1
|
Canderan G, Muehling LM, Kadl A, Ladd S, Bonham C, Cross CE, Lima SM, Yin X, Sturek JM, Wilson JM, Keshavarz B, Enfield KB, Ramani C, Bryant N, Murphy DD, Cheon IS, Solga M, Pramoonjago P, McNamara CA, Sun J, Utz PJ, Dolatshahi S, Irish JM, Woodfolk JA. Distinct type 1 immune networks underlie the severity of restrictive lung disease after COVID-19. Nat Immunol 2025; 26:595-606. [PMID: 40140496 DOI: 10.1038/s41590-025-02110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025]
Abstract
The variable origins of persistent breathlessness after coronavirus disease 2019 (COVID-19) have hindered efforts to decipher the immunopathology of lung sequelae. Here we analyzed hundreds of cellular and molecular features in the context of discrete pulmonary phenotypes to define the systemic immune landscape of post-COVID lung disease. Cluster analysis of lung physiology measures highlighted two phenotypes of restrictive lung disease that differed according to their impaired diffusion and severity of fibrosis. Machine learning revealed marked CCR5+CD95+CD8+ T cell perturbations in milder lung disease but attenuated T cell responses hallmarked by elevated CXCL13 in more severe disease. Distinct sets of cells, mediators and autoantibodies distinguished each restrictive phenotype and differed from those of patients without substantial lung involvement. These differences were reflected in divergent T cell-based type 1 networks according to the severity of lung disease. Our findings, which provide an immunological basis for active lung injury versus advanced disease after COVID-19, might offer new targets for treatment.
Collapse
Affiliation(s)
- Glenda Canderan
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alexandra Kadl
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Shay Ladd
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Science, Charlottesville, VA, USA
| | - Catherine Bonham
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Claire E Cross
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Sierra M Lima
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xihui Yin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey M Sturek
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jeffrey M Wilson
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Behnam Keshavarz
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kyle B Enfield
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Chintan Ramani
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Naomi Bryant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Deborah D Murphy
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - In Su Cheon
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Michael Solga
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Patcharin Pramoonjago
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Coleen A McNamara
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jie Sun
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Paul J Utz
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Sepideh Dolatshahi
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Judith A Woodfolk
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Valdez-Salas B, Salvador-Carlos J, Valdez-Salas E, Beltrán-Partida E, Castillo-Saenz J, Curiel-Álvarez M, Gonzalez-Mendoza D, Cheng N. Nasal Spray Disinfectant for Respiratory Infections Based on Functionalized Silver Nanoparticles: A Physicochemical and Docking Approach. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:533. [PMID: 40214578 PMCID: PMC11990716 DOI: 10.3390/nano15070533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Respiratory diseases have presented a remarkable challenge during modern history, contributing to important pandemics. The scientific community has focused its efforts on developing vaccines and blocking the transmission of viruses through the respiratory tract. In this study, we propose the use of stable silver nanoparticles (AgNPs) functionalized with tannic acid (TA) and sodium citrate (SC) as a nasal spray disinfectant (NSD). The non-ionic ethoxylated surfactant Tween 80 (T80) was added to enhance the wetting effect on nasal and oral tissues following spray application. We analyzed the physicochemical properties of the AgNPs and the NSD, including zeta potential, polarity, morphology, composition, particle size, and distribution. The results indicated spherical AgNPs ranging from 3 to 5 nm, stabilized by TA-SC. The addition of T80 resulted in particles with negative polarity, high stability, and improved coverage area. Furthermore, the colloidal stability was monitored over one year, showing no signs of degradation or precipitation. Interestingly, the interaction between the capped AgNP complex, the spike protein, and ACE2 was studied by molecular docking, indicating a strong and thermodynamically favorable complex interaction. These findings hold promise for the development of potential inhibitors, antagonist receptors, Ag-complex agonists (as observed here), and drug development for viral protection.
