
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou, 510507, China
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
24
|
Calcaterra G, Bassareo PP, Spaccarotella CA, Barillà F, Sciomer S, Vadalà G, de Gregorio C, Romeo F, Mehta JL, Mattioli AV. The long-term cardiovascular impairment of COVID 19: need for clarity in definition and terminology. Minerva Cardiol Angiol 2024; 72:24-31. [PMID: 37705368 DOI: 10.23736/s2724-5683.23.06316-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Clinical experience and several large studies in the field have found that SARS-CoV-2 infection can cause long-term persistent cardiovascular (CV) impairment beyond the acute phase of the disease. This has resulted in a major public health concern worldwide. Regarding COVID-related long-term involvement of various organs and systems, using specific definitions and terminology is crucial to point out time relationships, lingering damage, and outcome, mostly when symptoms and signs of CV disease persist beyond the acute phase. Due to a lack of a common standardized definition, investigators have used interchangeable terms such as "long COVID," "post-COVID," or "post-acute sequelae of COVID-19" to describe CV involvement, thus causing some confusion. For the sake of clarity, the aim of this paper is to discuss the definition and terminology used in defining sequelae after the acute phase of COVID-19, thus pointing out the meaning of definitions like acute cardiac injury, post-acute sequelae of COVID-19, long COVID syndrome, and increased risk of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
| | - Pier P Bassareo
- University College of Dublin, School of Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Carmen A Spaccarotella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy -
| | | | | | - Giuseppe Vadalà
- Division of Cardiology, P. Giaccone University Hospital, Palermo, Italy
| | - Cesare de Gregorio
- Department of Clinical and Experimental Cardiology, University of Messina, Messina, Italy
| | | | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences, VA Medical Center, Little Rock, AR, USA
| | - Anna V Mattioli
- Department of Medical and Surgical Sciences for Children and Adults University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
25
|
Dierckx W, De Backer W, De Meyer Y, Lauwers E, Franck E, De Backer J, Ides K. Personalized pulmonary rehabilitation program for patients with post-acute sequelae of COVID-19: A proof-of-concept retrospective study. Physiol Rep 2024; 12:e15931. [PMID: 38296347 PMCID: PMC10830387 DOI: 10.14814/phy2.15931] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/05/2024] Open
Abstract
Long-COVID patients present with a decline in physical fitness. The aim of this study is to reveal the impact of pulmonary rehabilitation (PR) on physical fitness, quality of life (QoL), and parameters of quantified thorax CT. Long-COVID patients enrolled in a 3-month PR program were retrospectively studied. PR included endurance and resistance training three times a week. Assessments pre- and post-rehabilitation included quantified chest CT, pulmonary function tests (PFT), six-minute walk test (6MWT), cardiopulmonary exercise test, and questionnaires: Hospital Anxiety and Depression Scale, post-COVID-19 Functional Status scale, Borg score, and EuroQol. Seventeen subjects (5M/12F), mean age 42 ± 13 years, were included. PR improved all questionnaires' results significantly. Only significant difference in PFT parameters was correlation between baseline total lung capacity (TLC) and difference in TLC pre- and post-rehabilitation (p = 0.002). 6MWT increased from 329 to 365 m (p < 0.001), VO2max changed from 21 to 24 mL/kg/min (p = 0.007), peak load increased from 116 to 141 Watt (p < 0.001). Blood volume in small pulmonary vessels of 1.25 to 5 mm2 in cross-sectional area (BV5%) was higher than observed in patients with acute COVID-19 infection. After rehabilitation, BV5% decreased from 65% to 62% (p = 0.020). These changes correlated directly with changes in TLC (p = 0.039). Quantified CT airway volume increased after rehabilitation (p = 0.013). After rehabilitation, TLC tended to normalize due to (re)opening of small airways, with decline in air trapping and recruitment of alveoli. Furthermore, this study revealed that pulmonary rehabilitation can improve QoL and physical fitness in long-COVID patients.
Collapse
Affiliation(s)
- Wendel Dierckx
- Centre for Research and Innovation in Care, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Multidisciplinary Medical CentreMedImprove BVKontichBelgium
| | - Wilfried De Backer
- Multidisciplinary Medical CentreMedImprove BVKontichBelgium
- Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- FLUIDDA NVKontichBelgium
| | - Yinka De Meyer
- Multidisciplinary Medical CentreMedImprove BVKontichBelgium
- Clinical OperationsFLUIDDA NVKontichBelgium
| | - Eline Lauwers
- Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Clinical OperationsFLUIDDA NVKontichBelgium
| | - Erik Franck
- Centre for Research and Innovation in Care, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | | | - Kris Ides
- Multidisciplinary Medical CentreMedImprove BVKontichBelgium
- Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- CoSys Research Lab, Faculty of Applied EngineeringUniversity of AntwerpAntwerpBelgium
- Flanders Make Strategic Research CentreLommelBelgium
- Department of PaediatricsAntwerp University HospitalEdegemBelgium
| |
Collapse
|
26
|
Marinkovic K, White DR, Alderson Myers A, Parker KS, Arienzo D, Mason GF. Cortical GABA Levels Are Reduced in Post-Acute COVID-19 Syndrome. Brain Sci 2023; 13:1666. [PMID: 38137114 PMCID: PMC10741691 DOI: 10.3390/brainsci13121666] [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: 11/11/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
After recovering from the acute COVID-19 illness, a substantial proportion of people continue experiencing post-acute sequelae of COVID-19 (PASC), also termed "long COVID". Their quality of life is adversely impacted by persistent cognitive dysfunction and affective distress, but the underlying neural mechanisms are poorly understood. The present study recruited a group of mostly young, previously healthy adults (24.4 ± 5.2 years of age) who experienced PASC for almost 6 months following a mild acute COVID-19 illness. Confirming prior evidence, they reported noticeable memory and attention deficits, brain fog, depression/anxiety, fatigue, and other symptoms potentially suggestive of excitation/inhibition imbalance. Proton magnetic resonance spectroscopy (1H-MRS) was used to examine the neurochemical aspects of cell signaling with an emphasis on GABA levels in the occipital cortex. The PASC participants were compared to a control (CNT) group matched in demographics, intelligence, and an array of other variables. Controlling for tissue composition, biological sex, and alcohol intake, the PASC group had lower GABA+/water than CNT, which correlated with depression and poor sleep quality. The mediation analysis revealed that the impact of PASC on depression was partly mediated by lower GABA+/water, indicative of cortical hyperexcitability as an underlying mechanism. In addition, N-acetylaspartate (NAA) tended to be lower in the PASC group, possibly suggesting compromised neuronal integrity. Persistent neuroinflammation may contribute to the pathogenesis of PASC-related neurocognitive dysfunction.
