1
|
Imtiaz N, Stoddard WA, Ghazy A, Day SW. Oxygen transport in nanoporous SiN membrane compared to PDMS and polypropylene for microfluidic ECMO. Biomed Microdevices 2025; 27:22. [PMID: 40437284 PMCID: PMC12119709 DOI: 10.1007/s10544-025-00750-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2025] [Indexed: 06/01/2025]
Abstract
Extracorporeal Membrane Oxygenation (ECMO) serves as a crucial intervention for patients with severe pulmonary dysfunction by facilitating oxygenation and carbon dioxide removal. While traditional ECMO systems are effective, their large priming volumes and significant blood-contacting surface areas can lead to complications, particularly in neonates and pediatric patients. Microfluidic ECMO systems offer a promising alternative by miniaturizing the ECMO technology, reducing blood volume requirements, and minimizing device surface area to improve safety and efficiency. This study investigates the oxygen transport performance of three membrane types- polydimethylsiloxane (PDMS), polypropylene, and a novel nanoporous silicon nitride (NPSiN) membrane-in a microfluidic ECMO platform. While nanoporous membranes rely on pore-mediated diffusion and PDMS on polymer lattice diffusion, results show no significant differences in device oxygenation efficiency (p > 0.05). Blood-side factors, including the diffusion rate of oxygen through the red blood cell (RBC) membrane, RBC residence time, and hemoglobin binding kinetics, were identified as primary bottlenecks. Even computational models of a hypothetical infinitely permeable membrane corroborate the limited impact of membrane material. These findings suggest a shift in ECMO design priorities from membrane material to blood-side enhancements. This research provides a foundation for optimizing ECMO systems.
Collapse
Affiliation(s)
- Nayeem Imtiaz
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY, 14623, USA
| | - William A Stoddard
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY, 14623, USA
| | - Abdelrahman Ghazy
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY, 14623, USA
| | - Steven W Day
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY, 14623, USA.
| |
Collapse
|
2
|
Kaneshita S, Chambers CD, Johnson D, Kavanaugh A, Garfein R, Bandoli G. Short-term side effects following COVID-19 vaccination in pregnancies complicated by autoimmune inflammatory rheumatic diseases: A prospective cohort study. Vaccine 2025; 56:127194. [PMID: 40315794 DOI: 10.1016/j.vaccine.2025.127194] [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: 02/25/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Pregnant women are at higher risk for severe Coronavirus disease 2019 (COVID-19) infection, leading the Centers for Disease Control and Prevention to recommend vaccination. However, the frequency of vaccine side effects in pregnant women with autoimmune inflammatory rheumatic diseases (AIIRD) remains unknown. We investigated the frequencies of short-term (≤ 7 days) adverse reactions following the initial COVID-19 vaccination ever received by pregnant women with AIIRD. METHODS A descriptive analysis of the incidence of side effects following the COVID-19 vaccination in pregnant women with AIIRD was conducted utilizing data from the MotherToBaby Study registry, a prospective cohort study designed to assess the safety of drug and vaccine exposure during pregnancy. This study enrolled pregnant women living in the United States or Canada between January 2021 and September 2022. RESULTS Of 1413 participants who received dose 1 of the COVID-19 vaccine in pregnancy, 79 had AIIRD. There was no large difference in the total number of adverse reactions between pregnant women with and without AIIRD (β = -0.01, 95 % Confidence Interval [CI]: -0.17, 0.17). Pregnant women with higher Health Assessment Questionnaire Disability Index (HAQ-DI) in AIIRD had a higher total number of systemic reactions (β = 0.56, 95 % CI: 0.04, 1.10) than those with lower HAQ-DI, although the total number of adverse reactions was not different (β = 0.27, 95 % CI: -0.08, 0.62). CONCLUSIONS We found no difference in the frequency of COVID-19 vaccine-related side effects between pregnant women with and without AIIRD. Patients with AIIRD who have higher levels of functional impairment may have a slightly higher frequency of short-term adverse effects. The results of this assessment may help provide information for pregnant women with AIIRD regarding COVID-19 vaccination.
Collapse
Affiliation(s)
- Shunya Kaneshita
- Herbert Wertheim School of Public Health and Longevity Science, University of California San Diego, La Jolla, CA, USA; Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Christina D Chambers
- Herbert Wertheim School of Public Health and Longevity Science, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, Division of Environmental Science and Health, University of California San Diego, La Jolla, CA, USA
| | - Diana Johnson
- Department of Pediatrics, Division of Environmental Science and Health, University of California San Diego, La Jolla, CA, USA
| | - Arthur Kavanaugh
- Division of Rheumatology, Autoimmunity and Inflammation, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Richard Garfein
- Herbert Wertheim School of Public Health and Longevity Science, University of California San Diego, La Jolla, CA, USA
| | - Gretchen Bandoli
- Herbert Wertheim School of Public Health and Longevity Science, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, Division of Environmental Science and Health, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
3
|
Zhang Y, Wu S, Li X. Expression and diagnostic values of ferroptosis-related genes in coronavirus-associated viral sepsis. Front Med (Lausanne) 2025; 12:1496834. [PMID: 40370730 PMCID: PMC12074931 DOI: 10.3389/fmed.2025.1496834] [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: 09/15/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Aim The aim of this study is to investigate the differential expression and diagnostic value of ferroptosis-related genes in coronavirus-associated viral sepsis. Methods This study was conducted in two sequential phases: (1) identification of differentially expressed genes through comprehensive analysis of the experimental dataset (GSE164805); and (2) clinical validation of the candidate molecular markers using both test set samples and clinical samples, followed by rigorous evaluation of their diagnostic performance. Firstly, the microchips associated with coronavirus-associated viral sepsis were retrieved from the GEO database, a public data platform of NCBI (National Center for Biotechnology Information), and differentially expressed genes (DEGs) were obtained through differential analysis. The identified DEGs were then intersected with the ferroptosis gene dataset to obtain ferroptosis-related DEGs. Subsequently, molecular signaling pathways of ferroptosis-related genes in coronavirus-associated viral sepsis were analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. CIBERSORT was employed to analyze immune cell infiltration in both the coronavirus-associated viral sepsis group and control group. Furthermore, a protein-protein interaction (PPI) network was constructed to identify hub genes involved in ferroptosis. Finally, the expression of ferroptosis hub genes in coronavirus-associated viral sepsis and its diagnostic value were analyzed in validation set GSE199816 and clinical case samples. Results In test set GSE164805, a total of 15,059 differentially expressed genes (DEGs) were identified, comprising 7,458 up-regulated and 7,601 down-regulated genes. Subsequently, an intersection analysis with the ferroptosis gene dataset yielded 189 DEGs associated with ferroptosis. Functional enrichment analyses using GO and KEGG revealed significant enrichment in signaling pathways related to ferroptosis, oxidative stress, and HIF-1. Additionally, CIBERSORT immune-infiltration analysis revealed enhanced infiltration of innate immune cells but reduced infiltration of CD8+ T cells and natural killer (NK) cells in the coronavirus-associated viral sepsis group compared with healthy controls. Furthermore, analysis identified that the distribution of these immune cells correlated with the expression levels of IL1-β and HMOX1, suggesting that viral infection in the septic pathological state disrupts the balance between immune activation and suppression. Notably, PPI network analysis also identified IL1-β and HMOX1 as hub genes involved in ferroptosis. Finally, the results were verified in the validation set and clinical case samples, and the results showed that the expressions of IL1-β and HMOX1 in the coronavirus-associated viral sepsis group were decreased compared with the case control group and the healthy control group. In clinical samples, the expression levels were as follows: IL1-β (0.390 ± 0.068 vs. 1.101 ± 0.107), HMOX1 (0.629 ± 0.117 vs. 1.101 ± 0.107), and the differences were statistically significant (all p < 0.05). Further diagnostic performance analysis demonstrated that IL1-β and HMOX1 exhibited AUROCs of 0.892 and 0.765, respectively, indicating their robust diagnostic potential for coronavirus-associated viral sepsis. Conclusion The present study has successfully identified two hub genes, IL1-β and HMOX1, associated with ferroptosis in coronavirus-associated viral sepsis, and their expression and diagnostic value for the disease. These findings provide effective diagnostic biomarkers and potential therapeutic targets for coronavirus-associated viral sepsis. Notably, this study specifically focused on coronavirus-induced viral sepsis, distinct from previously characterized bacterial sepsis and other viral etiologies, thus warranting future studies with expanded sample sizes for stratified analyses.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Respiratory and Critical Care Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Shaoduo Wu
- Department of Respiratory and Critical Care Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xiaoyan Li
- Department of Pulmonary Critical Care Medicine, Zhoupu Hospital in Pudong New Area, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
4
|
Jeon S, Kim YK, Seo GB, Oh GJ, Leem ST, Song J, Yang MJ, Hwang J, Kim MS. SARS-CoV-2 infection exacerbates fibrosis and develops new-onset asthma in damaged lung by polyhexamethylene guanidine phosphate. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138430. [PMID: 40319855 DOI: 10.1016/j.jhazmat.2025.138430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Modern society faces a variety of respiratory-related threats from the increased use of chemicals and periodic outbreaks of infectious diseases. This study investigates the connection between chemically-induced lung damage and SARS-CoV-2 infection, addressing a critical research gap. To investigate this connection, we conducted a study using a mouse model to assess SARS-CoV-2 infection symptoms in lungs injured by polyhexamethylene guanidine phosphate (PHMG-p). Our research revealed that PHMG-p-Induced Lung Injury (PILI) mice exhibited severe inflammatory responses and lung damage following infection. Cytokine storm-related factors were significantly elevated in the bronchoalveolar lavage fluids of infected PILI mice, indicating severe infection. RNA-seq analysis showed upregulated genes in infected PILI mice associated with respiratory tract diseases and increased inflammatory and immune responses. Downregulated genes were primarily involved in lipid metabolism processes. We also identified alterations in four _genes linked to asthma development in infected PILI mice, correlating with clinical observations in patients. Our findings suggest that SARS-CoV-2 infection in chemically damaged lungs may exacerbate symptoms and potentially lead to new-onset asthma. This study highlights the increased risk of infection severity in chemically damaged lungs and emphasizes the need for heightened awareness of respiratory health in individuals exposed to chemicals, especially during infectious disease outbreaks.
Collapse
Affiliation(s)
- Seulgi Jeon
- Center for Respiratory Safety Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Young Kyu Kim
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Gyun-Baek Seo
- Center for Respiratory Safety Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Gi Jun Oh
- Center for Respiratory Safety Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Sun-Taek Leem
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Jeongah Song
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Mi-Jin Yang
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea
| | - Jeongho Hwang
- Center for Large Animals Convergence Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea.
| | - Min-Seok Kim
- Center for Respiratory Safety Research, Korea Institute of Toxicology, Jeongeup-si, Jeonbuk-do 56212, Republic of Korea.
| |
Collapse
|
5
|
Wada H, Shiraki K, Ichikawa Y, Ito N, Inoue H, Moritani I, Masuda J, Yamamoto A, Tomida M, Yoshida M, Kawamura M, Shimaoka M, Iba T, Shimpo H. Implications of Soluble C-type Lectin-Like Receptor 2 Levels in Patients with Coronavirus Disease 2019-Associated with Thrombosis. Thromb Haemost 2025. [PMID: 40174637 DOI: 10.1055/a-2572-1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is often associated with thrombosis. Elevated levels of soluble C-type lectin-like receptor 2 (sCLEC-2), a biomarker for platelet activation, have been reported in COVID-19. Therefore, we examined the behavior of sCLEC-2 levels and their relationship with thrombosis.The clinical course of inflammatory and thrombotic biomarkers was assessed in 271 patients with COVID-19.Inflammatory biomarkers such as C-reactive protein, procalcitonin, and presepsin levels were significantly increased in patients with COVID-19, and these behaviors differed among the clinical course or stages. The plasma D-dimer levels increased slightly and gradually. Platelet counts were within the normal range, and plasma sCLEC-2 levels were markedly increased in most patients with COVID-19. There were 17 patients with thrombosis in this study. Although there was no significant difference in various biomarkers between COVID-19 patients with and without thrombosis, the super formula of sCLEC-2xD-dimer/platelet count in patients with thrombosis was significantly higher than in those without thrombosis. Furthermore, this super formula was significantly higher in COVID-19 patients with severe or critical illness than in those with mild or moderate illness.Elevation of the super formula of sCLEC-2xD-dimer/platelet count was associated with the thrombosis in patients with COVID-19 suggesting the thrombosis in COVID-19 may be caused by the development of microthrombosis.
Collapse
Affiliation(s)
- Hideo Wada
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Katsuya Shiraki
- Department of General Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Yuhuko Ichikawa
- Department of Central Laboratory, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Nobuo Ito
- Department of Neurology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Hidekazu Inoue
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Isao Moritani
- Department of Gastroenterology, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Jun Masuda
- Department of Cardiovascular Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Akitaka Yamamoto
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Masaki Tomida
- Department of Emergency and Critical Care Center, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Masamichi Yoshida
- Department of Respiratory Medicine, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | | | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Hideto Shimpo
- Mie Prefectural General Medical Center, Yokkaichi, Japan
| |
Collapse
|
6
|
Barbeiro HV, Barbeiro DF, de Souza HP, Soriano FG, Machado MC, Hajjar LA. LBP and iFABP mismatch in the evaluation of intestinal barrier dysfunction due to SARS-CoV-2 infection. Clinics (Sao Paulo) 2025; 80:100642. [PMID: 40273498 PMCID: PMC12051628 DOI: 10.1016/j.clinsp.2025.100642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
SARS-CoV-2 presents a hyperinflammatory scenario due to systemic inflammatory response syndrome with intense cytokine release, with consequent extrapulmonary involvement in 20 % of patients. The authors studied whether COVID-19 intestinal damage is a direct action of the virus on intestinal epithelial cells, with damage mainly at the tight junction. This is a retrospective observational study in a tertiary hospital emergency department. The authors studied 87 patients (46 patients over 61 years and 41 patients under 60 years old) with severe SARS-CoV-2 infection. The authors measured two plasma markers, LPS-Binding Protein (LBP) and ileal Fatty Acid-Binding Protein (iFABP). Furthermore, the authors evaluated the interaction between the two markers. TNF-α and IL-1 β were higher in bacterial co-infected patients and TNF-α was also higher in the older patients. Plasma iFABP levels were not statistically different in patients with bacterial co-infection; however, higher levels were found in the older population. Plasma LBP levels were higher in patients with bacterial co-infection when compared to patients without infection; however, when comparing plasma LBP levels in the older population with younger patients, no differences could be found. LBP, FABP, and cytokines can discriminate between bacterially infected patients and also discriminate elderly patients. The present study suggests that intestinal barrier dysfunction in SARS-CoV-2 infections is mainly due to damage to the intestinal tight junction complex with a disproportionately lower damage to enterocyte. In the older population, the authors also observed an increase in intestinal epithelial damage.