Collapse
Affiliation(s)
- Benjamín Valdez-Salas
- Core Facilities of Chemistry and Advanced Materials, Instituto de Ingeniería, Universidad Autónoma de Baja California, Calle de La Normal S/N and Boulevard Benito Juárez, Mexicali 21100, Baja California, Mexico; (B.V.-S.); (E.B.-P.); (J.C.-S.); (M.C.-Á.)
| | - Jorge Salvador-Carlos
- Core Facilities of Chemistry and Advanced Materials, Instituto de Ingeniería, Universidad Autónoma de Baja California, Calle de La Normal S/N and Boulevard Benito Juárez, Mexicali 21100, Baja California, Mexico; (B.V.-S.); (E.B.-P.); (J.C.-S.); (M.C.-Á.)
| | - Ernesto Valdez-Salas
- Centro Médico Ixchel, Av. Nicolás Bravo 270, Mexicali 21000, Baja California, Mexico;
| | - Ernesto Beltrán-Partida
- Core Facilities of Chemistry and Advanced Materials, Instituto de Ingeniería, Universidad Autónoma de Baja California, Calle de La Normal S/N and Boulevard Benito Juárez, Mexicali 21100, Baja California, Mexico; (B.V.-S.); (E.B.-P.); (J.C.-S.); (M.C.-Á.)
| | - Jhonathan Castillo-Saenz
- Core Facilities of Chemistry and Advanced Materials, Instituto de Ingeniería, Universidad Autónoma de Baja California, Calle de La Normal S/N and Boulevard Benito Juárez, Mexicali 21100, Baja California, Mexico; (B.V.-S.); (E.B.-P.); (J.C.-S.); (M.C.-Á.)
| | - Mario Curiel-Álvarez
- Core Facilities of Chemistry and Advanced Materials, Instituto de Ingeniería, Universidad Autónoma de Baja California, Calle de La Normal S/N and Boulevard Benito Juárez, Mexicali 21100, Baja California, Mexico; (B.V.-S.); (E.B.-P.); (J.C.-S.); (M.C.-Á.)
| | - Daniel Gonzalez-Mendoza
- Instituto de Ciencias Agrícolas, Universidad Autónoma de Baja California, Carretera a Delta s/n, Ejido Nuevo Leon, Mexicali 21705, Baja California, Mexico;
| | - Nelson Cheng
- Magna International Pte Ltd., 10 H Enterprise Road, Singapore 629834, Singapore;
| |
Collapse
|
3
|
Nyasulu PS, Tamuzi JL, Oliveira RKF, Oliveira SD, Petrosillo N, de Jesus Perez V, Dhillon N, Butrous G. COVID-19 and Parasitic Co-Infection: A Hypothetical Link to Pulmonary Vascular Disease. Infect Dis Rep 2025; 17:19. [PMID: 40126325 PMCID: PMC11932205 DOI: 10.3390/idr17020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Background/Objectives: Before the Coronavirus disease 2019 (COVID-19) era, the global prevalence of pulmonary arterial hypertension (PAH) was between 0.4 and 1.4 per 100,000 people. The long-term effects of protracted COVID-19 associated with pulmonary vascular disease (PVD) risk factors may increase this prevalence. According to preliminary data, the exact prevalence of early estimates places the prevalence of PVD in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection at 22%, although its predictive value remains unknown. PVD caused by COVID-19 co-infections is understudied and underreported, and its future impact is unclear. However, due to COVID-19/co-infection pathophysiological effects on pulmonary vascularization, PVD mortality and morbidity may impose a genuine concern-both now and in the near future. Based on reported studies, this literature review focused on the potential link between COVID-19, parasitic co-infection, and PVD. This review article also highlights hypothetical pathophysiological mechanisms between COVID-19 and parasitic co-infection that could trigger PVD. Methods: We conducted a systematic literature review (SLR) searching peer-reviewed articles, including link between COVID-19, parasitic co-infection, and PVD. Results: This review hypothesized that multiple pathways associated with pathogens such as underlying schistosomiasis, human immunodeficiency virus (HIV), pulmonary tuberculosis (PTB), pulmonary aspergillosis, Wuchereria bancrofti, Clonorchis sinensis, paracoccidioidomycosis, human herpesvirus 8, and scrub typhus coupled with acute or long COVID-19, may increase the burden of PVD and worsen its mortality in the future. Conclusions: Further experimental studies are also needed to determine pathophysiological pathways between PVD and a history of COVID-19/co-infections.