Collapse
Affiliation(s)
- Ksenija Marinkovic
- Spatio-Temporal Brain Imaging Lab, Department of Psychology, San Diego State University, San Diego, CA 92182, USA (A.A.M.); (D.A.)
- Department of Radiology, University of California, San Diego, CA 92093, USA
| | - David R. White
- Spatio-Temporal Brain Imaging Lab, Department of Psychology, San Diego State University, San Diego, CA 92182, USA (A.A.M.); (D.A.)
| | - Austin Alderson Myers
- Spatio-Temporal Brain Imaging Lab, Department of Psychology, San Diego State University, San Diego, CA 92182, USA (A.A.M.); (D.A.)
- Department of Psychiatry, University of California, San Diego, CA 92093, USA
| | - Katie S. Parker
- Spatio-Temporal Brain Imaging Lab, Department of Psychology, San Diego State University, San Diego, CA 92182, USA (A.A.M.); (D.A.)
| | - Donatello Arienzo
- Spatio-Temporal Brain Imaging Lab, Department of Psychology, San Diego State University, San Diego, CA 92182, USA (A.A.M.); (D.A.)
- Department of Radiology, University of California, San Diego, CA 92093, USA
| | - Graeme F. Mason
- Department of Radiology and Biomedical Imaging, Psychiatry, and Biomedical Engineering, Yale University, New Haven, CT 06520, USA;
| |
Collapse
|
27
|
Hammel IS, Tosi DM, Tang F, Pott H, Ruiz JG. Frailty as a risk factor for post-acute sequelae of COVID-19 among US veterans during the Delta and Omicron waves. J Am Geriatr Soc 2023; 71:3826-3835. [PMID: 37725480 DOI: 10.1111/jgs.18584] [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: 03/20/2023] [Revised: 07/17/2023] [Accepted: 08/14/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Older populations have suffered the highest rates of SARS-CoV-2 infection and associated complications, including Post-Acute Sequelae of SARS-CoV-2 infection (PASC). Frailty is a geriatric syndrome that often coexists with COVID-19 infection. The vulnerability to stressors caused by multisystemic dysfunction that characterizes frailty may predispose older adults to develop PASC. METHODS Retrospective cohort study using the VA COVID-19 Shared Data Resource to identify US veterans testing positive for SARS-CoV-2 between July 2021 and February 2022, without prior positive tests and who were alive 30 days after infection. Frailty was calculated using a 31-item VA Frailty Index generated from electronic health records. We categorized Veterans into robust (FI ≤ 0.10), prefrail (FI: >0.10- < 0.21), and frail (FI ≥ 0.21). We assessed the association between frailty and PASC and vaccination and PASC using Cox survival model, adjusting for covariates. RESULTS We identified 245,857 COVID-19-positive veterans surviving 30 days after infection. The mean age was 57.5 ± 16.5 years; 87.2% were males, 68.1% were white, and 9.0% were Hispanic. Almost half of the sample (48.9%) were classified as robust, while 28.3% were pre-frail and 22.7% were frail; 99,886 (40.6%) were fully vaccinated, and 33,516 (13.6%) received booster doses. Over a median follow-up of 143 days (IQR = 101), 23,890 (9.7%) patients developed PASC. Within 6 months after infection, frailty and pre-frailty were associated with a 41% (adjusted HR [aHR]:1.40 (95% CI: 1.35-1.47) and 15% (aHR: 1.17 (95% CI: 1.11-1.19) increase in the risk of PASC compared with the robust, respectively. Vaccination and booster doses before infection were associated with a 27% (aHR: 0.73 (95% CI: 0.71-0.75) and 33% (aHR: 0.66 (95% CI: 0.63-0.69) reduction in the risk of developing PASC, respectively. CONCLUSIONS Frailty was associated with an increased risk of developing PASC. Vaccination was associated with a decreased risk of PASC, further reduced by booster doses. Early recognition of frailty in patients with COVID-19 may assist in the early identification and management of PASC.