Collapse
Affiliation(s)
- Hermes Vieira Barbeiro
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil
| | - Denise Frediani Barbeiro
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil
| | - Heraldo Possolo de Souza
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil
| | - Francisco Garcia Soriano
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil.
| | - Marcel Cerqueira Machado
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil
| | - Ludhmila Abrahão Hajjar
- Laboratório de Investigação Médica da Disciplina de Emergências Clínicas, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo São Paulo, SP, Brazil
| |
Collapse
|
7
|
Alissa DA, Almasuood R, Alghalyini B, Tamim H, Raslan MA, Zaidi ARZ, Shaar BA, Masri MMA, Karim AA, Aburas W, Aburas W, Al-Jedai AH, Almaghrabi RS. Risk factors for mortality among kidney transplant recipients with COVID-19 in Saudi Arabia: a case-control study. BMC Infect Dis 2025; 25:560. [PMID: 40251535 PMCID: PMC12008917 DOI: 10.1186/s12879-025-10994-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/17/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has profoundly impacted global health, leading to over 74 million confirmed cases and 1.67 million deaths by December 2020. In Saudi Arabia, extensive measures were implemented to mitigate the spread of the virus. Kidney transplant recipients, due to their immunosuppressed status, are particularly vulnerable to severe COVID-19 outcomes. This study aims to identify risk factors associated with mortality in COVID-19-infected kidney transplant patients in Saudi Arabia. The primary objective is to identify mortality risk factors among COVID-19-infected kidney transplant patients. The secondary objective is to compare clinical management and outcomes between deceased and recovered patients. METHODOLOGY This case-control study matched 82 deceased kidney transplant patients (cases) with 151 survivors (controls). Data were collected from the National Registry for COVID-19 Mortality and King Faisal Specialist Hospital and Research Centre (KFSH&RC) for patients diagnosed between March 2020 and January 2021. Key variables included demographic information, comorbidities, clinical symptoms, and treatment details. Statistical analyses involved chi-square tests and multivariable logistic regression to assess associations with mortality. RESULTS Among cases, 93.9% required ICU admission, and 95.1% were intubated. Males constituted 73.2% of cases, with 53.7% aged over 60. Cardiovascular comorbidities were more prevalent among cases (97% vs. 87.4%, p = 0.01). and presented more frequently with fever, cough, and respiratory distress. In multivariable analysis, fever, shortness of breath, and desaturation were associated with increased mortality odds. Notably, patients who discontinued immunosuppressive therapy had higher mortality odds (OR = 63.2, p = 0.083), whereas those who held or adjusted their therapy had significantly lower odds (OR = 0.1, p = 0.042; OR = 0.0, p = 0.007). Bacterial infections also increased mortality risk (OR = 56.6, p = 0.009). CONCLUSION This study identifies critical risk factors for mortality among kidney transplant patients infected with COVID-19 in Saudi Arabia. The findings underscore the need for tailored clinical management strategies to improve outcomes in this vulnerable population. Further research is warranted to explore long-term implications and effective treatment protocols.
Collapse
Affiliation(s)
- Dema A Alissa
- Therapeutic Affairs Deputyship, Ministry of Health, Riyadh, Saudi Arabia.
- Colleges of Medicine and Pharmacy, Al-Faisal University, Riyadh, Saudi Arabia.
- College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Rawan Almasuood
- Therapeutic Affairs Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Baraa Alghalyini
- Department of Family and Community Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hala Tamim
- Department of Family and Community Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | | | - Abdul Rehman Zia Zaidi
- Department of Family and Community Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Bader Abou Shaar
- Department of Family and Community Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Mhd Malek Al Masri
- Colleges of Medicine and Pharmacy, Al-Faisal University, Riyadh, Saudi Arabia
| | - Adnan Abdul Karim
- Colleges of Medicine and Pharmacy, Al-Faisal University, Riyadh, Saudi Arabia
| | - Wejdan Aburas
- Therapeutic Affairs Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Waled Aburas
- College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Ahmed H Al-Jedai
- Therapeutic Affairs Deputyship, Ministry of Health, Riyadh, Saudi Arabia
- Colleges of Medicine and Pharmacy, Al-Faisal University, Riyadh, Saudi Arabia
| | - Reem S Almaghrabi
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Taib RR, Kozlov Y, Ekshtein A, Gordon B, Wand O, Ben-Ari O. A comparison of pulmonary function pre and post mild SARS-CoV-2 infection among healthy adults. BMC Pulm Med 2025; 25:163. [PMID: 40200177 PMCID: PMC11980318 DOI: 10.1186/s12890-025-03613-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND SARS-CoV-2 infection frequently involves the respiratory system and may impact on pulmonary function tests (PFT) of recovered individuals. Studies which compare post-COVID-19 PFT to pre-illness measurements are scarce. The primary objective of this study was to assess the effect of COVID-19 on PFT soon after infection. METHODS In this prospective observational study, PFT were measured early following recovery from COVID-19 among healthy military aircrew. Spirometry values were compared to pre-COVID-19 measurements, and abnormality rates of lung volumes and diffusion capacity for carbon monoxide (DLCO) were assessed. RESULTS The study included 252 aviators, 97.6% males, mean age 34.9-years, following recovery from SARS-CoV-2 infection. Participants manifested mild symptoms (79.4%) or were asymptomatic (20.6%). Post-COVID-19 spirometry results 10.79 ± 5.67 days following infection were compared to measurements performed 41.3 ± 28.59 months earlier. Pre- and post-COVID-19 results were comparable, with similar minimal abnormalities rates (2% and 4.4%, respectively). In addition, there were no restrictive abnormalities following infection, and just 7.7% of individuals had a marginally low DLCO of 70-80% of predicted. CONCLUSION Among vaccinated, healthy adults, mild COVID-19 had no significant impact on PFT early post-infection. The data suggest that systematic PFT testing might not be necessary for asymptomatic healthy individuals who recovered from mild COVID-19.
Collapse
Affiliation(s)
- Raz Roje Taib
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel
- Israel Defense Forces Medical Corps, Jerusalem, Israel
| | - Yuval Kozlov
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel
- Israel Defense Forces Medical Corps, Jerusalem, Israel
| | - Aya Ekshtein
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel
| | - Barak Gordon
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel
- Israel Defense Forces Medical Corps, Jerusalem, Israel
| | - Ori Wand
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel.
- Division of Pulmonary Medicine, Barzilai University Medical Center, Hahistadrut St. 2, Ashkelon, Israel.
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Oded Ben-Ari
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Israeli Air Force Aeromedical Center, Ramat-Gan, Israel
- Israel Defense Forces Medical Corps, Jerusalem, Israel
- The Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
9
|
Stoiber A, Gray G, Sailer G, Huf W, Tonna A. Frequency, type and severity of drug-related problems and pharmacist interventions in Paxlovid® prescribing: a descriptive analysis. Int J Clin Pharm 2025; 47:471-476. [PMID: 39708268 DOI: 10.1007/s11096-024-01852-5] [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: 08/25/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Paxlovid® (nirmatrelvir and ritonavir) is the only licensed oral antiviral for COVID-19. Ritonavir is a potent inhibitor of cytochrome P450 enzymes causing numerous drug-drug interactions (DDIs). AIM To describe the frequency, type, and severity of detected drug related problems (DRPs) associated with Paxlovid®. METHOD This study involved a retrospective quantitative analysis including all patients prescribed Paxlovid® at a public hospital in Vienna, Austria. Data were collected from the patients' records by a clinical pharmacist. A customised, piloted data collection form was used. A sample of data was checked for consistency by an independent clinical pharmacist. Any DDI and severity classification was recorded using an established interaction checker tool. Dosage adjustments due to renal impairment were recorded. RESULTS One hundred twenty-two of 140 patients (87.1%) required interventions to prevent DRPs. Pharmacists' intervention at dispensing was needed in 63.6% (n = 89) of cases. In 3 (2.1%) patients, Paxlovid® was prescribed despite being contraindicated due to severe renal impairment. The most common were DDIs (n = 80; 57.1%). Renal impairment and DDIs were noted in 24.3% (n = 34) of cases. A total of 313 DDIs were recorded in 114 (81.4%) patients, with severe interactions in 24 (17%) patients. CONCLUSION Pharmacists' involvement in prescribing highly interacting drugs such as Paxlovid® is essential to enhance patient safety.
Collapse
Affiliation(s)
- Alina Stoiber
- School of Pharmacy, Applied Sciences and Public Health, Robert Gordon University, Garthdee Road, Aberdeen, AB10 7QB, Scotland, UK
- Hospital Pharmacy, Clinic Hietzing, Vienna Healthcare Group, Vienna, Austria
| | - Gwen Gray
- School of Pharmacy, Applied Sciences and Public Health, Robert Gordon University, Garthdee Road, Aberdeen, AB10 7QB, Scotland, UK
| | - Gudrun Sailer
- Hospital Pharmacy, Clinic Hietzing, Vienna Healthcare Group, Vienna, Austria
| | - Wolfgang Huf
- Karl Landsteiner Institute for Clinical Risk Management, Vienna, Austria
| | - Antonella Tonna
- School of Pharmacy, Applied Sciences and Public Health, Robert Gordon University, Garthdee Road, Aberdeen, AB10 7QB, Scotland, UK.
| |
Collapse
|
10
|
Yuan J, Guan Y, Zhao Z, Shen J, Tan D, Zhao F, Ge L, Xie R, Li T. Enteral nutrition therapy for elderly patients with common-type COVID-19, a retrospective study based on medical records. Int Health 2025:ihaf012. [PMID: 40163463 DOI: 10.1093/inthealth/ihaf012] [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: 03/23/2024] [Revised: 12/07/2024] [Accepted: 02/13/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The objective was to investigate the implications of enteral nutrition for elderly patients with common-type coronavirus disease 2019 (COVID-19). METHODS Data were retrospectively extracted from medical records. Enteral nutritional supplementation was recommended for patients with a nutritional risk score >3. The preferred method was oral administration, and preparations included Ensure and TPF-T. Continuous variables were compared using analysis of two-tailed Student's t-tests or one-way analysis of variance for normally distributed data and the rank sum test for non-normally distributed data. Categorical variables were compared using the χ2 test or Fisher's exact test. Values of p <0.05 were considered to be statistically significant. RESULTS The mortality rate in the whole cohort was 9.54%. A total of 474 patients tested negative and were discharged; among them, 173 patients received enteral nutrition while 301 patients did not. There were significant correlations between mortality and age, serum albumin concentration, prognostic nutritional index, underlying severe disease status and diet condition. In patients with a poor diet, early use of enteral nutrition is associated with faster conversion to a negative polymerase chain reaction test. CONCLUSIONS The prognosis of elderly patients with common-type COVID-19 was related to their nutritional status. Enteral nutritional supplementation is the preferred method of nutrition because it is the simplest and most widely accepted method for patients. For patients with poor diet conditions, enteral nutritional intervention should be performed early.
Collapse
Affiliation(s)
- Jianming Yuan
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Yuening Guan
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Zhifeng Zhao
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, No.127, West Changle Road, Xi'an, Shaanxi Province, China
| | - Jiankang Shen
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Dan Tan
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Fang Zhao
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Lei Ge
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Rongli Xie
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| | - Tingting Li
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiaotong University School of Medicine, 149 Chongqing South Road, Huangpu District, Shanghai, China
| |
Collapse
|
11
|
Kim SH, Lee H, Kim MJ, Kim Y, Min KH, Yoo KH, Kim JS, Moon JY. Risk of acute exacerbation of chronic obstructive pulmonary disease after COVID-19 recovery: a nationwide population-based cohort study. Respir Res 2025; 26:116. [PMID: 40148876 PMCID: PMC11951598 DOI: 10.1186/s12931-025-03123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is associated with severe Coronavirus disease 2019 (COVID-19) outcomes. However, it is uncertain whether the risk of acute exacerbation of COPD (AECOPD) increases after recovering from COVID-19. METHODS This study included 2,118 individuals with COPD from the Korea National Health Insurance Service database who were also diagnosed with COVID-19. Matched controls were chosen using 1:1 propensity score (PS) matching. We compared the risk of AECOPD after COVID-19 recovery between the COVID-19 cohort and matched controls between October 8, 2020, and December 31, 2021, using PS-matched Cox proportional hazard regression models. RESULTS During a median follow-up of 62 days (interquartile range, 29-179 days), including a median of 14 days of recovery time after COVID-19, 68 people (5.6%) in the COVID-19 cohort and 50 (3.9%) in the matched control group experienced AECOPD. Compared to the matched controls, the COVID-19 cohort had a significantly higher risk of overall AECOPD (hazard ratio [HR] = 1.45, 95% confidence interval [CI] = 1.09-1.92). This increased risk was particularly evident for severe AECOPD among individuals who had severe COVID-19 within the first 30days post-recovery (aHR = 8.14, 95% CI = 3.32-19.97). When classified by COVID-19 severity, while severe COVID-19 significantly increased this risk (aHR = 2.97, 95% CI = 2.15-4.11), non-severe COVID did not significantly influence the risk of AECOPD, regardless of time duration or exacerbation severity. CONCLUSION Individuals with COPD who had severe COVID-19 have increased risk of AECOPD after COVID-19 recovery, especially within the first 30 days after COVID-19 recovery.
Collapse
Affiliation(s)
- Sang Hyuk Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju, Republic of Korea
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang Medical Center, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Min Ji Kim
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Youlim Kim
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Kyung Hoon Min
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kwang Ha Yoo
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Jong Seung Kim
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, Republic of Korea.
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.
- Department of Otorhinolaryngology-Head and Neck Surgery, Jeonbuk National University Medical School, Jeonju, Republic of Korea.
| | - Ji-Yong Moon
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea.
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05030, Republic of Korea.
| |
Collapse
|
12
|
Perestiuk V, Kosovska T, Volianska L, Boyarchuk O. Association of zinc deficiency and clinical symptoms, inflammatory markers, severity of COVID-19 in hospitalized children. Front Nutr 2025; 12:1566505. [PMID: 40201587 PMCID: PMC11975579 DOI: 10.3389/fnut.2025.1566505] [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: 01/24/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Zinc plays an important role in the functioning of the immune system. Zinc deficiency leads to increased susceptibility to inflammatory and infectious diseases. There are few studies investigating the role of zinc in the development and progression of COVID-19 in children, and their findings remain inconsistent. This study aimed to determine the zinc levels in children with COVID-19 and assess their association with symptoms, inflammation markers, and disease progression. Methods A prospective cohort study included hospitalized patients under 18 years who had a confirmed diagnosis of SARS-CoV-2 infection. Serum zinc concentrations were measured using a colorimetric method. Based on zinc levels, the children were divided into two groups: the first group had concentrations below 10.7 μmol/L, indicating zinc deficiency, while the second group had levels above 10.7 μmol/L, which was considered within the optimal range. Results In total, 140 hospitalized patients with COVID-19 were examined. Zinc deficiency was identified in 40 children (28.6%), while optimal levels were found in 100 children (71.4%). Zinc status did not depend on the age of the children. Among the symptoms of acute SARS-CoV-2 infection, children with zinc deficiency showed a trend toward more frequent fever occurrences (p = 0.0654). No significant impact of zinc status was observed on the severity of COVID-19 or the duration of hospitalization. Children with zinc deficiency had higher median values of the neutrophil-to-lymphocyte ratio (NLR) (1.84 vs. 1.09, p = 0.0010), C-reactive protein (CRP) levels (9.65 vs. 3.96 mg/L, p = 0.0053), and fibrinogen levels (2.88 vs. 2.07 g/L, p = 0.0057) compared to those with adequate zinc levels. Additionally, the percentage of patients with a NLR greater than 4, elevated CRP, and fibrinogen levels was higher in the zinc-deficient group (p = 0.0017, p = 0.0107, p = 0.0338, respectively). Conclusion Zinc deficiency was observed in 28.6% of children with COVID-19 and was not dependent on age. Children with hypozincemia had higher levels of inflammation markers, including the neutrophil-to-lymphocyte ratio and CRP.