Collapse
|
4
|
Ayyoub S, Dhillon NK, Tura-Ceide O. Genetics of Long COVID: Exploring the Molecular Drivers of Persistent Pulmonary Vascular Disease Symptoms. Infect Dis Rep 2025; 17:15. [PMID: 39997467 PMCID: PMC11855385 DOI: 10.3390/idr17010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
Background/ Objectives: Long COVID or post-acute sequelae of SARS-CoV-2 infection (PASC) are symptoms that manifest despite passing the acute infection phase. These manifestations encompass a wide range of symptoms, the most common being fatigue, shortness of breath, and cognitive dysfunction. Genetic predisposition is clearly involved in the susceptibility of individuals to developing these persistent symptoms and the variation in the severity and forms. This review summarizes the role of genetic factors and gene polymorphisms in the development of major pulmonary vascular disorders associated with long COVID. Methods: A comprehensive review of current literature was conducted to examine the genetic contributions to pulmonary complications following SARS-CoV-2 infection. Studies investigating genetic polymorphisms linked to pulmonary hypertension, pulmonary thromboembolism, and pulmonary vascular endothelialitis were reviewed and summarized. Results: Findings show that specific genetic variants contribute to increased susceptibility to pulmonary vascular complications in long COVID patients. Variants associated with endothelial dysfunction, coagulation pathways, and inflammatory responses have been implicated in the development of pulmonary hypertension and thromboembolic events. Genetic predispositions influencing vascular integrity and immune responses appear to influence disease severity and progression. Conclusions: Understanding these mechanisms and genetic predispositions could pave the way for targeted therapeutic interventions to alleviate the burden on patients experiencing long COVID.
Collapse
Affiliation(s)
- Sana Ayyoub
- Department of Medical Sciences, Faculty of Medicine, University of Girona, 17004 Girona, Spain;
| | - Navneet Kaur Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS 66160, USA;
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Olga Tura-Ceide
- Translational Research Group on Cardiovascular Respiratory Diseases (CAREs), Girona Biomedical Research Institute (IDIBGI-CERCA), Martí i Julià, Hospital Park Building M2, 17190 Salt, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
5
|
da Silva Nunes BB, Dos Santos Mendonça J, de Matos LP, Guimarães ATB, Soares WR, de Lima Rodrigues AS, Govindarajan M, Gomes AR, da Luz TM, Malafaia G. Beyond the virus: ecotoxicological and reproductive impacts of SARS-CoV-2 lysate protein in C57Bl/6j female mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2025; 32:1805-1829. [PMID: 39745629 DOI: 10.1007/s11356-024-35840-z] [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: 07/29/2024] [Accepted: 12/20/2024] [Indexed: 01/29/2025]
Abstract
Since the establishment of the COVID-19 pandemic, a range of studies have been developed to understand the pathogenesis of SARS-CoV-2 infection, vaccine development, and therapeutic testing. However, the possible impacts that these viruses can have on non-target organisms have been explored little, and our knowledge of the consequences of the COVID-19 pandemic for biota is still very limited. Thus, the current study aimed to address this knowledge gap by evaluating the possible impacts of oral exposure of C57Bl/6 J female mice to SARS-CoV-2 lysate protein (at 20 µg/L) for 30 days, using multiple methods, including behavioral assessments, biochemical analyses, and histopathological examinations. Although we did not have evidence of hematological, mutagenic, or genotoxic effects, we noted that the ingestion of SARS-CoV-2 lysate protein-induced behavioral disorders (hypoactivity, anxiety-like behavior, and short-term memory deficit), which were associated with oxidative stress and dopaminergic and cholinesterase imbalance in the animal brain. Furthermore, the elevation of bilirubin levels and lactate dehydrogenase levels in these animals suggests the occurrence of hepatic changes, and the redox imbalance, nitrosative stress, and elevated production of IFN-γ and inflammatory infiltration in the duodenum, disrupted follicular structure, and presence of vacuoles in granulosa cells, in ovarian, indicate that the SARS-CoV-2-exposed group showed significant toxicity. Principal component analysis (PCA) and cluster analysis confirmed that the