Collapse
Affiliation(s)
- Iriana S Hammel
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, Florida, USA
- Division of Geriatrics and Palliative Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Dominique M Tosi
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, Florida, USA
- Division of Geriatrics and Palliative Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Fei Tang
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, Florida, USA
| | - Henrique Pott
- Department of Medicine, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil
| | - Jorge G Ruiz
- Geriatric Research Education and Clinical Center, Miami VA Healthcare System, Miami, Florida, USA
- Division of Geriatric Medicine, Memorial Healthcare System, Hollywood, Florida, USA
| |
Collapse
|
28
|
Kim SA, Lee JS, Kim T, Kim TH, Kwon S, Kang JW. Efficacy and safety of acupuncture treatment for fatigue after COVID-19 infection: study protocol for a pilot randomized sham-controlled trial. Front Neurol 2023; 14:1302793. [PMID: 38033774 PMCID: PMC10684676 DOI: 10.3389/fneur.2023.1302793] [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: 10/04/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background As the coronavirus disease 2019 (COVID-19) pandemic has spread globally, its sequelae, called Long COVID, have persisted, troubling patients worldwide. Although fatigue is known to be the most frequent among Long COVID symptoms, its mechanism and treatment have not been clearly demonstrated. In 2022, we conducted a preliminary prospective case series and found that acupuncture and moxibustion were feasible interventions for fatigue. This study is a pilot patient-assessor-blinded randomized sham-controlled trial to evaluate the efficacy and safety of acupuncture treatment for patients with fatigue that has persisted for at least 4 weeks after recovery from COVID-19. Methods Thirty patients will be recruited and randomly assigned to either the acupuncture or sham acupuncture treatment groups. Treatment will be conducted thrice a week for both groups during 4 weeks. The primary outcome will be the efficacy and safety of acupuncture, including numeric rating scale (NRS), brief fatigue inventory (BFI), fatigue severity scale (FSS), and adverse event evaluation. Secondary outcomes will be evaluation of improvement in the comorbid symptoms of fatigue and feasibility variables. Outcome variables will be assessed before treatment, 4 weeks after treatment, and 8 weeks after treatment completion. Discussion The results of this study will be used to clarify the efficacy and safety of acupuncture treatment for persistent fatigue in patients with Long COVID. Additionally, the feasibility of the study design was validated to provide evidence for future full-scale randomized controlled trials.Clinical trial registration: identifier: KCT0008656 https://cris.nih.go.kr/cris/search/detailSearch.do?seq=24785&search_page=L.
Collapse
Affiliation(s)
- Sung-A. Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ji-Su Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Taegon Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Tae-Hun Kim
- Korean Medicine Clinical Trial Center, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sunoh Kwon
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Jung Won Kang
- Department of Acupuncture & Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Acupuncture and Moxibustion, Kyung Hee University Medical Center, Seoul, Republic of Korea
| |
Collapse
|
29
|
Soprano CM, Ngo R, Konys CA, Bazier A, Salamon KS. Post-Acute Sequelae of COVID-19 (PASC) in Pediatrics: Factors That Impact Symptom Severity and Referral to Treatment. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1805. [PMID: 38002896 PMCID: PMC10670189 DOI: 10.3390/children10111805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023]
Abstract
The post-acute sequelae of COVID-19 (PASC) is a complex condition. While there are emerging studies on its effects in adults, there is scarce research regarding the long-term effects of COVID-19 infection among youth. Several researchers have likened long-haul COVID-19 to chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME) and postural orthostatic tachycardia syndrome (POTS). In adults, the prognosis for these diagnoses is less promising than that in youth; however, there is currently very little information available on the presentation of youth with PASC. A better understanding of the specific symptom presentation for youth diagnosed with PASC is necessary. Retrospective chart reviews were conducted collecting demographic data, COVID-19 symptoms and disease progression, and vaccination status. Additional data on referrals to a PASC treatment program and appointments attended were collected. Overall, data suggested that youth present with less severe PASC symptoms than adults, and the role of vaccination is unclear. These youth are often not referred to treatment programs. More exploration is necessary to continue to build an understanding of how best to aid youth diagnosed with PASC.
Collapse
Affiliation(s)
- Catherine M. Soprano
- Nemours Children’s Health, Wilmington, DE 19803, USA;
- Sydney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ryan Ngo
- Bowdoin College, Brunswick, ME 04011, USA
| | - Casey A. Konys
- Lurie Children’s Hospital of Chicago, Chicago, IL 60614, USA
| | - Ashley Bazier
- School of Medicine, Indiana University, Indianapolis, IN 46805, USA
| | - Katherine S. Salamon
- Nemours Children’s Health, Wilmington, DE 19803, USA;
- Sydney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
30
|
ACTIV-6: Operationalizing a decentralized, outpatient randomized platform trial to evaluate efficacy of repurposed medicines for COVID-19. J Clin Transl Sci 2023; 7:e221. [PMID: 38028354 PMCID: PMC10643936 DOI: 10.1017/cts.2023.644] [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: 06/12/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Despite the availability of vaccinations, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to cause Coronavirus Disease 2019 (COVID-19) infection with a spectrum of disease in the acute setting. Transmission, infection, and severe disease remain common. There is a critical need to establish treatment regimens in the ambulatory setting that can reduce symptom burden and potentially prevent progression to severe disease and death. Many existing medicines previously approved for other uses may have benefit but remain unproven in informative clinical trials. Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV)-6 is a decentralized, placebo-controlled, double-blind, randomized, platform trial that has now enrolled more than 7500 participants and has reported on the effectiveness of ivermectin at two doses, fluticasone, and fluvoxamine for helping people with COVID-19. With additional repurposed therapies added to the platform, ACTIV-6 continues to enroll symptomatic outpatients aged ≥ 30 years with a confirmed positive PCR or antigen test for SARS-CoV-2. Potential participants are screened and enrolled online, through a call center, or facilitated by local study sites. Participants consent electronically and are randomized to placebo or to one of the open study drugs for which they are eligible at the time of enrollment. A shared, contemporary placebo approach is used. Participants receive study drug in the mail and remain on study for up to 180 days. While enrolled, electronic patient-reported outcome assessments are used to monitor symptoms, healthcare utilization, and mortality. The primary endpoint is time to recovery or a composite of hospitalization and mortality within 28 days. Symptoms, acute healthcare utilization, and the Patient-Reported Outcomes Measurement Information System-29 are collected for up to 180 days. Using a decentralized trial approach allowed the ACTIV-6 platform to increase both reach and rate of enrollment. The decentralized approach did not simplify regulatory oversight, and we found unanticipated challenges in patient behavior and the study drug delivery process. Despite challenges, ACTIV-6 has enrolled thousands of participants from across the USA and continues to test the effectiveness of repurposed medicines for treating COVID-19. Our lessons learned contribute to the emerging understanding of how to optimize decentralized trials.