Collapse
Affiliation(s)
- Vita Perestiuk
- Department of Children’s Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | | | | | | |
Collapse
|
13
|
İdikut A, Değer İ, Göktaş G, Karahan S, Sarınç S, Köksal D, Babaoğlu MO, Babaoğlu E. Association of Endothelial Nitric Oxide Synthase Polymorphisms with Clinical Severity in Patients with COVID-19. J Clin Med 2025; 14:1931. [PMID: 40142738 PMCID: PMC11943162 DOI: 10.3390/jcm14061931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: To elucidate the factors that contribute to individual variability in the progression of COVID-19, experiments on endothelial nitric oxide synthase polymorphisms have been reported. Nitric oxide synthase (NOS3) is located in the endothelium and is involved in the regulation of inflammation and vascular homeostasis. In this study, we investigated the association between COVID-19 severity and NOS3 G894T and NOS3 27-bp VNTR 4b/a genetic polymorphisms. Methods: Patients with COVID-19 (n = 178) were divided into Group 1 (mild disease) and Group 2 (severe disease) based on oxygen saturation levels in room air (Group 1, SpO2 ≥ 93%, n = 107; and Group 2, SpO2 < 93%, n = 73) and hospitalization requirements. Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism analysis. Results: Overall, genotype and allele frequencies of the NOS3 genetic polymorphisms were similar across the two study groups (p > 0.05). However, the subgroup analysis showed a notable trend for the 4b/4a allele distribution between Groups 1 and 2. In the younger subgroup of patients (≤50 years old) without chronic obstructive pulmonary disease, Group 2 tended to have a higher frequency of the 4b allele than Group 1 (97.4% vs. 85.4% p = 0.06) and a higher occurrence of 4b/4b genotype (94.7% vs. 74.0%, p = 0.05). Additionally, a rarely observed 4c allele was detected only in two subjects within Group 2 but not in Group 1. Conclusions: These findings suggest a trend of association between COVID-19 severity and NOS3 27-bp VNTR 4b/a genetic polymorphism. Genetic analysis may reveal patient susceptibility to disease, prognosis risk factors, and drug responsiveness.
Collapse
Affiliation(s)
- Aytekin İdikut
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - İlter Değer
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye; (İ.D.); (M.O.B.)
| | - Gamze Göktaş
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Sevilay Karahan
- Department of Bioistatistics, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye;
| | - Sevinç Sarınç
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Deniz Köksal
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| | - Melih O. Babaoğlu
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye; (İ.D.); (M.O.B.)
| | - Elif Babaoğlu
- Department of Chest Diseases, Faculty of Medicine, Hacettepe University, Ankara 06230, Türkiye (S.S.); (D.K.)
| |
Collapse
|
14
|
Jensen AL, Litorell J, Grip J, Dahlberg M, Joelsson‐Alm E, Jonmarker S. A descriptive, retrospective single-centre study of air-leak syndrome in intensive care unit patients with COVID-19. Acta Anaesthesiol Scand 2025; 69:e14582. [PMID: 39936659 PMCID: PMC11816560 DOI: 10.1111/aas.14582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Acute respiratory failure is the predominant presentation of intensive care unit (ICU) patients with COVID-19, and lung protective strategies are recommended to mitigate additional respiratory complications such as air-leak syndrome. The aim of this study is to investigate the prevalence, type, and timing of air-leak syndrome with regards to associated factors and patient outcome in patients with COVID-19 in ICUs at a large Swedish emergency hospital. METHODS This retrospective study included all adult patients admitted to an ICU for COVID-19-related respiratory failure at Södersjukhuset between March 6, 2020, and June 6, 2021. Primary outcomes were proportion of patients diagnosed with air-leak syndrome and its different types of manifestations, and timing of diagnoses in relation to ICU admission and initiation of invasive ventilation. Secondary outcomes included the highest level of respiratory support prior to the diagnosis of air-leak syndrome, patient characteristics and treatment variables associated with air-leak syndrome, and 90-day mortality for patients with air-leak syndrome compared to those without. RESULTS Out of a total of 669 patients, 81 (12%) were diagnosed with air-leak syndrome. Air-leak syndrome manifested as pneumomediastinum (PMD) (n = 58, 72%), pneumothorax (PTX) (n = 43, 53%), subcutaneous emphysema (SCE) (n = 28, 35%) and pneumatocele (PC) (n = 4, 4.9%). Air-leak syndrome was diagnosed at a median of 14 days (IQR 6-22) after ICU admission and 12 days (IQR 6-19) following the initiation of invasive ventilation. The highest respiratory support prior to diagnosis was invasive ventilation (IV) in 64 patients (79%), non-invasive ventilation in two patients (2.5%), and low- or high-flow oxygen in 15 patients (19%). Multiple logistic regression showed that pulmonary disease at baseline (OR 1.87, 95% CI 1.07-3.25), a lower body mass index (OR 0.95, 95% CI 0.9-0.99), admission later compared with earlier in the pandemic (OR 3.89, 95% CI 2.14-7.08), and IV (OR 3.92, 95% CI 2.07-7.44) were associated with an increased risk of air-leak syndrome. Compared with patients not diagnosed with air-leak syndrome, patients with air-leaks had a higher mortality at 90 days after ICU admission, 46% versus 26% (p <.001). However, the mortality rate differed with different air-leak manifestations, 47% for PMD, 47% for PTX, 50% for the combination of both PMD and PTX and 0% in patients with only SCE and/or PC, respectively. CONCLUSION In 669 ICU patients with COVID-19, 12% had one or more manifestations of air-leak syndrome. Notably, PMD, rather than PTX, was the most common manifestation, suggesting a potentially distinctive feature of COVID-19-related air-leak syndrome. Further research is needed to determine whether COVID-19 involves different pathophysiological or iatrogenic mechanisms compared with other critical respiratory conditions. REGISTRATION OF CLINICAL TRIAL Clinicaltrials.gov, identifying number, NCT05877443. EDITORIAL COMMENT This single-centre cohort study of air leakage into soft tissue in ventilated COVID cases presents findings for associated factors and clinical manifestations, including with different COVID-19 periods and treatments.
Collapse
Affiliation(s)
| | - Jacob Litorell
- Department of Anaesthesia and Intensive CareSödersjukhusetStockholmSweden
| | - Jonathan Grip
- Department of Clinical Science, Intervention and Technology, CLINTECKarolinska InstitutetStockholmSweden
- Function Perioperative Medicine and Intensive CareKarolinska University HospitalStockholmSweden
| | - Martin Dahlberg
- Department of SurgerySödersjukhusetStockholmSweden
- Department of Clinical Science and Education, SödersjukhusetKarolinska InstitutetStockholmSweden
| | - Eva Joelsson‐Alm
- Department of Anaesthesia and Intensive CareSödersjukhusetStockholmSweden
- Department of Clinical Science and Education, SödersjukhusetKarolinska InstitutetStockholmSweden
| | - Sandra Jonmarker
- Department of Anaesthesia and Intensive CareSödersjukhusetStockholmSweden
- Department of Clinical Science and Education, SödersjukhusetKarolinska InstitutetStockholmSweden
| |
Collapse
|
15
|
Zhang L, Wang X, Chen XW. The biogenesis and transport of triglyceride-rich lipoproteins. Trends Endocrinol Metab 2025; 36:262-277. [PMID: 39164120 DOI: 10.1016/j.tem.2024.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024]
Abstract
Triglyceride-rich lipoproteins (TRLs) play essential roles in human health and disease by transporting bulk lipids into the circulation. This review summarizes the fundamental mechanisms and diverse factors governing lipoprotein production, secretion, and regulation. Emphasizing the broader implications for human health, we outline the intricate landscape of lipoprotein research and highlight the potential coordination between the biogenesis and transport of TRLs in physiology, particularly the unexpected coupling of metabolic enzymes and transport machineries. Challenges and opportunities in lipoprotein biology with respect to inherited diseases and viral infections are also discussed. Further characterization of the biogenesis and transport of TRLs will advance both basic research in lipid biology and translational medicine for metabolic diseases.
Collapse
Affiliation(s)
- Linqi Zhang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China
| | - Xiao Wang
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China.
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, PR China; Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, PR China; Peking University (PKU)-Tsinghua University (THU) Joint Center for Life Sciences, Peking University, Beijing 100871, PR China.
| |
Collapse
|
16
|
Browne SH, Bernstein M, Bickler PE. Evaluation of Pulse Oximetry Accuracy in a Commercial Smartphone and Smartwatch Device During Human Hypoxia Laboratory Testing. SENSORS (BASEL, SWITZERLAND) 2025; 25:1286. [PMID: 40096012 PMCID: PMC11902706 DOI: 10.3390/s25051286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Background: The US Food and Drug Administration (FDA) and International Organization for Standardization (ISO) clearance standards for the clinical use of smart device pulse oximetry require in-laboratory human hypoxemia testing in healthy human individuals using arterial blood gas analysis. Methods: We evaluated the SpO2 measurements of the Samsung smartphone (Galaxy S9/10) and smartwatch (Galaxy 4) at stable arterial oxygen saturations (SaO2) between 70 and 100% in 24 healthy participants. Testing followed FDA/ISO-stipulated procedures for pulse oximetry performance validation, which include questionnaire estimation of skin tone based on Fitzpatrick estimation of skin types I-VI. During testing, inspired oxygen, nitrogen, and carbon dioxide partial pressures were monitored and adjusted via partial rebreathing circuits to achieve stable target arterial blood oxygen (SaO2) plateaus between 70% and 100%. Arterial blood samples were taken at each plateau, with device SpO2 readings taken at each sample extraction. An ABL-90FLEX blood gas analyzer determined arterial blood sample SaO2. Bias, calculated from device readings minus corresponding arterial blood measurements, was reported as root mean square deviation (RMSD). Results: Combined Participants demographics were: 62.5% female; median age 26 years (range 21-46); and race/ethnicity 16.7% African American, 33.3% Asian, 12.5% multi-ethnic, and 37.5% Caucasian. Fitzpatrick Skin Scale-identified skin tones were: white-fair (I&II), 20.8%; average-light brown (III-IV), 54% and brown-black (V-VI), 25%. There were no adverse events. The RMSD values of SpO2 measurements were: smartphone 2.6% (257 data pairs) and smartwatch 1.8% (247 data pairs). Conclusions: Device SpO2 demonstrated RMSD < 3.0% to SaO2, meeting FDA/ISO clearance standards at the time of study. However, additional testing in persons with darker skin tones is necessary. Smartphones and paired wearables, when cleared for clinical use following revision of FDA clearance standards, may expand access to remote pulse oximetry.
Collapse
Affiliation(s)
- Sara H. Browne
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Specialists in Global Health, Encinitas, CA 92024, USA
| | - Michael Bernstein
- Physio Monitor, LLC, San Ramon, CA 94583, USA
- Department of Anesthesia, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Philip E. Bickler
- Department of Anesthesia, University of California San Francisco, San Francisco, CA 94143, USA;
| |
Collapse
|
17
|
Lee H, Kim SH, Jeong CY, Chung JE, Kim Y, Min KH, Yoo KH, Kim JS, Moon JY. COVID-19 and risk of long-term mortality in COPD: a nationwide population-based cohort study. BMJ Open Respir Res 2025; 12:e002694. [PMID: 39961706 PMCID: PMC11836811 DOI: 10.1136/bmjresp-2024-002694] [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: 07/04/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a risk factor for severe COVID-19. However, mortality after COVID-19 recovery in this population remains unclear. METHODS We retrospectively enrolled individuals with COPD from the Korean National Health Insurance database. We compared the mortality rate in individuals with COPD who recovered from COVID-19 between 8 October 2020 and 31 December 2021 (COVID-19 cohort, n=2499) with that in 1:1 propensity score-matched controls (n=2499). The study population was followed until either death or 30 September 2022, whichever came first. RESULTS The COVID-19 cohort had a 4.8% mortality rate vs 2.7% in matched controls during a median follow-up of 319 days (IQR, 293-422 days), including 14 days of recovery time. The COVID-19 cohort had a higher risk of death than matched controls (adjusted HR (aHR)=1.81, 95% CI=1.35 to 2.45). The risk of mortality was notably higher in individuals with severe COVID-19 (aHR=5.05, 95% CI=3.65 to 6.97), especially during the first 180 days of recovery (highest during the first 30 days (aHR=20.25, 95% CI=7.79 to 52.64)). Non-severe COVID-19 does not increase the risk of mortality compared with controls (aHR=0.85, 95% CI=0.57 to 1.28). CONCLUSION Individuals with COPD recovering from COVID-19 showed an increased risk of long-term mortality, particularly within the first 180 days post-recovery, especially those who experienced severe COVID-19.
Collapse
Affiliation(s)
- Hyun Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Sang Hyuk Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Cho Yun Jeong
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Jee-Eun Chung
- College of Pharmacy, Hanyang University, Seoul, South Korea
| | - Youlim Kim
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Kyung Hoon Min
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Kwang Ha Yoo
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Jong Seung Kim
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Ji-Yong Moon
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Mallol R, Rombauts A, Abelenda-Alonso G, Gudiol C, Balsalobre M, Carratalà J. Metabolomic profile of severe COVID-19 and a signature predictive of progression towards severe disease status: a prospective cohort study (METCOVID). Sci Rep 2025; 15:4963. [PMID: 39929875 PMCID: PMC11811168 DOI: 10.1038/s41598-025-87288-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Profound metabolomic alterations occur during COVID-19. Early identification of the subset of hospitalised COVID-19 patients at risk of developing severe disease is critical for optimal resource utilization and prompt treatment. This work explores the metabolomic profile of hospitalised adult COVID-19 patients with severe disease, and establishes a predictive signature for disease progression. Within 48 hours of admission, serum samples were collected from 148 hospitalised patients for nuclear magnetic resonance (NMR) spectroscopy. Lipoprotein profiling was performed using the 1H-NMR-based Liposcale test, while low molecular weight metabolites were analysed using one-dimensional Carr-Purcell-Meiboom-Gill pulse spectroscopy and an adaptation of the Dolphin method for lipophilic extracts. Severe COVID-19, per WHO's Clinical Progression Scale, was characterized by altered lipoprotein distribution, elevated signals of glyc-A and glyc-B, a shift towards a catabolic state with elevated levels of branched-chain amino acids, and accumulation of ketone bodies. Furthermore, COVID-19 patients initially presenting with moderate disease but progressing to severe stages exhibited a distinct metabolic signature. Our multivariate model demonstrated a cross-validated AUC of 0.82 and 72% predictive accuracy for severity progression. NMR spectroscopy-based metabolomic profiling enables the identification of moderate COVID-19 patients at risk of disease progression, aiding in resource allocation and early intervention.