groups were clearly separated and showed that the largest changes upon SARS-CoV-2 exposure were related to ROS, MDA, nitrite, IFN-γ/IL-10 levels and SOD and catalase activity in the ovary; IFN-γ/IL-10 production and SOD activity in the duodenum; BChE activity in the brain; bilirubin levels and lactate dehydrogenase activity in the serum; number of primary follicles in the ovary. In conclusion, our study provides new insights into the toxicity of SARS-CoV-2 lysate proteins in a non-target terrestrial organism of infection and, therefore, expands our understanding of the real extent of the ecological/environmental impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Bárbara Beatriz da Silva Nunes
- Post-Graduation Program in Ecology, Conservation, and Biodiversity, Federal University of Uberlândia, Uberlândia, MG, 38408144, Brazil
| | - Juliana Dos Santos Mendonça
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, 75790-000, Brazil
| | - Letícia Paiva de Matos
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, 75790-000, Brazil
| | - Abraão Tiago Batista Guimarães
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, 75790-000, Brazil
| | - Wesley Rodrigues Soares
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, 75790-000, Brazil
| | - Aline Sueli de Lima Rodrigues
- Post-Graduation Program in Conservation of Cerrado Natural Resources, Goiano Federal Institute, Urutaí, GO, 75790-000, Brazil
| | | | - Alex Rodrigues Gomes
- Post-Graduation Program in Ecology, Conservation, and Biodiversity, Federal University of Uberlândia, Uberlândia, MG, 38408144, Brazil
| | - Thiarlen Marinho da Luz
- Post-Graduation Program in Ecology, Conservation, and Biodiversity, Federal University of Uberlândia, Uberlândia, MG, 38408144, Brazil
| | - Guilherme Malafaia
- Post-Graduation Program in Ecology, Conservation, and Biodiversity, Federal University of Uberlândia, Uberlândia, MG, 38408144, Brazil.
- Laboratory of Toxicology Applied to the Environment, Goiano Federal Institute - Urutaí Campus, Rodovia Geraldo Silva Nascimento, 2,5 Km, Zona Rural, Urutaí, GO, 75790-000, Brazil.
- Post-Graduation Program in Conservation of Cerrado Natural Resources, Goiano Federal Institute, Urutaí, GO, 75790-000, Brazil.
| |
Collapse
|
6
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
7
|
Ljilja Posavec A, Cvetković Kučić D, Zagorec N, Malnar L, Lalić K, Zelenika M, Kovačević I, Kušter D, Mutvar A, Piskač Ž ivković N. Prolonged corticosteroid therapy and lung abnormalities in patients after severe COVID-19 pneumonia. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024052. [PMID: 39655598 PMCID: PMC11708960 DOI: 10.36141/svdld.v41i4.14331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/06/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Some of the hospitalized patients after severe COVID-19 pneumonia experience significant fall in peripheral saturation despite optimal treatment. Because of immune dysregulation in COVID-19 there are indications that prolonged corticosteroids could be considered in treating patients for persistent radiological sequelae and respiratory symptoms. OBJECTIVES to investigate lung function and lung sequelae on high-resolution CT (HRCT) im COVID-19 patients who were treated with glucocorticoid therapy in two dose regimens with a control group of patients who did not receive additional glucocorticoid therapy. METHODS In this prospective cohort research we studied patients who suffered from prolonged respiratory insufficiency after severe COVID-19 pneumonia. Patients received corticosteroid therapy in two dose regimens: for 14 days and for 3 months after discharge from the hospital. Control group of patients did not receive additional corticosteroid therapy. Lung function, post-COVID-19 symptoms, and lung abnormalities on CT scans were analyzed in three months follow-up and compared with the control group of patients. RESULTS Patients who received prolonged corticosteroid therapy for three months did not have better CT findings of lung abnormalities, lung function, or symptoms recovery in comparison to the patients with 14 days of therapy and control group of patients. Onwards, control group had significantly fewer dyspnea symptoms (Chi-square test, p=0,04) and higher DLCO (Kruskal Wallis test, p=0,03). CONCLUSIONS Supplementary corticosteroid therapy for patients after severe pneumonia and prolonged respiratory insufficiency with lung abnormalities after COVID-19 did not improve lung function or lung lesions on CT.