Collapse
|
31
|
Martelletti P, Leonardi M, Ashina M, Burstein R, Cho SJ, Charway-Felli A, Dodick DW, Gil-Gouveia R, Grazzi L, Lampl C, MaassenVanDenBrink A, Minen MT, Mitsikostas DD, Olesen J, Owolabi MO, Reuter U, Ruiz de la Torre E, Sacco S, Schwedt TJ, Serafini G, Surya N, Tassorelli C, Wang SJ, Wang Y, Wijeratne T, Raggi A. Rethinking headache as a global public health case model for reaching the SDG 3 HEALTH by 2030. J Headache Pain 2023; 24:140. [PMID: 37884869 PMCID: PMC10604921 DOI: 10.1186/s10194-023-01666-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/05/2023] [Indexed: 10/28/2023] Open
Abstract
The 2030 Agenda for Sustainable Development sets out, through 17 Sustainable Development Goals (SDGs), a path for the prosperity of people and the planet. SDG 3 in particular aims to ensure healthy lives and promote well-being for all at all ages and includes several targets to enhance health. This review presents a "headache-tailored" perspective on how to achieve SDG 3 by focusing on six specific actions: targeting chronic headaches; reducing the overuse of acute pain-relieving medications; promoting the education of healthcare professionals; granting access to medication in low- and middle-income countries (LMIC); implementing training and educational opportunities for healthcare professionals in low and middle income countries; building a global alliance against headache disorders. Addressing the burden of headache disorders directly impacts on populations' health, as well as on the possibility to improve the productivity of people aged below 50, women in particular. Our analysis pointed out several elements, and included: moving forward from frequency-based parameters to define headache severity; recognizing and managing comorbid diseases and risk factors; implementing a disease management multi-modal management model that incorporates pharmacological and non-pharmacological treatments; early recognizing and managing the overuse of acute pain-relieving medications; promoting undergraduate, postgraduate, and continuing medical education of healthcare professionals with specific training on headache; and promoting a culture that favors the recognition of headaches as diseases with a neurobiological basis, where this is not yet recognized. Making headache care more sustainable is an achievable objective, which will require multi-stakeholder collaborations across all sectors of society, both health-related and not health-related. Robust investments will be needed; however, considering the high prevalence of headache disorders and the associated disability, these investments will surely improve multiple health outcomes and lift development and well-being globally.
Collapse
Affiliation(s)
- Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Matilde Leonardi
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy
| | - Messoud Ashina
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rami Burstein
- John Hedley-Whyte Professor of Anesthesia and Neuroscience at the Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Soo-Jin Cho
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Military Hospital, Hwaseong, Korea
| | | | - David W Dodick
- Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | - Raquel Gil-Gouveia
- Neurology Department, Hospital da Luz Headache Center, Hospital da Luz Lisboa., Lisbon, Portugal
- Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Lisbon, Portugal
| | - Licia Grazzi
- Neuroalgology Unit and Headache Center, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Christian Lampl
- Department of Neurology and Stroke Unit, Koventhospital Barmherzige Brüder Linz, Linz, Austria
- Headache Medical Center Linz, Linz, Austria
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Mia T Minen
- Department of Neurology, NYU Langone Health, NY, New York, USA
| | - Dimos Dimitrios Mitsikostas
- 1st Neurology Department, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Jes Olesen
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mayowa Ojo Owolabi
- Faculty of Clinical Sciences; Center for Genomic and Precision Medicine, College of Medicine,, University of Ibadan, Ibadan, Nigeria
| | - Uwe Reuter
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Universitätsmedizin Greifswald, Greifswald, Germany
| | | | - Simona Sacco
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Todd J Schwedt
- Department of Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | - Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Cristina Tassorelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Headache Science and Neurorehabilitation Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Shuu-Jiun Wang
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Neurology, The Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yonggang Wang
- Headache Center, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tissa Wijeratne
- Department of Neurology, Sunshine Hospital, St Albans, VIC, Australia
| | - Alberto Raggi
- Neurology, Public Health and Disability Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan, 20133, Italy.