Collapse
Affiliation(s)
- Roger Mallol
- Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08007, Barcelona, Spain
| | - Alexander Rombauts
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain.
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Institut Català d'Oncologia (ICO), Hospital Duran i Reynals, 08908, Barcelona, Spain
| | - Marc Balsalobre
- Human Environment Research, La Salle-Universitat Ramon Llull, 08022, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge-IDIBELL, 08907, Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, 08007, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
19
|
Schultz IC, Dos Santos Pereira Andrade AC, Dubuc I, Laroche A, Allaeys I, Doré E, Bertrand N, Vallières L, Fradette J, Flamand L, Wink MR, Boilard E. Targeting Cytokines: Evaluating the Potential of Mesenchymal Stem Cell Derived Extracellular Vesicles in the Management of COVID-19. Stem Cell Rev Rep 2025; 21:564-580. [PMID: 39340739 DOI: 10.1007/s12015-024-10794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
The Coronavirus Disease 2019 (COVID-19), caused by virus SARS-CoV-2, is characterized by massive inflammation and immune system imbalance. Despite the implementation of vaccination protocols, the accessibility of treatment remains uneven. Furthermore, the persistent threat of new variants underscores the urgent need for expanded research into therapeutic options for SARS-CoV-2. Mesenchymal stem cells (MSCs) are known for their immunomodulatory potential through the release of molecules into the extracellular space, either as soluble elements or carried by extracellular vesicles (EVs). The aim of this study was to evaluate the anti-inflammatory potential of EVs obtained from human adipose tissue (ASC-EVs) against SARS-CoV-2 infection. ASC-EVs were purified by size-exclusion chromatography, and co-culture assays confirmed that ASC-EVs were internalized by human lung cells and could colocalize with SARS-CoV-2 into early and late endosomes. To determine the functionality of ASC-EVs, lung cells were infected with SARS-CoV-2 in the presence of increasing concentrations of ASC-EVs, and the release of cytokines, chemokines and viruses were measured. While SARS-CoV-2 replication was significantly reduced only at the highest concentrations tested, multiplex analysis highlighted that lower concentrations of ASC-EV sufficed to prevent the production of immune modulators. Importantly, ASC-EVs did not contain detectable inflammatory cytokines, nor did they trigger inflammatory mediators, nor affect cellular viability. In conclusion, this work suggests that ASC-EVs have the potential to attenuate inflammation by decreasing the production of pro-inflammatory cytokines in lung cells following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Iago Carvalho Schultz
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Ana Claudia Dos Santos Pereira Andrade
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Dubuc
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Isabelle Allaeys
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Etienne Doré
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Nicolas Bertrand
- Axe Endocrinologie et Néphrologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Département de Chirurgie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
- Division of Regenerative Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
| | - Louis Flamand
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada
| | - Marcia Rosangela Wink
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Eric Boilard
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC, Canada.
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine de l'Université Laval, Québec, QC, Canada.
| |
Collapse
|
20
|
Wei J, Liu S, Bian Y, Li L, Qian B, Shen Z, Zhang Y, Abuduaini A, Dong F, Zhang X, Li J, Yu Y, Zhang W, Wang J, Zhai W, Song Q, Zheng Y, Pan W, Yu L, Zhan Q, Zhang N, Zheng J, Pan S, Yao C, Li H. Safety and efficacy of oral administrated cepharanthine in non-hospitalized, asymptomatic or mild COVID-19 patients: a Double-blind, randomized, placebo-controlled trial : Author detials. Sci Rep 2025; 15:3875. [PMID: 39890847 PMCID: PMC11785718 DOI: 10.1038/s41598-024-75891-3] [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: 04/28/2024] [Accepted: 10/09/2024] [Indexed: 02/03/2025] Open
Abstract
Cepharanthine (CEP) is a natural remedy that potently inhibits SARS-CoV-2 activity both in vitro and in vivo. To evaluate the efficacy and safety of CEP compared with placebo in adults with asymptomatic or mild coronavirus disease 2019 (COVID-19), we conducted a proof-of-concept, double-blind, randomized, placebo-controlled trial. Patients were randomized to receive 120 mg/day of CEP, 60 mg/day CEP or placebo for 5 days. Main outcome was the time from randomization to negative nasopharyngeal swab and safety. Among 262 randomized participants, 188 completed the trial among group of 120 mg/day CEP (n = 65), 60 mg/day CEP (n = 68) and placebo (n = 55). Neither 120 mg/day or 60 mg/day CEP shortened the time to negative significantly compared with placebo. However, 60 mg/day CEP showed a slight trend (difference=-0.77 days, hazard ratio (HR) = 1.40, 95% CI 0.97-2.01, p = 0.072). In analysis of participants with good medication compliance, 60 mg/day CEP significantly shortened the time to negative (difference=-0.87 days, HR = 1.56, 95% CI 1.03-2.37, p = 0.035). Adverse events were not different among the three groups, and no serious adverse events occurred. In conclusion, treatment of asymptomatic or mild Covid-19 with 120 mg/day or 60 mg/day did not shorten the time to negative significantly. However, 60 mg/day CEP showed a slight trend which needs future confirmatory trials to validate. (NCT05398705).
Collapse
Affiliation(s)
- Jianyi Wei
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Shupeng Liu
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yuexiang Bian
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Lei Li
- Department of Otorhinolaryngology-Head & Neck Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Biyun Qian
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zixuan Shen
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yan Zhang
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Adila Abuduaini
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Fuchen Dong
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Xin Zhang
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Jinhui Li
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China
| | - Yongpei Yu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China
| | - Weituo Zhang
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wang
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qixiang Song
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zheng
- Department of Respiratory Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihua Pan
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lanlan Yu
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China
| | - Qimin Zhan
- Peking University - Yunnan Baiyao International Medical Research Center, Beijing, China
| | - Ning Zhang
- Peking University - Yunnan Baiyao International Medical Research Center, Beijing, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| | - Chen Yao
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, 100083, China.
| | - Hai Li
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), 1630 Dong Fang Road, Shanghai, 200127, China.
| |
Collapse
|
21
|
Kochi M, Yokoyama Y. [Posterior reversible encephalopathy syndrome after COVID-19 in a patient with chronic renal failure]. Rinsho Shinkeigaku 2025; 65:32-38. [PMID: 39710393 DOI: 10.5692/clinicalneurol.cn-002013] [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: 12/24/2024]
Abstract
A 61-year-old man with chronic renal failure had an embolic stroke of undetermined source that was treated with warfarin. Five weeks later, the patient contracted coronavirus disease (COVID-19). Six days after the onset of COVID-19, high blood pressure (>200 mmHg) and consciousness disturbance were reported. CT demonstrated symmetrical hypodensity areas in the bilateral cerebellar hemispheres. MRI revealed hyperintensity lesions in the bilateral cerebellar hemispheres and pons on the T2-weighted and fluid-attenuated inversion recovery images. Moreover, cerebellar lesions appeared as hyperintensity areas on apparent diffusion coefficient mapping. Based on these findings, a diagnosis of posterior reversible encephalopathy syndrome (PRES) was made. The patient was treated with antihypertensive drugs, and the consciousness level improved gradually. MRI after one month showed that the lesions had disappeared. PRES should be considered if the brain CT of patients with COVID-19 shows a low-density lesion, especially in patients with risk factors for PRES such as chronic renal failure or hypertension.
Collapse
|
22
|
Lee JS, Dittmar M, Miller J, Li M, Ayyanathan K, Ferretti M, Hulahan J, Whig K, Etwebi Z, Griesman T, Schultz DC, Cherry S. Pressure to evade cell-autonomous innate sensing reveals interplay between mitophagy, IFN signaling, and SARS-CoV-2 evolution. Cell Rep 2025; 44:115115. [PMID: 39708319 DOI: 10.1016/j.celrep.2024.115115] [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: 07/03/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
SARS-CoV-2 emerged, and continues to evolve, to efficiently infect humans worldwide. SARS-CoV-2 evades early innate recognition, interferon signaling occurring only in bystander cells. How the virus continues to evolve in the face of innate responses has important consequences, but the pathways involved are incompletely understood. Here, we find that autophagy genes regulate innate immune signaling, impacting the basal set point of interferons and, thus, permissivity to infection. Mechanistically, autophagy (mitophagy) genes negatively regulate MAVS, and this low basal level of MAVS is efficiently antagonized by SARS-CoV-2 ORF9b, blocking interferon activation in infected cells. However, loss of autophagy increased MAVS and overcomes ORF9b-mediated antagonism. This has driven the evolution of SARS-CoV-2 to express more ORF9b, allowing SARS-CoV-2 to replicate under conditions of increased MAVS signaling. Altogether, we find a critical role of mitophagy in the regulation of innate immunity and uncover an evolutionary trajectory of SARS-CoV-2 ORF9b to overcome host defenses.
Collapse
Affiliation(s)
- Jae Seung Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Dittmar
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Minghua Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kasirajan Ayyanathan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Ferretti
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse Hulahan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanupriya Whig
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zienab Etwebi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trevor Griesman
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David C Schultz
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Ferrando-Terradez I, Núñez-Cortés R, López-Bueno L, Alcántara E, Calatayud J, Casaña J, Parčina I, Dueñas L. Relationship between psychological empowerment, physical activity enjoyment, and response to a HIIT Program in physically inactive young women: a prospective multicentre study. BMC Womens Health 2025; 25:38. [PMID: 39871301 PMCID: PMC11771111 DOI: 10.1186/s12905-025-03576-8] [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: 10/08/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND This study aimed to investigate the relationship of psychological empowerment and enjoyment of physical activity with changes in physical activity levels, sleep quality, and muscular endurance following a high-intensity interval training (HIIT) program in physically inactive young women. METHODS A total of 61 physically inactive young women (age: 20.1 ± 2.7 y) were recruited to participate in a six-month HIIT intervention delivered via a smartphone app. Outcome measures included physical activity levels (MET-min/week), muscular endurance (plank test), and sleep quality through the Pittsburgh Sleep Quality Index (PSQI). The Healthy Lifestyle and Personal Control Questionnaire (HLPCQ) and the Physical Activity Enjoyment Scale (PACES) were used to assess psychological empowerment and enjoyment, respectively. Repeated measures ANOVA and covariate analyses were performed to evaluate the impact of the intervention and the role of psychological empowerment and enjoyment. RESULTS At 6 months, significant improvements in physical activity (p < 0.001; ηp²=0.336) and muscular endurance (p = 0.005; ηp²=0.085) were observed, with large and moderate effect sizes, respectively. The PACES showed a significant interaction with time for MET-min/week (F = 11.67, p = 0.001, ηp²=0.129), suggesting that enjoyment influenced the increase in physical activity. No significant differences in sleep quality were observed (p > 0.05). CONCLUSION Enjoyment plays a crucial role in the response to HIIT programs among physically inactive young women, particularly in improving weekly physical activity levels. Psychological training showed no significant relationship with the outcomes studied.
Collapse
Affiliation(s)
| | - Rodrigo Núñez-Cortés
- Department of Physical Therapy, Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago, Chile.
| | - Laura López-Bueno
- Department of Physiotherapy, University of Valencia, Valencia, 46010, Spain
- Exercise Intervention for Health Research Group (EXINH-RG), Department of Physiotherapy, Universitat de Valencia, Valencia, 46010, Spain
| | - Enrique Alcántara
- Dr. Q. Data Driven Innovation and Association of the valencian community for driving R&D and impact innovation in sports (4ivlcesport), Valencia, Spain
| | - Joaquín Calatayud
- Exercise Intervention for Health Research Group (EXINH-RG), Department of Physiotherapy, Universitat de Valencia, Valencia, 46010, Spain
| | - José Casaña
- Exercise Intervention for Health Research Group (EXINH-RG), Department of Physiotherapy, Universitat de Valencia, Valencia, 46010, Spain
| | - Ivana Parčina
- Faculty of Sport, University "Union-Nikola Tesla", Belgrade, 11000, Serbia
| | - Lirios Dueñas
- Department of Physiotherapy, University of Valencia, Valencia, 46010, Spain
- Physiotherapy in Motion, Multi-Specialty Research Group (PTinMOTION), Department of Physiotherapy, University of Valencia, Valencia, 46010, Spain
| |
Collapse
|
24
|
Rabe DC, Choudhury A, Lee D, Luciani EG, Ho UK, Clark AE, Glasgow JE, Veiga S, Michaud WA, Capen D, Flynn EA, Hartmann N, Garretson AF, Muzikansky A, Goldberg MB, Kwon DS, Yu X, Carlin AF, Theriault Y, Wells JA, Lennerz JK, Lai PS, Rabi SA, Hoang AN, Boland GM, Stott SL. Ultrasensitive detection of intact SARS-CoV-2 particles in complex biofluids using microfluidic affinity capture. SCIENCE ADVANCES 2025; 11:eadh1167. [PMID: 39792670 PMCID: PMC11721714 DOI: 10.1126/sciadv.adh1167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025]
Abstract
Measuring virus in biofluids is complicated by confounding biomolecules coisolated with viral nucleic acids. To address this, we developed an affinity-based microfluidic device for specific capture of intact severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our approach used an engineered angiotensin-converting enzyme 2 to capture intact virus from plasma and other complex biofluids. Our device leverages a staggered herringbone pattern, nanoparticle surface coating, and processing conditions to achieve detection of as few as 3 viral copies per milliliter. We further validated our microfluidic assay on 103 plasma, 36 saliva, and 29 stool samples collected from unique patients with COVID-19, showing SARS-CoV-2 detection in 72% of plasma samples. Longitudinal monitoring in the plasma revealed our device's capacity for ultrasensitive detection of active viral infections over time. Our technology can be adapted to target other viruses using relevant cell entry molecules for affinity capture. This versatility underscores the potential for widespread application in viral load monitoring and disease management.