Collapse
Affiliation(s)
| | - Daria Cvetković Kučić
- Clinical Department of Diagnostic and Interventional Radiology, University Hospital Dubrava, Zagreb, Croatia
| | - Nikola Zagorec
- Department of Nephrology, University Hospital Dubrava, Zagreb, Croatia
| | - Linda Malnar
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Kristina Lalić
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Marina Zelenika
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Ivona Kovačević
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Dinka Kušter
- Department of Nuclear Medicine, University Hospital Dubrava, Zagreb, Croatia
| | - Andreja Mutvar
- Department of Nuclear Medicine, University Hospital Dubrava, Zagreb, Croatia
| | | |
Collapse
|
8
|
Tintore C, Cuartero J, Camps-Vilaró A, Subirana, Elosua R, Marrugat J, Degano IR. Increased risk of arrhythmias, heart failure, and thrombosis in SARS-CoV-2 positive individuals persists at one year post-infection. Comput Struct Biotechnol J 2024; 24:476-483. [PMID: 39050244 PMCID: PMC11266869 DOI: 10.1016/j.csbj.2024.06.024] [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: 03/19/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Risk of cardiovascular events is increased after COVID-19. However, information on cardiovascular risk trends after COVID-19 infection is lacking and estimates by sex are inconsistent. Our aim was to examine cardiovascular outcomes and mortality in a large cohort (164,346 participants) of SARS-CoV-2 positive individuals compared to non-positive individuals, stratified by sex. Data were obtained from the Spanish Health System's electronic medical records. Selected individuals were ≥ 45 years old with/without a positive SARS-CoV-2 test in the period March-May 2020. Follow-up was obtained until January 31, 2021, for cardiovascular events (angina/myocardial infarction, arrhythmias, bypass/revascularization, heart failure, peripheral artery disease, stroke/transient ischemic attack, and thrombosis), and until March 31, 2021, for mortality. Individuals were matched by propensity score. Incidence of cardiovascular events and mortality was compared with accelerated failure time models. The effect of matching and of COVID-19 severity was assessed with sensitivity analyses. In the first 3 months of follow-up, SARS-CoV-2 positive individuals had a higher risk of mortality and of all cardiovascular events. From 4-12 months, there was increased risk of mortality in SARS-CoV-2 positive individuals overall, of heart failure in SARS-CoV-2 positive females (HR= 1.26 [1.11-1.42]), and of arrhythmias and thrombosis in SARS-CoV-2 positive males (HR= 1.29 [1.14-1.47] and HR= 1.35 [1.03-1.77], respectively). When COVID-19 patients admitted to the ICU were excluded, incidence of thrombosis was similar in males regardless of positive/non-positive SARS-CoV-2 status. In the full year of follow-up, increased incidence of heart failure and of arrhythmias and thrombosis was observed in SARS-CoV-2 positive females and males, respectively.
Collapse
Affiliation(s)
- C. Tintore
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
| | - J. Cuartero
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Department of Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - A. Camps-Vilaró
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Subirana
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - R. Elosua
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cardiovascular Epidemiology and Genetics Research Group, IMIM, 08003 Barcelona, Spain
| | - J. Marrugat
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - IR Degano
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Registre Gironí del Cor (REGICOR) Study Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- Institute for Research and Innovation in Life Sciences and Health in Central Catalonia (IRIS-CC), 08500 Vic, Spain
| |
Collapse
|
9
|
Zhang L, Cai M, Zhang X, Wang S, Pang L, Chen X, Zheng C, Sun Y, Liang Y, Guo S, Wei F, Zhang Y. Integrated analysis of microbiome and host transcriptome unveils correlations between lung microbiota and host immunity in bronchoalveolar lavage fluid of pneumocystis pneumonia patients. Microbes Infect 2024; 26:105374. [PMID: 38849069 DOI: 10.1016/j.micinf.2024.105374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
OBJECTIVE The lung microbiota of patients with pulmonary diseases is disrupted and impacts the immunity. The microbiological and immune landscape of the lungs in patients with pneumocystis pneumonia (PCP) remains poorly understood. METHODS Multi-omics analysis and machine learning were performed on bronchoalveolar lavage fluid to explore interaction between the lung microbiota and host immunity in PCP. Then we constructed a diagnostic model using differential genes with LASSO regression and validated by qPCR. The immune infiltration analysis was performed to explore the landscape of lung immunity in patients with PCP. RESULTS Patients with PCP showed a low alpha diversity of lung microbiota, accompanied by the elevated abundance of Firmicutes, and the differential expressed genes (DEGs) analysis displayed a downregulation of MAPK signaling. The MAPK10, TGFB1, and EFNA3 indicated a potential to predict PCP (AUC = 0.86). The lung immune landscape in PCP showed the lower levels of naïve CD4+ T cells and activated dendritic cells. The correlation analysis of the MAPK signaling pathway-related DEGs and the differential microorganisms at the level of phylum showed that the Firmicutes was negatively correlated with these DEGs. CONCLUSION We profiled the characteristics of lung microbiota and immune landscape in PCP, which may contribute to elucidating the mechanism of PCP.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Miaotian Cai
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Sitong Wang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Lijun Pang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xue Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China
| | - Caopei Zheng
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Ying Liang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China
| | - Shan Guo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Feili Wei
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China; Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing Key Laboratory for HIV/AIDS Research, Beijing, 100069, China; Laboratory for Clinical Medicine, Capital Medical University, China; Beijing Research Center for Respiratory Infectious Diseases, China.