| |
Collapse
|
32
|
Sahnoun I, Znegui T, Moussa I, Rejeb S, Ayedi Y, Hattab S, Mokaddem S, Jameleddine S, El Gharbi LD. Impact of respiratory muscle training on clinical and functional parameters in COVID-19 recovered patients. Pan Afr Med J 2023; 46:65. [PMID: 38282784 PMCID: PMC10822100 DOI: 10.11604/pamj.2023.46.65.36615] [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: 08/02/2022] [Accepted: 03/08/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction early respiratory rehabilitation is required for patients with coronavirus virus disease 2019 (COVID-19) sequelae to reduce the risk of serious disabilities after hospital discharge. Methods it was a comparative prospective study including patients with persistent symptoms one month after discharge. The patients were hospitalized at the pneumology department D of Abderahman Mami hospital for COVID-19 pneumonia. The study involved two groups: (G1) included patients who participated in respiratory muscle training program (twice a week during 6 weeks), and a control group (G2). The groups were matched based on age, sex and body mass index (BMI). Persistent symptoms and pulmonary lung function (forced vital capacity (FVC), forced expiratory volume in one second (FEV1), total lung capacity (TLC) and diffusion capacity for carbon monoxide (DLCO), maximal inspiratory pressure (PI max) and maximal expiratory pressure (PE max), 6 Minute Walk distance (6-MWD) at baseline and after 6 weeks were compared between the two groups. Results the two groups of patients were comparable in terms of age, sex, BMI, comorbidities, and extent of lung computed tomography (CT) lesions. Compared to G2, a significant improvement of persistent symptoms was noted in G1, including dry cough (p=0.002), dyspnea (p=0.001), chest pain (p=0.002), and fatigue (p=0.001). The mean of percutaneous oxygen saturation (SpO2) increased from 96.68% to 97.93% (p<0.01) in G1. A significant improvement in the percentages of change of FEV1 (p=0.005), FVC (p=0.003), TLC (p<0.001), DLCO (p<0.001), and 6-MWD (p=0.015) was also noted in G1 after this program. Nevertheless, only the percentage of FEV1 (p=0.02) increased in the control group. No impact of respiratory muscle training on PI max and PE max was noted. Conclusion the present study demonstrated a significant improvement of persistent symptoms and exercise tolerance after short-term respiratory muscle training in patients suffering from COVID-19 sequelae.
Collapse
Affiliation(s)
- Imen Sahnoun
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Tasnim Znegui
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Ines Moussa
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Siwar Rejeb
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Yosor Ayedi
- University of Tunis El Manar, Tunis Medical School, Epidemiology and Statistics Department, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Sirine Hattab
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Salma Mokaddem
- University of Tunis El Manar, Tunis Medical School, Department of Physiology, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Saloua Jameleddine
- University of Tunis El Manar, Tunis Medical School, Department of Physiology, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Leila Douik El Gharbi
- University of Tunis El Manar, Tunis Medical School, Pneumology Department D, Abderrahmen Mami Hospital, Ariana, Tunisia
| |
Collapse
|
33
|
Hasan A, Rahim R, Nakayama EE, Uno K, Hasan N, Rahman M, Shioda T. Enhancement of IL-6 Production Induced by SARS-CoV-2 Nucleocapsid Protein and Bangladeshi COVID-19 Patients' Sera. Viruses 2023; 15:2018. [PMID: 37896795 PMCID: PMC10611338 DOI: 10.3390/v15102018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory tract infection caused by severe acute respiratory syndrome coronavirus 2 that can have detrimental effects on multiple organs and accelerate patient mortality. This study, which encompassed 130 confirmed COVID-19 patients who were assessed at three different time points (i.e., 3, 7, and 12 days) after the onset of symptoms, investigated interleukin-6 (IL-6) enhancement induced by a viral nucleocapsid (N) protein from a myeloid cell line. Disease severity was categorized as mild, moderate, or severe. The severe cases were characterized as having significant elevations in serum IL-6, C-reactive protein, D-dimer, ferritin, creatinine, leukocytes, and neutrophil-to-lymphocyte ratio and decreased hemoglobin, hematocrit, and albumin levels compared with mild and moderate cases. To evaluate IL-6-inducing activity, heat-inactivated sera from these patients were incubated with and without the N protein. The findings showed a progressive increase in IL-6 production in severe cases upon N protein stimulation. There was a strong correlation between anti-N antibodies and levels of IL-6 secreted by myeloid cells in the presence of N protein and sera, indicating the crucial role that the anti-N antibody plays in inducing IL-6 production. Uncontrolled IL-6 production played a pivotal role in disease pathogenesis, exacerbating both disease severity and mortality. Efficiently targeting the N protein could potentially be employed as a therapeutic strategy for regulating the immune response and alleviating inflammation in severe cases.
Collapse
Affiliation(s)
- Abu Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Rummana Rahim
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Emi E. Nakayama
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| | - Kazuko Uno
- IFN & Host-Defense Research Laboratory, Louis Pasteur Center for Medical Research, Kyoto 606-8225, Japan;
| | - Nazmul Hasan
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Mizanur Rahman
- Evercare Hospital Dhaka, Plot-81, Block-E, Bashundhara R/A, Dhaka 1229, Bangladesh; (A.H.); (R.R.); (N.H.)
| | - Tatsuo Shioda
- Research Institute for Microbial Diseases, Osaka University, Suita 565-0781, Japan;
| |
Collapse
|
34
|
Price JK, de Avila L, Stepanova M, Weinstein AA, Pham H, Keo W, Racila A, Gerber S, Lam BP, Gerber LH, Younossi ZM. Severe, Persistent, Disruptive Fatigue Post-SARS-CoV-2 Disproportionately Affects Young Women. Int J Gen Med 2023; 16:4393-4404. [PMID: 37789881 PMCID: PMC10544141 DOI: 10.2147/ijgm.s423910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/14/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction Post-acute SARS-CoV-2 (PASC) symptoms are often persistent, disruptive, and difficult to treat effectively. Fatigue is often among the most frequently reported symptoms and may indicate a more challenging road to recovery. Purpose To describe the natural history, symptomology, and risk profile of long-term post-acute SARS-CoV-2. Patients and Methods Participants treated for SARS-CoV-2 within a large, community health system in the US were enrolled prospectively in a longitudinal, observational PASC study examining participants at enrollment and 6 months. Medical history, symptom reporting, validated measures of cognition, and patient-reported outcomes (PROs), were performed for all participants and repeated during study follow-up visits. Results A total of 323 participants completed baseline evaluations. Sixty one participants indicated clinically significant fatigue (23.1% at baseline); a representative sample of 141 enrollees also completed a baseline Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) in-depth fatigue reporting questionnaire, 37 had severe fatigue. The severely fatigued (FACIT-F ≤29.7) were significantly younger, female, had more anxiety and depression, had a higher resting heart rate, reported more sick days, and were less physically active post-COVID. They were more likely to have a diagnosis of chronic kidney disease (13.5% vs 2.9%) but less likely to have a history of cancer (8.1% vs 23.1). Participants who were severely fatigued reported health, diet, weight, and sleep were worse than those not severely fatigued post-COVID (p = 0.02 to 0.0002). Fatigue was significantly correlated with impairment of all PROs administered after COVID-19 infection. Conclusion Fatigue is a common symptom post-COVID-19 infection and is associated with lower reported well-being and function. Those with severe fatigue tended to be younger and female and have a past medical history of anxiety, depression, kidney disease, and more sedentary lifestyles.