Collapse
Affiliation(s)
- Daniel C. Rabe
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adarsh Choudhury
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dasol Lee
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Evelyn G. Luciani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Uyen K. Ho
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alex E. Clark
- Departments of Pathology and Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey E. Glasgow
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sara Veiga
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - William A. Michaud
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Diane Capen
- Microscopy Core of the Program in Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth A. Flynn
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicola Hartmann
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron F. Garretson
- Departments of Pathology and Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alona Muzikansky
- Massachusetts General Hospital Biostatistics, Harvard Medical School, Boston, MA, USA
| | - Marcia B. Goldberg
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Douglas S. Kwon
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Xu Yu
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Aaron F. Carlin
- Departments of Pathology and Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yves Theriault
- Qualcomm Institute, University of California, San Diego, La Jolla, CA, USA
| | - James A. Wells
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jochen K. Lennerz
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peggy S. Lai
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sayed Ali Rabi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anh N. Hoang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Departments of Pathology and Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Microscopy Core of the Program in Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
- Massachusetts General Hospital Biostatistics, Harvard Medical School, Boston, MA, USA
- Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Qualcomm Institute, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Genevieve M. Boland
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shannon L. Stott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
25
|
Antonogiannaki EM, Grigoropoulos I, Manali ED, Thomas K, Kallieri M, Alexopoulou P, Papaioannou AI, Prountzos S, Karachaliou A, Kontopoulou C, Karageorgou V, Lampadakis S, Blizou M, Tomos I, Grigoropoulou S, Kavatha D, Loukides S, Antoniadou A, Papiris SA. Long-Term Lung Sequelae in Survivors of Severe/Critical COVID-19 Pneumonia: The "Non-Steroid", "Non-Interventional" Approach. J Clin Med 2025; 14:347. [PMID: 39860353 PMCID: PMC11766020 DOI: 10.3390/jcm14020347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/02/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: Long-term lung sequelae in severe COVID-19 survivors, as well as their treatment, are poorly described in the current literature. Objective: To investigate lung fibrotic sequelae in survivors of severe/critical COVID-19 pneumonia and their fate according to a "non-interventional" approach. Methods: Prospective study of the above COVID-19 survivors after hospital discharge from March 2020 to October 2022. Re-evaluation lasted 3-12 months and included chest HRCT, PFTs, dyspnea, and overall health evaluation by modified Medical Research Council (mMRC) and St. George's Respiratory Questionnaire (SGRQ), respectively. Results: In this study, 198 patients (61.1% male) with a median age of 57 years (IQR 49-66). After 3 months, 187 (94.4%) patients were assessed; after 6 months, 82 (41.1%) patients were assessed; and after 12 months, 16 (8%) patients were assessed. At each time point, a significant reduction was observed in the extent of COVID-19-associated opacities (p < 0.001 and p = 0.002) and of parenchymal bands (p = 0.014 and p = 0.025). Persisting fibrotic-like changes were observed in 18 (9%) patients (apical findings in 2 patients, fibrotic non-specific interstitial pneumonia-like changes in 14 patients, minimal fibrotic changes in 2 patients). At 3 months, the predicted median FVC% was 93% (80-100%) and the predicted DLCO% was 65% (58-78%) with a statistically significant improvement at 6 months in both (p = 0.001). Moreover, 81.1% had mMRC ≤ 1 and the median SGRQ was 11.65 [0-24.3] with a significant reduction at 6 months in both dyspnea (p < 0.001) and SGRQ (p = 0.027) persisting at 12 months. Conclusions: This prospective study, including only survivors of severe/critical COVID-19 pneumonia, documented the significant improvement in all imaging, functional, and clinical parameters by applying the "non-interventional" approach. These data do not indicate any post-COVID-19 severe/critical pneumonia and "epidemic of widespread pulmonary fibrosis".
Collapse
Affiliation(s)
- Elvira-Markela Antonogiannaki
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Effrosyni D. Manali
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Konstantinos Thomas
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Maria Kallieri
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Panagiota Alexopoulou
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Andriana I. Papaioannou
- 1st Respiratory Department, Athens Chest Hospital “Sotiria”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Spyridon Prountzos
- 2nd Department of Radiology, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (C.K.)
| | - Anastasia Karachaliou
- 2nd Department of Radiology, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (C.K.)
| | - Christina Kontopoulou
- 2nd Department of Radiology, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (S.P.); (A.K.); (C.K.)
| | - Vagia Karageorgou
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Stefanos Lampadakis
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Myrto Blizou
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Ioannis Tomos
- 5th Respiratory Department, Athens Chest Hospital “Sotiria”, 11527 Athens, Greece;
| | - Sotiria Grigoropoulou
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Dimitra Kavatha
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Stelios Loukides
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (I.G.); (K.T.); (P.A.); (S.G.); (D.K.); (A.A.)
| | - Spyros A. Papiris
- 2nd Pulmonary Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece; (E.-M.A.); (E.D.M.); (M.K.); (V.K.); (S.L.); (M.B.); (S.A.P.)
| |
Collapse
|
26
|
Yasseen BA, Elkhodiry AA, El-sayed H, Zidan M, Kamel AG, Badawy MA, Hamza MS, El-Messiery RM, El Ansary M, Abdel-Rahman EA, Ali SS. The role of neutrophilia in hyperlactatemia, blood acidosis, impaired oxygen transport, and mortality outcome in critically ill COVID-19 patients. Front Mol Biosci 2025; 11:1510592. [PMID: 39834785 PMCID: PMC11743367 DOI: 10.3389/fmolb.2024.1510592] [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: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction COVID-19 severity and high in-hospital mortality are often associated with severe hypoxemia, hyperlactatemia, and acidosis, yet the key players driving this association remain unclear. It is generally assumed that organ damage causes toxic acidosis, but since neutrophil numbers in severe COVID-19 can exceed 80% of the total circulating leukocytes, we asked if metabolic acidosis mediated by the glycolytic neutrophils is associated with lung damage and impaired oxygen delivery in critically ill patients. Methods Based on prospective mortality outcome, critically ill COVID-19 patients were divided into ICU- survivors and ICU-non-survivors. Samples were analyzed to explore if correlations exist between neutrophil counts, lung damage, glycolysis, blood lactate, blood pH, hemoglobin oxygen saturation, and mortality outcome. We also interrogated isolated neutrophils, platelets, and PBMCs for glycolytic activities. Results Arterial blood gas analyses showed remarkable hypoxemia in non-survivors with no consistent differences in PCO2 or [HCO3 -]. The hemoglobin oxygen dissociation curve revealed a right-shift, consistent with lower blood-pH and elevated blood lactate in non-survivors. Metabolic analysis of different blood cells revealed increased glycolytic activity only when considering the total number of neutrophils. Conclusion This indicates the role of neutrophilia in hyperlactatemia and lung damage, subsequently contributing to mortality outcomes in severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Basma A. Yasseen
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Aya A. Elkhodiry
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Hajar El-sayed
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Mona Zidan
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Azza G. Kamel
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | | | - Marwa S. Hamza
- Department of Clinical Pharmacy Practice, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Riem M. El-Messiery
- Infectious Disease Unit, Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed El Ansary
- Department of Intensive Care, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Engy A. Abdel-Rahman
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
- Pharmacology Department, Faculty of Medicine, Assuit University, Assuit, Egypt
| | - Sameh S. Ali
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| |
Collapse
|
27
|
Imtiaz N, Stoddard WA, Day SW. Oxygen Transport in Nanoporous SiN Membrane Compared to PDMS and Polypropylene for Microfluidic ECMO. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.04.631337. [PMID: 39803510 PMCID: PMC11722228 DOI: 10.1101/2025.01.04.631337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Extracorporeal Membrane Oxygenation (ECMO) serves as a crucial intervention for patients with severe pulmonary dysfunction by facilitating oxygenation and carbon dioxide removal. While traditional ECMO systems are effective, their large priming volumes and significant blood-contacting surface areas can lead to complications, particularly in neonates and pediatric patients. Microfluidic ECMO systems offer a promising alternative by miniaturizing the ECMO technology, reducing blood volume requirements, and minimizing device surface area to improve safety and efficiency. This study investigates the oxygen transport performance of three membrane types- polydimethylsiloxane (PDMS), polypropylene, and a novel nanoporous silicon nitride (SiN) membrane-in a microfluidic ECMO platform. While nanoporous membranes rely on pore-mediated diffusion and PDMS on polymer lattice diffusion, results show no significant differences in device oxygenation efficiency (p > 0.05). Blood-side factors, including the diffusion rate of oxygen through the red blood cell (RBC) membrane, RBC residence time, and hemoglobin binding kinetics, were identified as primary bottlenecks. Even computational models of a hypothetical infinitely permeable membrane corroborate the limited impact of membrane material. These findings suggest a shift in ECMO design priorities from membrane material to blood-side enhancements. This research provides a foundation for optimizing ECMO systems.
Collapse
Affiliation(s)
- Nayeem Imtiaz
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY 14623, USA
| | - William A Stoddard
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY 14623, USA
| | - Steven W Day
- Rochester Institute of Technology, Kate Gleason College of Engineering, Rochester, NY 14623, USA
| |
Collapse
|
28
|
Mf Z, DA PN, C L, Sp N, Ad D, J W, Yf K, G S. Supplemental oxygen prescriptions after hospitalization for coronavirus disease 2019. Heart Lung 2025; 69:208-216. [PMID: 39500209 DOI: 10.1016/j.hrtlng.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND The role of home oxygen therapy for patients recovering from Coronavirus Disease 2019 (COVID-19) pneumonia, characterized by impaired gas exchange, is not well-defined. OBJECTIVES To compare the characteristics, duration, odds of receiving, and continuing to receive home oxygen prescriptions between patients discharged home after COVID-19 pneumonia hospitalization and those discharged after non-COVID-19 pneumonia. METHODS From April 2020 to March 2021, 52,951 patients with COVID-19 pneumonia (53.6 % women, 64 % White) were identified, and from January 2019 to December 2019, 26,701 patients with non-COVID pneumonia (53.9 % women, 76.7 % White) were identified, using the Optum Clinformatics Data Mart Database. New oxygen prescriptions were identified through Healthcare Common Procedure Coding Systems codes. Propensity score matching adjusted for confounders, and Cox regression analysis was conducted to compare post-discharge oxygen use. RESULTS Following hospitalization, oxygen was prescribed to 52,951 patients with COVID-19 pneumonia and 26,701 patients with non-COVID pneumonia in the United States. The COVID-19 pneumonia group were four times more likely to be prescribed supplemental oxygen compared to the non-COVID-19 pneumonia group (OR 4.22; 95 % confidence interval [CI] 3.76-4.74). This trend persisted in sensitivity analyses: ICU patients (OR 4.05; 95 % CI 3.36-4.88) and those who received both ICU admission and mechanical ventilation (OR 3.84; 95 % CI 2.32-6.37). Hispanic patients had the highest likelihood of receiving a supplemental oxygen prescription after discharge (OR 6.75; 95 % CI 5.03-9.05). CONCLUSIONS Post-hospitalization, one in five patients with COVID-19 received prescriptions for supplemental oxygen, which was significantly higher than the proportion of patients with non-COVID-19 pneumonia.
Collapse
Affiliation(s)
- Zaidan Mf
- Division of Pulmonary Critical Care and Sleep Medicine, University of Texas Medical Branch, Galveston, TX, United States.
| | - Puebla Neira DA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ, United States
| | - Lau C
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Arizona College of Medicine Phoenix, Phoenix, AZ, United States
| | - Nishi Sp
- Division of Pulmonary Critical Care and Sleep Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Duarte Ad
- Division of Pulmonary Critical Care and Sleep Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Wang J
- Office of Biostatistics, University of Texas Medical Branch (UTMB), Galveston, TX
| | - Kuo Yf
- Office of Biostatistics, University of Texas Medical Branch (UTMB), Galveston, TX
| | - Sharma G
- Division of Pulmonary Critical Care and Sleep Medicine, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
29
|
Keller K, Sagoschen I, Schmitt VH, Sivanathan V, Farmakis IT, Hahad O, Koelmel S, Schmidt FP, Espinola-Klein C, Konstantinides S, Münzel T, Lurz P, Hobohm L. Risk factors for intensive care admission in childhood patients with COVID-19 - Results of the German nationwide inpatient sample. Respir Med 2025; 236:107880. [PMID: 39586334 DOI: 10.1016/j.rmed.2024.107880] [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: 06/27/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND COVID-19 pandemic research efforts have mainly focused on adults, but data in paediatric populations are sparse. METHODS We used the German nationwide-inpatient-sample analysing all hospitalized children ≤18 years with confirmed COVID-19-diagnosis in Germany during the year 2020, stratified for intensive care unit (ICU) admission. RESULTS Overall, 3360 hospitalization-cases of children ≤18 years with COVID-19-infection were diagnosed in Germany 2020 (median age 7.0 [IQR 0.0-15.0] years, 49.8 % female); among them 4.3 % were admitted on an ICU. In-hospital death occurred in five patients with and three without ICU admission (3.5 % vs. 0.1 %, P < 0.001) and ICU admission was independently associated with increased case-fatality (OR 21.573 [95%CI 4.191-111.044], P < 0.001). Obesity (OR 3.419 [95%CI 1.300-8.993], P = 0.013), diabetes mellitus (OR 6.929 [95%CI 3.327-14.432], P < 0.001), pneumonia (OR 7.373 [95%CI 4.823-11.271], P < 0.001), acute respiratory distress syndrome (ARDS) (OR 48.058 [95%CI 11.689-197.588], P < 0.001) and multisystem inflammatory syndrome caused by COVID-19 (OR 9.573 [95%CI 3.036-30.191], P < 0.001), heart failure (OR 64.509 [95%CI 24.462-170.121], P < 0.001), myocarditis (OR 4.682 [95%CI 1.278-17.149], P = 0.020), acute and/or chronic kidney failure (OR 7.946 [95%CI 3.606-17.508], P < 0.001), cancer (OR 5.220 [95%CI 2.599-10.485], P < 0.001) and liver diseases (OR 5.501 [95%CI 2.177-13.899], P < 0.001) were associated with an ICU admission. CONCLUSION Proportion of hospitalized paediatric COVID-19-patients admitted on ICU in Germany was low with 4.3 % accompanied by 3.5 % case-fatality rate. Independent factors for ICU admission comprised cardio-vascular risk factors, comorbidities, and complications of COVID-19.
Collapse
Affiliation(s)
- Karsten Keller
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Medical Clinic VII, Department of Sports Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Ingo Sagoschen
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Volker H Schmitt
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Visvakanth Sivanathan
- Department of Gastroenterology, University Medical Center Mainz (Johannes Gutenberg-University Mainz), Mainz, Germany
| | - Ioannis T Farmakis
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Sebastian Koelmel
- Department of Internal Medicine, Triemli Hospital Zurich, Zurich, Switzerland
| | | | - Christine Espinola-Klein
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece
| | - Thomas Münzel
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Philipp Lurz
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Lukas Hobohm
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
30
|
Rezaei M, Babamahmoodi A, Mirahmadi A, Bineshfar N, Mahmoudi S, Ghadimi S, Valizadeh M, Malakouti T, Taheri FT, Mohammadpour H, Azadani FN, Ziai SA, Poorhosseini SM, Marjani M. The Relationship between the Clinical Course of SARS-CoV-2 Infections and Expression of Bruton's Tyrosine Kinase. Infect Disord Drug Targets 2025; 25:e270624231361. [PMID: 38939988 DOI: 10.2174/0118715265301312240529044923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/23/2024] [Accepted: 04/17/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Bruton's Tyrosine Kinase (BTK), an important element for the production of several inflammatory cytokines, may play a role in the pathogenesis of COVID-19. This study aimed to assess BTK gene expression levels in COVID-19 cases based on disease severity and outcome. METHODS In this study, 33 hospitalized patients with COVID-19 were recruited and divided into two groups based on the severity of the disease: "mild to moderate" and "severe to critical". A blood sample was taken from each patient, peripheral blood mononuclear cells (PBMCs) were extracted, and BTK gene expression was measured. The level of BTK gene expression was compared based on the demographic data, laboratory results, and the severity and outcome of the disease. RESULTS Among the 33 patients, 22 (66.7%) were male, with nearly half having at least one underlying condition. The severity groups comprised 12 patients in the "mild to moderate" category and 21 in the "severe to critical" category, with eight (24.2%) experiencing fatal outcomes. Age, weight, and BMI showed no significant associations with BTK expression. BTK expression was notably lower in "severe to critical" and ICU-admitted cases, as well as in individuals with low O2 saturation. However, no significant difference in BTK expression was observed between cured and deceased patients (p = 0.117). CONCLUSION BTK gene expression in PBMCs exhibited an inverse correlation with COVID- 19 severity. However, no difference was found between BTK expression and disease outcome.