| |
Collapse
|
10
|
Oliveira SD, Almodóvar S, Butrous G, De Jesus Perez V, Fabro A, Graham BB, Mocumbi A, Nyasulu PS, Tura‐Ceide O, Oliveira RKF, Dhillon NK. Infection and pulmonary vascular diseases consortium: United against a global health challenge. Pulm Circ 2024; 14:e70003. [PMID: 39534510 PMCID: PMC11555293 DOI: 10.1002/pul2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/16/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024] Open
Abstract
Leveraging the potential of virtual platforms in the post-COVID-19 era, the Infection and Pulmonary Vascular Diseases Consortium (iPVDc), with the support of the Pulmonary Vascular Research Institute (PVRI), launched a globally accessible educational program to highlight top-notch research on inflammation and infectious diseases affecting the lung vasculature. This innovative virtual series has already successfully brought together distinguished investigators across five continents - Asia, Europe, South and North America, and Africa. Moreover, these open global forums have contributed to a comprehensive understanding of the complex interplay among immunology, inflammation, infection, and cardiopulmonary health, especially concerning pulmonary hypertension and related pulmonary disorders. These enlightening discussions have not only heightened awareness about the impact of various pathogenic microorganisms, including fungi, parasites, and viruses, on the onset and development of pulmonary vascular diseases but have also cast a spotlight on co-infections and neglected illnesses like schistosomiasis - a disease that continues to impose a heavy socioeconomic burden in numerous regions worldwide. Thus, the overall goal of this review article is to present the most recent breakthroughs from infectious PVDs as well as bring to light the scientific and educational insights from the 2023 iPVDc/PVRI virtual symposium series, shaping our understanding of these crucial health issues in this more than ever interconnected world.
Collapse
Affiliation(s)
- S. D. Oliveira
- Vascular Immunobiology Lab, Department of Anesthesiology, Department of Physiology and Biophysics, College of MedicineUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - S. Almodóvar
- Department of Immunology & Molecular MicrobiologyTexas Tech University Health Sciences Center, School of MedicineLubbockTexasUSA
| | - G. Butrous
- Medway School of PharmacyUniversity of KentMedwayKentUnited Kingdom
| | - V De Jesus Perez
- Division of Pulmonary and Critical CareStanford UniversityPalo AltoCaliforniaUSA
| | - A. Fabro
- Division of Respiratory DiseasesFederal University of São PauloSao PauloBrazil
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical SchoolUniversidade de São PauloRibeirão PretoBrazil
| | - B. B. Graham
- Department of Medicine, Zuckerberg San Francisco General HospitalUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - A. Mocumbi
- Department of MedicineUniversidade Eduardo MondlaneMaputoMozambique
- Division of Determinants of Chronic Diseases, Instituto Nacional de SaúdeVila de MarracueneMozambique
| | - P. S. Nyasulu
- Department of Global Health, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
- School of Public Health, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - O. Tura‐Ceide
- Biomedical Research Institute‐IDIBGIGironaSpain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES)MadridSpain
| | - R. K. F. Oliveira
- Division of Respiratory Diseases, Department of MedicineFederal University of São Paulo (Unifesp)São PauloBrazil
| | - N. K. Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal MedicineUniversity of Kansas Medical CenterKansas CityKansasUSA
| | | |
Collapse
|
11
|
Dondossola D, Lonati C, Pini A, Bignamini D, Zanella A, Lombardi R, Scaravilli V, La Mura V, Forzenigo L, Biondetti P, Grasselli G, Fracanzani A, Paleari C, Cespiati A, Todaro S, Cattaneo E, Di Feliciantonio M, Sigon G, Valsecchi C, Guzzardella A, Battistin M, Iuculano F. Portal hypertension-like pattern in coronavirus disease 2019 acute respiratory distress syndrome. J Crit Care 2024; 82:154759. [PMID: 38461659 DOI: 10.1016/j.jcrc.2024.154759] [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: 10/04/2023] [Revised: 01/12/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVES Although respiratory failure is the most common feature in coronavirus disease 2019 (COVID-19), abdominal