Collapse
Affiliation(s)
- Jillian Kallman Price
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Leyla de Avila
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Maria Stepanova
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| | - Ali A Weinstein
- Department of Global and Community Health, George Mason University, Fairfax, VA, USA
| | - Huong Pham
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Wisna’odom Keo
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Andrei Racila
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Suzannah Gerber
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
| | - Brian P Lam
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| | - Lynn H Gerber
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| | - Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| |
Collapse
|
35
|
Kopplin N, Garcia A, Reczek A, Wilkinson K, Yekkaluri S, Murphy CC, Tiro J, Muthukumar AR, Masica A, Singal AG. Post-acute sequelae of COVID-19 and longitudinal antibody levels in a community-based cohort. PLoS One 2023; 18:e0291259. [PMID: 37682916 PMCID: PMC10490864 DOI: 10.1371/journal.pone.0291259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) infection invokes variable immune responses and poses a risk of post-acute sequelae SARS-CoV-2 infection (PASC) symptoms; however, most data on natural history are derived from patients with severe infection. Further data are needed among patients with mild infection, who comprise most cases. METHODS The Dallas Fort-Worth (DFW) COVID-19 Prevalence Study included 21,597 community-dwelling adults (ages 18-89) who underwent COVID-19 PCR and anti-nucleocapsid antibody testing between July 2020 and March 2021. We invited participants with positive COVID-19 results (cases) and a subset with negative results (controls), matched on age, sex, race/ethnicity, and ZIP code, to complete a follow-up questionnaire for PASC symptoms and repeat anti-nucleocapsid testing, and anti-spike antibody testing between July and December 2021. RESULTS Of 3,917 adults invited to participate, 2260 (57.7%) completed the questionnaire- 1150 cases and 1110 controls. Persistent symptoms were reported in 21.1% of cases, with the most common being shortness of breath, fatigue, and loss of taste or smell. Among 292 cases with asymptomatic infection, >15% reported new fatigue and 8-10% reported new loss of taste/smell, myalgias, or headache. Median anti-nucleocapsid levels in cases decreased from 3.5U to 0.7U over a median follow-up of 8.6 months. Anti-spike antibody levels at 6-7 months post-vaccination in cases were similar to that of controls. CONCLUSIONS More than 1 in 5 patients with COVID-19 infection, including those with mild infection, reported persistent symptoms during follow-up. Both nucleocapsid and spike protein antibody levels decreased within six months following a COVID-19 infection and vaccination.
Collapse
Affiliation(s)
- Noa Kopplin
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Angie Garcia
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Annika Reczek
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Kate Wilkinson
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Sruthi Yekkaluri
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Caitlin C. Murphy
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Jasmin Tiro
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Alagar R. Muthukumar
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Andrew Masica
- Texas Health Resources, Fort Worth, TX, United States of America
| | - Amit G. Singal
- University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| |
Collapse
|
36
|
Torres-Ruiz J, Lomelín-Gascón J, Lira Luna J, Vargas-Castro AS, Pérez-Fragoso A, Nuñez-Aguirre M, Alcalá-Carmona B, Absalón-Aguilar A, Balderas-Miranda JT, Maravillas-Montero JL, Mejía-Domínguez NR, Núñez-Álvarez C, Llorente L, Romero-Ramírez S, Sosa-Hernández VA, Cervantes-Díaz R, Juárez-Vega G, Meza-Sánchez D, Rull-Gabayet M, Martínez-Juárez LA, Morales L, López-López LN, Negrete-Trujillo JA, Falcón-Lezama JA, Valdez-Vázquez RR, Gallardo-Rincón H, Tapia-Conyer R, Gómez-Martín D. Novel clinical and immunological features associated with persistent post-acute sequelae of COVID-19 after six months of follow-up: a pilot study. Infect Dis (Lond) 2023; 55:243-254. [PMID: 36637466 DOI: 10.1080/23744235.2022.2158217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Currently, there is scant information regarding the features associated to the persistence of post-COVID-19 syndrome, which is the main aim of the present study. METHODS A cohort study of 102 COVID-19 patients was conducted. The post-COVID-19 symptoms were assessed by a standardised questionnaire. Lymphocyte immunophenotyping was performed by flow cytometry and chemokines/cytokines, neutrophil extracellular traps, the tripartite motif 63, anti-cellular, and anti-SARS-CoV-2 IgG antibodies were addressed in serum. The primary outcome was the persistence of post-COVID-19 syndrome after six months follow-up. RESULTS Thirteen patients (12.7%) developed the primary outcome and had a more frequent history of post-COVID-19 syndrome 3 months after infection onset (p = .044), increased levels of IL-1α (p = .011) and IP-10 (p = .037) and increased CD57 expression in CD8+ T cells (p = .003). There was a trend towards higher levels of IFN-γ (p = .051), IL-1β (p = .062) and IL-6 (p = .087). The history of post COVID-19 in the previous 3 months, obesity, baseline serum MIP-1α and IP-10, and CD57 expression in CD8+ T cells were independently associated with the persistence of post-COVID-19 syndrome. CONCLUSION Our data suggest an important relationship between a pro-inflammatory state mediated through metabolic pathways related to obesity and increased cellular senescence as a key element in the persistence of post-COVID-19 syndrome at six months of follow-up.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jaquelin Lira Luna