Collapse
Affiliation(s)
- Mitra Rezaei
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pathology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Clinical Tuberculosis and Epidemiology Research Center, `National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Babamahmoodi
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Clinical Tuberculosis and Epidemiology Research Center, `National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Mirahmadi
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloufar Bineshfar
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Mahmoudi
- Pediatric Infectious Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Somayeh Ghadimi
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Melika Valizadeh
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tannaz Malakouti
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hadiseh Mohammadpour
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farinaz Nasr Azadani
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Poorhosseini
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Majid Marjani
- Clinical Tuberculosis and Epidemiology Research Center, `National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Chavez-Tapia N, Sayeed MA, Luxmi S, Kasper DJ, Xue F, Shen Y, Fan W, Yuan W, Du B. Safety and efficacy of selective RIPK1 inhibitor SIR1-365 in hospitalized patients with severe COVID-19: A multicenter, randomized, double-blind, phase 1b trial. JOURNAL OF INTENSIVE MEDICINE 2025; 5:70-78. [PMID: 39872839 PMCID: PMC11763862 DOI: 10.1016/j.jointm.2024.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/26/2024] [Accepted: 07/29/2024] [Indexed: 01/30/2025]
Abstract
Background Receptor-interacting protein kinase 1 (RIPK1), a serine/threonine protein kinase, is mainly activated by pro-inflammatory cytokines and pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and its activation could result in apoptosis, necroptosis, or inflammation. This study was conducted to evaluate the safety and efficacy of a potent and selective inhibitor of RIPK1, SIR1-365, in hospitalized patients with severe coronavirus disease 2019 (COVID-19). Methods This multicenter, randomized, double-blind, phase 1b study screened patients from December 18, 2020 until November 27, 2021. Adults hospitalized with severe COVID-19 (diagnosed ≤2 weeks before screening) were randomized 1:1 to receive oral placebo or SIR1-365 100 mg three times daily for ≤14 consecutive days, with standard-of-care. The primary objective was to evaluate SIR1-365 safety and tolerability. Secondary objectives included an assessment of SIR1-365 efficacy. Descriptive statistics were used to summarize safety. The study was not powered for efficacy testing. Relevant inferential statistical tests were used to aid interpretation of differences in clinical efficacy. Results Forty-five patients were randomized, 42 were treated. Eighteen patients experienced treatment-emergent adverse events (TEAEs) and 7 patients were ≥ grade 3. Fewer SIR1-365-treated vs. placebo-treated patients experienced TEAEs (30.4% vs. 57.9%) and serious TEAEs (13.0% vs. 26.3%) within 28 days of the first dose. There were no serious treatment-related TEAEs or deaths. Compare to placebo, SIR1-365 significantly increased arterial oxygenation from baseline to day 7 (least-squares mean change [standard error]: 109.4 [26.4] vs. -24.2 [23.6]; P=0.0095), significantly reduced hospitalization duration after treatment (mean±standard deviation: [4.7±3.7] days vs. [8.6±5.6] days; P=0.0145) and respiratory failure incidence (8.3% vs. 38.1%; two-sided P=0.0291) during the study, and numerically shortened the time to clinical improvement in World Health Organization ordinal scale (median: 5.0 days vs. 9.0 days, P=0.0766). Conclusions SIR1-365 was well tolerated and demonstrated a trend toward quicker recovery than placebo in hospitalized patients with severe COVID-19.Trial Registration ClinicalTrials.gov number: NCT04622332.
Collapse
Affiliation(s)
- Norberto Chavez-Tapia
- Obesity and Digestive Disease Unit, Medica Sur Clinic and Foundation, Mexico City, Mexico
| | - Muneeba Ahsan Sayeed
- Sindh Infectious Diseases Hospital and Research Centre, Dow University of Health Sciences, Karachi, Sindh, Pakistan
| | - Shobha Luxmi
- Sindh Infectious Diseases Hospital and Research Centre, Dow University of Health Sciences, Karachi, Sindh, Pakistan
| | - Douglas J. Kasper
- Department of Medicine, Division of Infectious Disease, University of Illinois School of Medicine, OSF HealthCare System d/b/a Saint Francis Medical Center, Peoria, IL, USA
| | | | | | | | - Wei Yuan
- Sironax USA, Inc., Edison, NJ, USA
| | - Bin Du
- Medical Intensive Care Unit, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
32
|
Mahrokhian SH, Tostanoski LH, Vidal SJ, Barouch DH. COVID-19 vaccines: Immune correlates and clinical outcomes. Hum Vaccin Immunother 2024; 20:2324549. [PMID: 38517241 PMCID: PMC10962618 DOI: 10.1080/21645515.2024.2324549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/24/2024] [Indexed: 03/23/2024] Open
Abstract
Severe disease due to COVID-19 has declined dramatically as a result of widespread vaccination and natural immunity in the population. With the emergence of SARS-CoV-2 variants that largely escape vaccine-elicited neutralizing antibody responses, the efficacy of the original vaccines has waned and has required vaccine updating and boosting. Nevertheless, hospitalizations and deaths due to COVID-19 have remained low. In this review, we summarize current knowledge of immune responses that contribute to population immunity and the mechanisms how vaccines attenuate COVID-19 disease severity.
Collapse
Affiliation(s)
- Shant H. Mahrokhian
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Lisa H. Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Samuel J. Vidal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
33
|
Su TH, Lu YC, Li JW, Liu WD, Lee MS, Hung CS, Chou CF, Wang JT, Ho YL, Kao JH. Telemedicine facilitates medical care for hospitalized patients in quarantine. J Formos Med Assoc 2024:S0929-6646(24)00575-8. [PMID: 39690047 DOI: 10.1016/j.jfma.2024.12.016] [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: 09/20/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND/PURPOSE Telemedicine is an innovative medical care system that facilitates visual communication between patients and healthcare workers (HCWs), mainly in a community-based setting. We investigated the clinical effectiveness of in-patient telemedicine care for patients in quarantine for the Coronavirus disease 2019 (COVID-19). METHODS We conducted a prospective study to include patients with mild-to-moderate severity of COVID-19 and their primary-care HCWs. Patients were stratified into the "telemedicine" group and the "standard-care" group. The telemedicine group received additional telemedicine communications and wearable devices for vital signs monitoring during hospitalization. The primary endpoint was the usability of the telemedicine system assessed by the modified telehealth usability questionnaire (TUQ) on a 7-point ordinal Likert scale. The secondary endpoints were the outcomes of COVID-19. RESULTS Overall, we included 30 and 48 patients in the respective standard-care group and telemedicine group. In addition, 31 primary care residents and 56 nurses were included in the HCW groups. For all participants, the modified TUQ responses in the categories of usefulness (6.2), ease of use (6.1), effectiveness (6.0), and satisfaction (6.1) were significantly greater than 5 (somewhat agree), except for the reliability sectors (5.3). The telemedicine group liked the telemedicine system significantly more than the HCWs. The doctors scored significantly higher than the nurses in the categories of effectiveness and satisfaction of the telemedicine systems. The outcomes of COVID-19 were comparable between the telemedicine and the standard-care group. CONCLUSIONS Our study demonstrated the clinical effectiveness and satisfaction of the telemedicine for the care of quarantined COVID-19 patients during hospitalization.
Collapse
Affiliation(s)
- Tung-Hung Su
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Cheng Lu
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jia-Wei Li
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Wang-Da Liu
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ming-Sui Lee
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Fu Chou
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan
| | - Jann-Tay Wang
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Lwun Ho
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jia-Horng Kao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan; Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
34
|
Woodhouse EW, McClain MT, Woods CW. Harnessing the host response for precision infectious disease diagnosis. Clin Microbiol Rev 2024; 37:e0007824. [PMID: 39404266 PMCID: PMC11629621 DOI: 10.1128/cmr.00078-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024] Open
Abstract
SUMMARYDetection of the presence of infection and its etiology must be accurate and timely to facilitate appropriate antimicrobial use. Diagnostic strategies that rely solely on pathogen detection often are insufficient due to poor test characteristics, inability to differentiate colonization from infection, or protracted delay to result. Understanding the human response across different pathogens on a clinical and molecular level can provide more accurate, timely, and useful answers, especially in critical illness and diagnostic uncertainty. Improvements in understanding the human immune response including genomics, protein analysis, gene expression, and cellular morphology have led to rapid innovation of new host response-based diagnostic tests. This review describes the limitations of pathogen-focused technology and the benefits of examining the breadth of immune response to diagnose infection. It then explores biomarkers that have been studied for this purpose and scrutinizes the performance of host-based multianalyte testing. Currently cleared diagnostics and those in late-stage development are described in depth, with a focus on the purpose of testing and its utility for clinicians. Finally, it concludes by examining opportunities for further host response-derived diagnostic innovation.
Collapse
Affiliation(s)
- E. Wilbur Woodhouse
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Micah T. McClain
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Christopher W. Woods
- Department of Medicine, Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, North Carolina, USA
- Section of Infectious Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| |
Collapse
|
35
|
Fernández-Pedruelo D, Juárez-Vela R, Ruiz de Viñaspre-Hernández R, Alonso-Alonso J, Criado-Gutiérrez JM, Sancho-Sánchez C. Influence of smoking and obesity on post-COVID-19 sequelae and risk of hospitalization. Front Med (Lausanne) 2024; 11:1499239. [PMID: 39703524 PMCID: PMC11655222 DOI: 10.3389/fmed.2024.1499239] [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: 09/20/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has significantly impacted the global healthcare system, with particularly harmful effects on the human respiratory system. Beyond the acute symptoms, there is growing concern about persistent symptoms that last for weeks or months after the initial infection, known as long COVID syndrome. This study focuses on investigating the relationship between smoking, obesity, and the presence of post-COVID-19 sequelae, as well as their influence on the risk of hospitalization. Materials and methods An observational and retrospective study was conducted using medical records of patients diagnosed with COVID-19 in Castilla y León, Spain, between November 1 and 30, 2020. The patients were divided into three groups: smoking (current and former), obesity/overweight, and control group. Various variables were analyzed, including age, sex, and the presence of post-COVID-19 sequelae, chronic pathologies, cardiovascular diseases, psychological conditions, and hospitalization. Descriptive statistics and Odds Ratio analysis were used for comparisons. Results The results revealed that obesity was significantly associated with a higher risk of post-COVID-19 sequelae, particularly memory disorders and neurological, mental, or psychological symptoms. In contrast, smoking was correlated with an increase in memory problems but did not show a direct influence on post-COVID-19 sequelae or hospitalization. Additionally, women were found to have a higher prevalence of obesity in the studied population. Conclusion This study provides evidence that obesity increases the risk of post-COVID-19 sequelae, especially in terms of memory disorders and neuropsychological symptoms. On the other hand, smoking is related to memory problems. Regarding cardiovascular pathologies, there was not enough statistical evidence for analysis, while for hospitalization, it was determined that smoking and obesity do not have a direct influence on these post-COVID consequences.
Collapse
Affiliation(s)
- Daniel Fernández-Pedruelo
- Doctoral Program in Health, Disability, Dependency, and Well-being, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Raúl Juárez-Vela
- Deparment of Nursing, Faculty of Health Sciences, University of La Rioja, Logroño, Spain
| | | | | | | | - Consuelo Sancho-Sánchez
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| |
Collapse
|
36
|
Sun J, Peters M, Yu LR, Vijay V, Bidarimath M, Agrawal M, Flores-Torres AS, Green AM, Burkhart K, Oliphant J, Smallwood HS, Beger RD. Untargeted metabolomics and lipidomics in COVID-19 patient plasma reveals disease severity biomarkers. Metabolomics 2024; 21:3. [PMID: 39636373 DOI: 10.1007/s11306-024-02195-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Coronavirus disease 2019 (COVID-19) has widely varying clinical severity. Currently, no single marker or panel of markers is considered standard of care for prediction of COVID-19 disease progression. The goal of this study is to gain mechanistic insights at the molecular level and to discover predictive biomarkers of severity of infection and outcomes among COVID-19 patients. METHOD This cohort study (n = 76) included participants aged 16-78 years who tested positive for SARS-CoV-2 and enrolled in Memphis, TN between August 2020 to July 2022. Clinical outcomes were classified as Non-severe (n = 39) or Severe (n = 37). LC/HRMS-based untargeted metabolomics/lipidomics was conducted to examine the difference in plasma metabolome and lipidome between the two groups. RESULTS Metabolomics data indicated that the kynurenine pathway was activated in Severe participants. Significant increases in short chain acylcarnitines, and short and medium chain acylcarnitines containing OH-FA chain in Severe vs. Non-severe group, which indicates that (1) the energy pathway switched to FA β-oxidation to maintain the host energy homeostasis and to provide energy for virus proliferation; (2) ROS status was aggravated in Severe vs. Non-severe group. Based on PLS-DA and correlation analysis to severity score, IL-6, and creatine, a biomarker panel containing glucose (pro-inflammation), ceramide and S1P (inflammation related), 4-hydroxybutyric acid (oxidative stress related), testosterone sulfate (immune related), and creatine (kidney function), was discovered. This novel biomarker panel plus IL-6 with an AUC of 0.945 provides a better indication of COVID-19 clinical outcomes than that of IL-6 alone or the three clinical biomarker panel (IL-6, glucose and creatine) with AUCs of 0.875 or 0.892.
Collapse
Affiliation(s)
- Jinchun Sun
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Megan Peters
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Li-Rong Yu
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Vikrant Vijay
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Mallikarjun Bidarimath
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Mona Agrawal
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, USA
| | | | - Amanda M Green
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, USA
- Department of Infectious Disease, St Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Keith Burkhart
- Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, USA
| | - Jessica Oliphant
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, USA
- Children's Foundation Research Institute, Memphis, TN, 38105, USA
| | - Richard D Beger
- Division of Systems Biology, National Center for Toxicological Research, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| |
Collapse
|
37
|
Jayasimhan D, Matthay MA. Definitions of Acute Respiratory Distress Syndrome: Present Recommendations and Challenges. Clin Chest Med 2024; 45:785-795. [PMID: 39442997 DOI: 10.1016/j.ccm.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory process resulting in diffuse lung injury precipitated by an underlying risk factor. However, current definitions may pose barriers to the accurate diagnosis of this syndrome. These include changes in risk factors and associated disease evolution of ARDS, changes in contemporary clinical practice, and access to diagnostic tools required to diagnose ARDS in resource-limited settings. A consensus conference has proposed changes for an expanded global definition of ARDS. In this review article, we review challenges in defining ARDS and present recommendations of the global definition of ARDS.