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Sección de Estudios de Posgrado e Investigación Escuela Superior de Medicina-IPN, Mexico City, Mexico
| | - Ana Sofia Vargas-Castro
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Becaria o Becario de la Dirección General de Calidad y Educación en Salud, Secretaría de Salud, México
| | - Alfredo Pérez-Fragoso
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Miroslava Nuñez-Aguirre
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Beatriz Alcalá-Carmona
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Químicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional
| | - Abdiel Absalón-Aguilar
- Department of Internal Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jennifer T Balderas-Miranda
- Program of Combined Studies in Medicine, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis Llorente
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Victor Andrés Sosa-Hernández
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David Meza-Sánchez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Marina Rull-Gabayet
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Linda Morales
- Carlos Slim Foundation, Mexico City, Mexico
- Temporary COVID-19 Hospital, Hipódromo de las Américas, Mexico City, Mexico
| | | | | | - Jorge Abelardo Falcón-Lezama
- Carlos Slim Foundation, Mexico City, Mexico
- División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco
| | | | - Héctor Gallardo-Rincón
- Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara, Mexico
| | - Roberto Tapia-Conyer
- National Autonomous University of Mexico, School of Medicine, Mexico City, Mexico
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
37
|
Bozkurt N, Yalcin AN. Prevention of Scarring of Lung Parenchyma in COVID-19 Patients through Glucocorticoid Treatment. Am J Health Behav 2023; 47:182-193. [PMID: 36945083 DOI: 10.5993/ajhb.47.1.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Objective: The objective of this research was to determine the impact of glucocorticoid treatment on preventing scarring of lung parenchyma in COVID-19 patients by considering their health behavior. Methods: A sample of 65 Turkish patients who had pneumonia and were hospitalized between March/December 2020 were included in this research. The data for this research was collected after the consent of the hospital. The structural equation model approach was used in data analysis and empirical findings. Results: The research identified that the patients with appropriate health behavior were satisfied with their clinical treatment of scarring of lung parenchyma by the method of glucocorticoid treatment. The research also identified that the patient's health behavior was a significant indicator to improve their perception of the clinical treatment. Conclusion: This study has reliable theoretical implications that are significantly important in the literature because of the nature and uniqueness of the findings. However, this research also has some practical implications related to the patient's lungs mostly infected by COVID-19. Furthermore, the findings of this research can be generalized in a significant way because the respondents of this research belonged to a diverse population.
Collapse
Affiliation(s)
- Nurgul Bozkurt
- Akdeniz University Medical Faculty Department of Chest Diseases Pınarbaşı Mah. Dumlupınar Bulvarı Akdeniz Üniversitesi Hastanesi 07059, Kampüs, Konyaaltı/Antalya/Turkey;,
| | - Ata N Yalcin
- Akdeniz University Medical Faculty Department of Clinic Microbiology. Pınarbaşı Mah. Dumlupınar Bulvarı Akdeniz Üniversitesi Hastanesi 07059, Kampüs, Konyaaltı/Antalya/Turkey
| |
Collapse
|
38
|
Neurological Consequences of Pulmonary Emboli in COVID-19 Patients: A Study of Incidence and Outcomes in the Kingdom of Saudi Arabia. Brain Sci 2023; 13:brainsci13020343. [PMID: 36831886 PMCID: PMC9954757 DOI: 10.3390/brainsci13020343] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Pulmonary embolism (PE) is a significant consequence that is becoming more common in COVID-19 patients. The current study sought to determine the prevalence and risk factors for PE in a study population of COVID-19 patients, as well as the relationship between PE and neurological sequelae. The research also sought to analyze the consistency of neurological examination and imaging techniques in detecting neurological problems. The research comprised a total of 63 individuals with COVID-19. The incidence of PE in the study group was 9.5% for smokers, 23.8% for obese patients, 33.3% for hypertensive patients, and 19% for diabetic patients, according to the findings. After adjusting for possible confounders such as age, gender, BMI, smoking, hypertension, and diabetes, a logistic regression analysis indicated that the probabilities of having neurological complications were 3.5 times greater in individuals who had PE. In conclusion, the present study highlights the high incidence of PE among patients with COVID-19 and the association between PE and neurological complications. The study also emphasizes the importance of a thorough neurological examination and imaging studies in the detection of neurological complications in patients with PE.
Collapse
|
39
|
Munipalli B, Paul S, Mohabbat A, Siddiqui H, Li Z, Abu Dabrh AM. Clinical differences in symptomology, characteristics, and risk factors in patients with post-acute sequelae of COVID-19: an experience from a tertiary-care academic center. J Investig Med 2023; 71:495-501. [PMID: 36760093 PMCID: PMC9922663 DOI: 10.1177/10815589231153604] [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] [Indexed: 02/11/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by the novel coronavirus SARS-CoV-2 and has caused significant mortality and morbidity since it was first recognized in Wuhan, China in December 2019. Patients may suffer from a constellation of symptoms termed post-acute sequelae of COVID-19 (PASC) . Here we present findings of a retrospective cohort study describing the prevalence and predicting factors of patient-reported post-acute sequelae of COVID-19 (PASC). Categorical variables were summarized as frequency (percentage) and compared between vaccine status groups using Fisher's exact test. Continuous variables were reported as median (range) and compared between the groups using Kruskal-Wallis test. All tests were two-sided with p value <0.05 considered statistically significant. Survey data from 132 patients with a median age of 45 years, 68% female, 83% Caucasian/Non-Hispanic. The most frequently reported PASC symptoms include fatigue (84.8%), dyspnea (54.5%), cognitive dysfunction (53%), myalgias (37.1%), lightheadedness or vertigo (36.4%), chest pain (34.8%), palpitations (34.8%), headaches (34.1%), arthralgias (31.8%), and unrefreshing sleep (31.1%). There is mounting evidence that supports higher prevalence of PASC in women, White/Caucasian, and middle-aged individuals. This knowledge can provide guidance to clinical practices to anticipate and support healthcare and self-care needs for patients at higher risk to developing PASC.
Collapse
Affiliation(s)
- Bala Munipalli
- Division of General Internal Medicine,
Mayo Clinic Florida, Jacksonville, FL, USA,Bala Munipalli, Division of General
Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL
32224, USA.
| | - Stefan Paul
- Division of General Internal Medicine,
Mayo Clinic Florida, Jacksonville, FL, USA
| | - Arya Mohabbat
- Division of General Internal Medicine,
Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Habeeba Siddiqui
- Department of Quantitative Health
Sciences, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Zhuo Li
- Department of Quantitative Health
Sciences, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Abd Moain Abu Dabrh
- Division of General Internal Medicine,
Mayo Clinic Florida, Jacksonville, FL, USA
| |
Collapse
|
40
|
Chiang M, Shih L, Lu C, Fang S. The COVID-19 vaccine did not affect the basal immune response and menstruation in female athletes. Physiol Rep 2023; 11:e15556. [PMID: 36750121 PMCID: PMC9904960 DOI: 10.14814/phy2.15556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 06/18/2023] Open
Abstract
The COVID-19 pandemic restricted the regular training and competition program of athletes. Vaccines against COVID-19 are known to be beneficial for the disease; however, the unknown side effects of vaccines and postvaccination reactions have made some athletes hesitant to get vaccinated. We investigated the changes in inflammatory responses and menstrual cycles of female athletes before and after vaccination. Twenty female athletes were enrolled in this study. Blood was collected from each subject before the first COVID-19 vaccination and after the first and second vaccinations. Laboratory data, including white blood cell, neutrophil, lymphocyte, and platelet counts, and inflammatory markers, including NLR (neutrophil-to-lymphocyte ratio), PLR (platelet lymphocyte ratio), RPR (red cell distribution width to platelet ratio), SII (systemic immune-inflammation index), and NeuPla (neutrophil-platelet ratio), were analyzed statistically. The menstrual changes before and after vaccination and the side effects were collected by questionnaires. No significant changes in the laboratory data were found after the first and second shots when compared to those at prevaccination: white blood cell, neutrophil, lymphocyte, platelet, NLR, PLR, SII, RPR, and NeuPla (p > 0.05). In addition, there were no significant changes in the menstruation cycle or days of the menstrual period (p > 0.05). All side effects after vaccination were mild and subsided in 2 days. The blood cell counts, inflammatory markers, and menstruation of female athletes were not affected by COVID-19 vaccines.
Collapse
Affiliation(s)
- Ming‐Ru Chiang
- Department of PediatricsJen‐Ai HospitalTaichungTaiwan
- Department of Exercise Health ScienceNational Taiwan University of SportTaichungTaiwan
| | - Li‐Chun Shih
- Department of Obstetrics and Gynecology, Puli BranchTaichung Veterans General HospitalNantouTaiwan
| | - Chi‐Cheng Lu
- Institute of AthleticsNational Taiwan University of SportTaichungTaiwan
| | - Shih‐Hua Fang
- Institute of AthleticsNational Taiwan University of SportTaichungTaiwan
| |
Collapse
|
41
|
Dong J, Varbanov M, Philippot S, Vreken F, Zeng WB, Blay V. Ligand-based discovery of coronavirus main protease inhibitors using MACAW molecular embeddings. J Enzyme Inhib Med Chem 2023; 38:24-35. [PMID: 36305272 PMCID: PMC9621234 DOI: 10.1080/14756366.2022.2132486] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ligand-based drug design methods are thought to require large experimental datasets to become useful for virtual screening. In this work, we propose a computational strategy to design novel inhibitors of coronavirus main protease, Mpro. The pipeline integrates publicly available screening and binding affinity data in a two-stage machine-learning model using the recent MACAW embeddings. Once trained, the model can be deployed to rapidly screen large libraries of molecules in silico. Several hundred thousand compounds were virtually screened and 10 of them were selected for experimental testing. From these 10 compounds, 8 showed a clear inhibitory effect on recombinant Mpro, with half-maximal inhibitory concentration values (IC50) in the range 0.18–18.82 μM. Cellular assays were also conducted to evaluate cytotoxic, haemolytic, and antiviral properties. A promising lead compound against coronavirus Mpro was identified with dose-dependent inhibition of virus infectivity and minimal toxicity on human MRC-5 cells.
Collapse
Affiliation(s)
- Jie Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, P. R. China
| | - Mihayl Varbanov
- Université de Lorraine, CNRS, Nancy, France
- Laboratoire de Virologie, CHRU de Nancy Brabois, Vandoeuvre-lès-Nancy, France
| | | | | | - Wen-bin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, P. R. China
| | - Vincent Blay
- Department of Microbiology and Environmental Toxicology, University of California at Santa Cruz, Santa Cruz, CA, USA
| |
Collapse
|