Collapse
Affiliation(s)
- Dilip Jayasimhan
- Intensive Care Unit, Wellington Regional Hospital, Te Whatu Ora Health New Zealand- Capital, Coast and Hutt Valley, 49 Riddiford Street, Wellington 6021, New Zealand
| | - Michael A Matthay
- Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA; Department of Anesthesia, University of California-San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California-San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
38
|
López-Vinasco AM, Montero-Vargas JM, García-Guillén MDL, De la Peña-Hernández LDJ, Teran LM. Clinical characteristics of adult asthma patients hospitalized by COVID-19 in Mexico City: a real-world study. Ann Med 2024; 56:2424448. [PMID: 39623785 PMCID: PMC11616761 DOI: 10.1080/07853890.2024.2424448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 06/03/2024] [Accepted: 10/18/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic raised concerns about whether individuals with chronic respiratory diseases, such as asthma, were at higher risk of severe outcomes. Although several studies were published on this topic, not all included asthma as a risk factor. Therefore, describing the clinical characteristics of COVID-19-infected asthma patients in a specialized respiratory center is valuable as a real-life study. OBJECTIVE To investigate the clinical characteristics and disease severity in SARS-CoV-2-infected adults with pre-existing asthma hospitalized at the National Institute of Respiratory Diseases (INER) in Mexico City. METHODS We conducted a retrospective, observational study on adults with confirmed COVID-19 hospitalized from March 2020 to June 2021. Out of 2,249 reviewed medical records, we identified asthmatic patients and compared them with a matched non-asthmatic control group to assess asthma's impact on COVID-19 severity and outcomes. RESULTS Based on the clinical records, asthma prevalence among hospitalized patients was low (1.51%); of these, 73% had allergic and 27% had non-allergic asthma. COVID-19 severity did not vary significantly between asthma phenotypes, although there was higher mortality among patients with non-allergic asthma. Most patients in both groups developed a severe form of the disease and higher mortality rates than non-asthmatics, though the differences were not statistically significant. CONCLUSION Asthma prevalence among patients with COVID-19 was low, but mortality was higher in asthma patients. Although the small sample size limits the generalizability of these findings, this study in a Mexican population hospitalized in a reference hospital provides insights for improving asthma management in future pandemics.
Collapse
Affiliation(s)
- Andrea Marcela López-Vinasco
- Departamento de Investigación en Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
- Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Josaphat Miguel Montero-Vargas
- Departamento de Investigación en Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Ma. de Lourdes García-Guillén
- Departamento de Investigación en Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Leonardo de Jesús De la Peña-Hernández
- Departamento de Investigación en Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Luis M. Teran
- Departamento de Investigación en Inmunogenética y Alergia, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
- Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
39
|
Tsiftsoglou SA, Gavriilaki E. A potential bimodal interplay between heme and complement factor H 402H in the deregulation of the complement alternative pathway by SARS-CoV-2. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 126:105698. [PMID: 39643072 DOI: 10.1016/j.meegid.2024.105698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
The recent discovery that the trimeric SARS-CoV-2 spike S glycoprotein carries heme within an NTD domain pocket of the S1 subunits, suggested that this virus may be cleverly utilizing heme, in addition to the S1 RBD domains, for invading target cells carrying a specific entry receptor like ACE2, TMEM106B and others. Studies during the COVID-19 pandemic revealed that the infectivity of this virus depends on cell surface heparan sulfate and that the infection induces non-canonical activation of the Complement Alternative pathway (AP) on the surface of infected cells. In our recent COVID-19 genomic studies, among the coding SNPs of interest we also detected the presence of the CFH rs1061170, rs800292 and rs1065489 within all the infected patient subgroups examined. The minor C allele of rs1061170 encodes CFH 402H that over the years has been associated with diseases characterized by complement dysregulation namely the age-related macular degeneration (AMD) and the atypical haemolytic uremic syndrome (aHUS). Also, more recently with the diminishment of CD4+ T cell responses with ageing. The rs800292 minor allele A encodes CFH 62I that supports enhanced cofactor activity for Complement factor I (CFI). Also, the rs1065489 minor allele T encodes CFH 936D and is located within the CCP16 domain that influences the affinity of CFH with extracellular laminins. A subsequent computational analysis revealed that the CFH residue 402 is located centrally within a heme-binding motif (HBM) in domain CCP7 (398YNQNYGRKF406). Heme on the viral spike glycoprotein S1 subunit could recruit CFH 402H for masking free viral particles from opsonisation, and when in proximity to cell surface, act as a bait disrupting CFH 402H from the heparan sulphate coat of the target cells. Publicly available genetic data for European populations indicate that the minor C allele of rs1061170 is present only in haplotypes that carry the major alleles of rs800292 and rs1065489. This combination encodes for CFH 402H that exhibits increased biochemical affinity for heme in proximity, without enhanced cofactor activity for CFI and weaker association with the extracellular matrix. In the theatre of infection, this combination can promote heme-mediated viral infection with weaker complement opsonisation and potential AP deregulation. This strategy may be evolutionary conserved among various classes of infectious agents.
Collapse
Affiliation(s)
- Stefanos A Tsiftsoglou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Department of Biomedical Sciences, School of Health Sciences, Alexander Campus, International Hellenic University, Sindos, 57400, Greece.
| | - Eleni Gavriilaki
- 2(nd) Propedeutic Department of Internal Medicine, Hippocration General Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
40
|
Almazroo R, Almazroa H. Associations between personality traits and emotional experiences as predictors of obesity during the COVID-19 pandemic: A cross-sectional study. Heliyon 2024; 10:e40002. [PMID: 39605822 PMCID: PMC11600080 DOI: 10.1016/j.heliyon.2024.e40002] [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: 08/13/2023] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
This quantitative cross-sectional descriptive research aimed to investigate the relationship between personality traits, emotions, and obesity among Saudi adults during the COVID-19 pandemic. The study recruited 383 participants from Riyadh using non-probability sampling. A validated online survey, including the PANAS-SF and BFI-10 measures, was used and hosted on Qualtrics. Descriptive and multiple regression analyses were performed on the data. The study found that conscientiousness was the highest personality trait among the sample, with an average score of 7.74, while the mean for openness was the lowest. The incidence of obesity was high among participants, with a rate of 29.2 %. Furthermore, there was a significant relationship between negative affect and obesity (p = 0.033). Based on these findings, public health specialists and health promoters should design programs using the socio-ecological model as a guide to address the important factors at every level of the model.
Collapse
Affiliation(s)
| | - Hiya Almazroa
- Department of Teaching and Learning, College of education and Human Development, Princess Nourah bint Abdulrahman University, Saudi Arabia
| |
Collapse
|
41
|
Gao L, Liu Y, He QY, Wang Y, Jiang YL, Yang J, Fu L, Zhao H. Serum transgelin is a novel prognostic biomarker for COVID-19 patients. Front Immunol 2024; 15:1423182. [PMID: 39676863 PMCID: PMC11638039 DOI: 10.3389/fimmu.2024.1423182] [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: 04/26/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Background Transgelin is a central actin-binding protein of the calponin family and involved in the process of multiple pulmonary diseases. Nevertheless, the role of transgelin in Coronavirus disease 2019 (COVID-19) patients is confusing. Methods All 317 COVID-19 patients were recruited from two hospital. Peripheral blood was collected from the fasting patients at the onset and convalescent phases. Demographic data and clinical information were obtained. The expression of serum transgelin was estimated using ELISA. Results The expression of serum transgelin on admission was gradually elevated in parallel with the increased severity scores of COVID-19. After treatment, serum transgelin expression was reduced during the convalescent phase. Spearman correlative analyses found that serum transgelin expression was closely correlated to lots of clinical parameters. Besides, serum transgelin was positively associated with severity scores. Follow-up research found that serum higher transgelin on admission elevated the risks of mechanical ventilation, vasoactive agent utilization, ICU admission, death, and longer hospital stays during hospitalization through a prospective cohort study. Additionally, there were similarly predictive capacities for critical patients and death between serum transgelin on admission and severity scores among COVID-19 patients. Conclusions The expression of serum transgelin is positively with the severity and poorly prognostic outcomes among COVID-19 patients, indicating that transgelin is implicated in the pathological process of COVID-19. Transgelin can assist in the risk stratification and revealing the pathological mechanisms of COVID-19.
Collapse
Affiliation(s)
- Lei Gao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ying Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qi-Yuan He
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ya-Lin Jiang
- Department of Respiratory and Critical Care Medicine, Bozhou People’s Hospital, Bozhou, Anhui, China
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
42
|
Karcioglu O, Akman C, Ozturk GA. Prothrombotic state and thrombotic events in COVID-19 pandemic period, including portal vein and splenic artery thromboses. World J Clin Cases 2024; 12:6595-6603. [PMID: 39600474 PMCID: PMC11514335 DOI: 10.12998/wjcc.v12.i33.6595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024] Open
Abstract
This editorial article is intended to perform a discussion on the manuscript entitled "Simultaneous portal vein thrombosis and splenic vein thrombosis in a COVID-19 patient: A case report and review of literature" written by Abramowitz et al. The article focuses on the diagnostic processes in a 77-year-old-male patient with a simultaneous portal vein and splenic artery thrombosis accompanying coronavirus disease 2019 (COVID-19). The authors postulated that splanchnic thrombosis should be on the list of differential diagnoses in a patient presenting with abdominal pain in presence of a COVID-19 infection. The tendency for venous and arterial thrombosis in COVID-19 patients is encountered, largely attributed to hypercoagulopathy. In general, venous thromboembolism mostly manifest as deep vein thrombosis (DVT), pulmonary embolism (PE) or catheter-related thromboembolic events. Acute PE, DVT, cerebrovascular events and myocardial infarction are seen as the most common thromboembolic complications in COVID-19 patients. COVID-19-associated hemostatic abnormalities include mild thrombocytopenia and increased D-dimer level. Similar to other coagulopathies, the treatment of the underlying condition is the mainstay. Addition of antiplatelet agents can be considered in critically ill patients at low bleeding risk, not on therapeutic anticoagulation, and receiving gastric acid suppression Early administration of antithrombotic drugs will have a beneficial effect in both the prevention and treatment of thrombotic events, especially in non-ambulatory patients. Low molecular weight heparin (LMWH) should be started if there is no contraindication, including in non-critical patients who are at risk of hospitalization LMWH (enoxaparin) is preferred to standard heparin.
Collapse
Affiliation(s)
- Ozgur Karcioglu
- Department of Emergency Medicine, University of Health Sciences, Istanbul Education and Research Hospital, Istanbul 34140, Bakırkoy, Türkiye
| | - Canan Akman
- Department of Emergency Medicine, Canakkale Onsekiz Mart University, Canakkale 17000, Çanakkale, Türkiye
| | - Göksu Afacan Ozturk
- Department of Emergency Medicine, Istanbul Aydin University, Istanbul 34295, Kucukcekmece, Istanbul, Türkiye
| |
Collapse
|
43
|
Laikitmongkhon J, Tassaneyasin T, Sutherasan Y, Phuphuakrat A, Srichatrapimuk S, Petnak T, Eksombatchai D, Thammavaranucupt K, Sungkanuparph S. A comparative study between methylprednisolone versus dexamethasone as an initial anti-inflammatory treatment of moderate COVID-19 pneumonia: an open-label randomized controlled trial. BMC Pulm Med 2024; 24:562. [PMID: 39529039 PMCID: PMC11555798 DOI: 10.1186/s12890-024-03364-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The most appropriate anti-inflammatory treatment for moderate COVID-19 pneumonia remains uncertain. We aimed to compare the effectiveness of a high-dose methylprednisolone versus a high-dose dexamethasone in hospitalized moderate COVID-19 pneumonia, regarding the WHO clinical progression scales, mortality, and the length of hospitalization. METHODS In this open-labeled randomized controlled trial, we enrolled patients with age > 18 years old who were diagnosed moderate COVID-19 pneumonia confirmed by real-time PCR, evidence of pneumonia by chest imaging and resting oxygen saturation between 90 and 94%. Patients were randomized at a 1:1 ratio to receive methylprednisolone 250 mg/day or dexamethasone 20 mg/day over the first three days. Then the patients in both groups received dexamethasone 20 mg/day on days 4-5, and 10 mg/day on days 6-10. Primary outcome was assessed by a 10-point WHO clinical progression scales ranging from uninfected (point 0) to death (point 10) on the fifth day of treatment. Secondary outcomes including 90-day mortality, length of hospitalization, rate of intensive care unit (ICU) transfer and complications were determined. RESULTS Of 98 eligible patients, the mean age was 76.0 ± 13.3 years. The median date of illness at the time of randomization was 3 days (interquartile range 2, 5). Baseline clinical characteristics and severity did not differ between groups. The WHO clinical progression scales were similar between methylprednisolone and dexamethasone group at 5 and 10 days of treatment [4.84, (95% confidence interval(CI), 4.35-5.33) vs. 4.76 (95% CI, 4.27-5.25), p = 0.821 and 4.32 (95% CI, 3.83-4.81) vs. 3.80 (95% CI, 3.31-4.29), p = 0.140, respectively)]. Both groups did not differ in-hospital mortality, length of hospitalization, and rate of ICU transfer. There were also no differences in steroid-related complications between groups until 90 days of follow-up. CONCLUSIONS In patients with moderate COVID-19 pneumonia, initial anti-inflammatory treatment with 250 mg/day of methylprednisolone for three days does not yield better outcomes over high-dose dexamethasone. TRIAL REGISTRATION This study was registered at Thai Clinical Trials Registry on October 17, 2021, with the identifier TCTR20211017001.
Collapse
Affiliation(s)
- Jakkrit Laikitmongkhon
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Tanapat Tassaneyasin
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan, Thailand
| | - Yuda Sutherasan
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.
| | - Angsana Phuphuakrat
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sirawat Srichatrapimuk
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan, Thailand
| | - Tananchai Petnak
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Dararat Eksombatchai
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Kanin Thammavaranucupt
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan, Thailand
| | - Somnuek Sungkanuparph
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan, Thailand
| |
Collapse
|
44
|
Santos-de-Araújo AD, Oliveira MR, Pontes-Silva A, Rodrigues LN, Costa CPS, Marinho RS, de Sousa Dos Santos S, Arena R, Phillips SA, Bassi-Dibai D, Borghi-Silva A. Inter- and intra-examiner reliability of short-term measurement of heart rate variability on rest in patients hospitalized with COVID-19. Sci Rep 2024; 14:26622. [PMID: 39496768 PMCID: PMC11535482 DOI: 10.1038/s41598-024-77558-5] [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: 02/15/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Measures reflecting cardiac sympathovagal activity, particularly those associated with heart rate variability (HRV), are widely recognized and utilized in both scientific and clinical contexts. This study aimed to assess the inter- and intra-examiner reliability of short-term HRV parameters in patients hospitalized with coronavirus disease 2019 (COVID-19). A total of 103 patients (both sexes) diagnosed with COVID-19 were included in the study. HRV was analyzed using both linear and nonlinear methods. Reliability was evaluated through intraclass correlation coefficient (ICC2.1), minimum detectable change (MDC), standard error of measurement (SEM), and coefficient of variation (CV). According to Fleiss' criteria, excellent reliability was demonstrated, with ICC values ranging from 0.970 to 0.999 for Examiner 1, and from 0.956 to 0.999, for Examiner 2. In the inter-examiner analysis, the ICCs of HRV parameters ranged from 0.972 to 0.999. SEM values for intra-examiner reliability for Examiner 1 ranged from 0.02 to 5.64, with MDC values from 0.05 to 15.64, and CV (%) from 0.28 to 8.04. For Examiner 2, SEM values ranged from 0.02 to 8.18, MDC values from 0.05 to 22.68, and CV (%) from 0.24 to 8.14. For inter-examiner reliability, SEM values ranged from 0.02 to 6.17, MDC from 0.06 to 17.11, and CV (%) from 0.34 to 9.81. Across all analyses, CVs for HRV parameters remained below 10%. Considering different time points and different examiners, short-term resting HRV measurements in patients hospitalized with COVID-19, as evaluated using a portable heart rate device, exhibit high reliability.
Collapse
Affiliation(s)
| | | | - André Pontes-Silva
- Physical Therapy Departarment, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | | | | | - Renan Shida Marinho
- Physical Therapy Departarment, Universidade Federal de São Carlos, São Carlos, SP, Brazil
| | | | - Ross Arena
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois, Chicago, Chicago, USA
| | - Shane A Phillips
- Department of Physical Therapy, College of Applied Health Sciences, University of Illinois, Chicago, Chicago, USA
| | - Daniela Bassi-Dibai
- Postgraduate Program in Management in Health Programs and Services, Universidade CEUMA, São Luís, MA, Brazil
| | - Audrey Borghi-Silva
- Physical Therapy Departarment, Universidade Federal de São Carlos, São Carlos, SP, Brazil.
- Cardiopulmonary Physiotherapy Laboratory, Physical Therapy Departament, Universidade Federal de São Carlos, Rodovia Washington Luiz, Postal Code - 13565- 905, São Carlos, SP, Brazil.
| |
Collapse
|
45
|
Rubens M, Saxena A, Ramamoorthy V, Appunni S, Ahmed MA, Zhang Z, Zhang Y, Sha R, Fahmy S. Impact of Frailty on COVID-19 Hospitalizations: Results from the California State Inpatient Database. South Med J 2024; 117:646-650. [PMID: 39486449 DOI: 10.14423/smj.0000000000001754] [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: 11/04/2024]
Abstract
OBJECTIVES Frail patients are at greater risk of experiencing adverse clinical outcomes in any critical illness due to decreased physiologic reserves, greater susceptibility to the adverse effects of treatment, and greater needs for intensive care. In this study, we sought to assess the prevalence of frailty and associated adverse in-hospital outcomes among coronavirus disease 2019 (COVID-19) hospitalizations using the 2020 California State Inpatient Database (SID). METHODS For this study, we conducted a retrospective analysis of data from all COVID-19 hospital patients aged 18 years and older. We identified hospitalizations that were at high risk of frailty using the Hospital Frailty Risk Score. The primary outcome of our study was in-hospital mortality, and the secondary outcomes were prolonged length of stay, vasopressor use, mechanical ventilation, and intensive care unit admission. RESULTS The prevalence of frailty was 44.3% among COVID-19 hospitalizations. Using propensity score matching analysis, we found that the odds of mortality (odds ratio [OR] 4.54, 95% confidence interval [CI] 4.28-4.82), prolonged length of stay (OR 2.81, 95% CI 2.70-2.90), vasopressor use (OR 8.65, 95% CI 7.45-10.03), mechanical ventilation (OR 6.90, 95% CI 6.47-7.35), and intensive care unit admission (OR 7.17, 95% CI 6.71-7.66) were significantly higher among the group of frail patients. CONCLUSION Our findings show that frailty could be used for assessing and risk stratifying patients for improved hospital outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Md Ashfaq Ahmed
- the Center for Advanced Analytics, Baptist Health South Florida, Miami
| | - Zhenwei Zhang
- the Center for Advanced Analytics, Baptist Health South Florida, Miami
| | - Yanjia Zhang
- the Center for Advanced Analytics, Baptist Health South Florida, Miami
| | - Rehan Sha
- the School for Advanced Studies, Miami, Florida
| | | |
Collapse
|
46
|
Michot JM, Dozio V, Rohmer J, Pommeret F, Roumier M, Yu H, Sklodowki K, Danlos FX, Ouali K, Kishazi E, Naigeon M, Griscelli F, Gachot B, Groh M, Bacciarello G, Stoclin A, Willekens C, Sakkal M, Bayle A, Zitvogel L, Silvin A, Soria JC, Barlesi F, Beeler K, André F, Vasse M, Chaput N, Ackermann F, Escher C, Marabelle A. Circulating Proteins Associated with Anti-IL6 Receptor Therapeutic Resistance in the Sera of Patients with Severe COVID-19. J Proteome Res 2024; 23:5001-5015. [PMID: 39352225 DOI: 10.1021/acs.jproteome.2c00422] [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
Circulating proteomes provide a snapshot of the physiological state of a human organism responding to pathogenic challenges and drug interventions. The outcomes of patients with COVID-19 and acute respiratory distress syndrome triggered by the SARS-CoV2 virus remain uncertain. Tocilizumab is an anti-interleukin-6 treatment that exerts encouraging clinical activity by controlling the cytokine storm and improving respiratory distress in patients with COVID-19. We investigate the biological determinants of therapeutic outcomes after tocilizumab treatment. Overall, 28 patients hospitalized due to severe COVID-19 who were treated with tocilizumab intravenously were included in this study. Sera were collected before and after tocilizumab, and the patient's outcome was evaluated until day 30 post-tocilizumab infusion for favorable therapeutic response to tocilizumab and mortality. Hyperreaction monitoring measurements by liquid chromatography-mass spectrometry-based proteomic analysis with data-independent acquisition quantified 510 proteins and 7019 peptides in the serum of patients. Alterations in the serum proteome reflect COVID-19 outcomes in patients treated with tocilizumab. Our results suggested that circulating proteins associated with the most significant prognostic impact belonged to the complement system, platelet degranulation, acute-phase proteins, and the Fc-epsilon receptor signaling pathway. Among these, upregulation of the complement system by activation of the classical pathway was associated with poor response to tocilizumab, and upregulation of Fc-epsilon receptor signaling was associated with lower mortality.
Collapse
Affiliation(s)
- Jean-Marie Michot
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Vito Dozio
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Julien Rohmer
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Fanny Pommeret
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Haochen Yu
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | | | - François-Xavier Danlos
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Kaissa Ouali
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Edina Kishazi
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Marie Naigeon
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
| | - Franck Griscelli
- Département de biologie et pathologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Bertrand Gachot
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Giulia Bacciarello
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Annabelle Stoclin
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Christophe Willekens
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Madona Sakkal
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Arnaud Bayle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Jean-Charles Soria
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Fabrice Barlesi
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Fabrice André
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Unité INSERM U981, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Marc Vasse
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
- Service de biologie clinique, Hôpital Foch, Suresnes 92150, France
- Unité INSERM U1176, Le Kremlin-Bicêtre, Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Nathalie Chaput
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Felix Ackermann
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | | | - Aurélien Marabelle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Centre d'investigation clinique - biothérapie, INSERM CICBT1428, Villejuif 94800, France
| |
Collapse
|
47
|
Giuliato ME, de Carvalho D, Baptistella AR. Functional status and quality of life after ICU discharge in severe COVID-19 patients. Respir Med 2024; 234:107810. [PMID: 39305966 DOI: 10.1016/j.rmed.2024.107810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE To assess the health status of individuals affected by COVID-19 after discharge from the ICU. METHOD Cross-sectional study, with patients discharged from the ICU due to severe COVID-19, in which Quality of Life (QoL) was assessed using the 12-Item SFHF, functionality using the Post-COVID-19 FSS, and the level of physical activity using the IPAQ. RESULTS Of the sixty patients, 51.7 % were male, with a mean age of 58 years. The physical component of QoL scored worse than the mental component and older patients had worse QoL in the physical component. These patients were shown to have low functionality scores and an irregularly active level of physical activity B. A lower level of physical activity was associated with individuals who remained in the prone position during hospitalization, while worse functionality was associated with the 70+ age group, although all age groups had functional losses. There was no association between QoL, functionality and level of physical activity and the clinical characteristics of the patients during hospitalization or the time they were discharged. CONCLUSION The majority of patients discharged from the ICU after severe COVID-19 have altered functional capacity, QoL and physical activity levels, which is not associated with the clinical characteristics during hospitalization.
Collapse
Affiliation(s)
- Márcia Eliane Giuliato
- University of the West of Santa Catarina (UNOESC), Joaçaba, SC, Brazil; Postgraduate Program in Biosciences and Health/(UNOESC), Joaçaba, SC, Brazil
| | - Diego de Carvalho
- University of the West of Santa Catarina (UNOESC), Joaçaba, SC, Brazil; Postgraduate Program in Biosciences and Health/(UNOESC), Joaçaba, SC, Brazil
| | - Antuani Rafael Baptistella
- University of the West of Santa Catarina (UNOESC), Joaçaba, SC, Brazil; Santa Terezinha University Hospital, Joaçaba, SC, Brazil; Postgraduate Program in Biosciences and Health/(UNOESC), Joaçaba, SC, Brazil.
| |
Collapse
|
48
|
Yoo JW, Kim WY, Chung CR, Cho YJ, Lee J, Jegal Y, Kim J, Joh JS, Park TY, Baek AR, Park JH, Chae G, Hwang JH, Song JW. Early pulmonary fibrosis-like changes between delta and pre-delta periods in patients with severe COVID-19 pneumonia on mechanical ventilation. Sci Rep 2024; 14:26101. [PMID: 39478105 PMCID: PMC11525473 DOI: 10.1038/s41598-024-77405-7] [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: 02/13/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
It remains unclear whether pulmonary fibrosis-like changes differ in patients with different SARS-CoV-2 variants. This study aimed to compare pulmonary fibrotic changes between two SARS-CoV-2 variant periods (delta vs. pre-delta) in critically ill patients with SARS-CoV-2 pneumonia. Clinical data and chest CT images of patients with SARS-CoV-2 pneumonia receiving mechanical ventilation were collected from 10 hospitals in South Korea over two periods: delta (July-December, 2021; n = 64) and pre-delta (February, 2020-June, 2021; n = 120). Fibrotic changes on chest CT were evaluated through visual assessment. Of 184 patients, the mean age was 64.6 years, and 60.5% were ale. Fibrosis-like changes on chest CT (median 51 days from enrollment to follow up CT scan, interquartile range 27-76 days) were identified in 75.3%. Delta group showed more fibrosis-like changes (≥ 2) (69.8% vs. 43.1%, P = 0.001) and more frequent reticulation and architectural distortion+/-parenchymal band than pre-delta group. Even after propensity score matching with clinical variables, delta group had more severe (≥ 2) fibrosis-like changes (71.4% vs. 38.8%, P = 0.001), and more frequent reticulation and architectural distortion+/-parenchymal band than pre-delta group. Our data suggest that critically ill patients with SARS-CoV-2 in delta period had more severe pulmonary fibrosis-like changes than those in pre-delta period.
Collapse
Affiliation(s)
- Jung-Wan Yoo
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Won-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chung Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Chi Ryang Chung
- Department of Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jinwoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yangjin Jegal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Junghyun Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Republic of Korea
| | - Joon-Sung Joh
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Tae Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Ae-Rin Baek
- Division of Allergy and Pulmonology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Joo Hun Park
- Department of Pulmonary and Critical Care Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Ganghee Chae
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jung Hwa Hwang
- Department of Radiology, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-Ro 43-Gil, Songpa-Gu 05505, Seoul, Republic of Korea.
| |
Collapse
|
49
|
Pellman J, Goldstein A, Słabicki M. Human E3 ubiquitin ligases: accelerators and brakes for SARS-CoV-2 infection. Biochem Soc Trans 2024; 52:2009-2021. [PMID: 39222407 PMCID: PMC11555711 DOI: 10.1042/bst20230324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
E3 ubiquitin ligases regulate the composition of the proteome. These enzymes mono- or poly-ubiquitinate their substrates, directly altering protein function or targeting proteins for degradation by the proteasome. In this review, we discuss the opposing roles of human E3 ligases as effectors and targets in the evolutionary battle between host and pathogen, specifically in the context of SARS-CoV-2 infection. Through complex effects on transcription, translation, and protein trafficking, human E3 ligases can either attenuate SARS-CoV-2 infection or become vulnerabilities that are exploited by the virus to suppress the host's antiviral defenses. For example, the human E3 ligase RNF185 regulates the stability of SARS-CoV-2 envelope protein through the ubiquitin-proteasome pathway, and depletion of RNF185 significantly increases SARS-CoV-2 viral titer (iScience (2023) 26, 106601). We highlight recent advances that identify functions for numerous human E3 ligases in the SARS-CoV-2 life cycle and we assess their potential as novel antiviral agents.
Collapse
Affiliation(s)
- Jesse Pellman
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
| | - Anna Goldstein
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
| | - Mikołaj Słabicki
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, U.S.A
| |
Collapse
|
50
|
Nikolic VN, Popadic V, Jankovic SM, Govedarović N, Vujić S, Andjelković J, Stosic LS, Stevanović NČ, Zdravkovic M, Todorovic Z. The silent predictors: exploring galectin-3 and Irisin's tale in severe COVID-19. BMC Res Notes 2024; 17:324. [PMID: 39465409 PMCID: PMC11514771 DOI: 10.1186/s13104-024-06978-3] [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: 04/24/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVE This study aimed to evaluate the roles of galectin-3 and irisin as biomarkers in predicting severe outcomes in COVID-19 patients. RESULTS We analyzed serum levels of galectin-3 and irisin in 59 patients with severe COVID-19 and 30 healthy controls. Elevated galectin-3 levels were associated with increased risks of mortality, need for intensive care, and severe acute respiratory distress syndrome. The optimal cut-off value for galectin-3 was 13.47 ng/ml, with a sensitivity of 72.7% and specificity of 76.6%. Irisin levels did not differ significantly between survivors and non-survivors at admission or on the 3rd day post-admission, but approached significance on the 7th day. These findings suggest that galectin-3 could be a valuable prognostic biomarker for severe COVID-19 outcomes, while irisin's role remains to be clarified in further studies.
Collapse
Affiliation(s)
- Valentina N Nikolic
- Department of Pharmacology with Toxicology, University of Nis Faculty of Medicine, Bul. dr Zorana Djindjica 81 Nis, Nis, 18000, Serbia.
| | - Višeslav Popadic
- University Hospital Medical Center, Bezanijska kosa, Belgrade, Serbia
| | - Slobodan M Jankovic
- Department of Pharmacology and Toxicology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nenad Govedarović
- Department of Internal Medicine, University of Nis Faculty of Medicine, Nis, Serbia
| | - Stevan Vujić
- University of Nis Faculty of Medicine, Nis, Serbia
| | | | | | | | - Marija Zdravkovic
- University Hospital Medical Center, Bezanijska kosa, Belgrade, Serbia
- Department of Internal Medicine, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zoran Todorovic
- University Hospital Medical Center, Bezanijska kosa, Belgrade, Serbia
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|