organ involvement is likewise frequently observed. To investigate visceral and thoracic circulation and abdominal organ damage in COVID-19 patients. MATERIALS AND METHODS A monocentric observational study was carried on. In COVID-19 patients affected by acute respiratory distress syndrome (ARDS) (n = 31) or mild pneumonia (n = 60) thoracoabdominal circulation was evaluated using Doppler-ultrasound and computed tomography. The study also included non-COVID-19 patients affected by ARDS (n = 10) or portal hypertension (n = 10) for comparison of the main circulatory changes. RESULTS Patients affected by COVID-19 ARDS showed hyperdynamic visceral flow and increased portal velocity, hepatic artery resistance-index, and spleen diameter relative to those with mild-pneumonia (p = 0.001). Splanchnic circulatory parameters significantly correlated with the main respiratory indexes (p < 0.001) and pulmonary artery diameter (p = 0.02). The chest and abdominal vascular remodeling pattern of COVID-19 ARDS patients resembled the picture observed in the PH group, while differed from that of the non-COVID ARDS group. A more severe COVID-19 presentation was associated with worse liver dysfunction and enhanced inflammatory activation; these parameters both correlated with abdominal (p = 0.04) and chest imaging measures (p = 0.03). CONCLUSION In COVID-19 ARDS patients there are abdominal and lung vascular modifications that depict a portal hypertension-like pattern. The correlation between visceral vascular remodeling, pulmonary artery enlargement, and organ damage in these critically ill patients is consistent with a portal hyperlfow-like syndrome that could contribute to the peculiar characteristics of respiratory failure in these patients. CLINICAL RELEVANCE STATEMENT our data suggest that the severity of COVID-19 lung involvement is directly related to the development of a portal hyperflow-like syndrome. These observations should help in defining the need for a closer monitoring, but also to develop dedicated therapeutic strategies.
Collapse
Affiliation(s)
- Daniele Dondossola
- General and Liver Transplant Surgery Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi of Milan, 20019 Milan, Italy.
| | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Alessia Pini
- Department of Statistical Sciences, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Daniela Bignamini
- Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Alberto Zanella
- Department of Pathophysiology and Transplantation, Università degli Studi of Milan, 20019 Milan, Italy; Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Rosa Lombardi
- Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Vittorio Scaravilli
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Vincenzo La Mura
- Internal Medicine, Hemostasis and Thrombosis Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy; Department of Biomedical Science for Health, Università degli Studi of Milan, 20019 Milan, Italy
| | - Laura Forzenigo
- Division of Radiology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Pierpaolo Biondetti
- Division of Radiology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Giacomo Grasselli
- Department of Pathophysiology and Transplantation, Università degli Studi of Milan, 20019 Milan, Italy; Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Anna Fracanzani
- Department of Pathophysiology and Transplantation, Università degli Studi of Milan, 20019 Milan, Italy; Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Chiara Paleari
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Annalisa Cespiati
- Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Serena Todaro
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Emanuele Cattaneo
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Marianna Di Feliciantonio
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Giordano Sigon
- Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Carlo Valsecchi
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Amedeo Guzzardella
- Department of Anesthesia and Critical Care, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan 20019, Italy
| | - Michele Battistin
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| | - Federica Iuculano
- Medicine and Metabolic Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20019 Milan, Italy
| |
Collapse
|
12
|
Metzdorf K, Jacobsen H, Kim Y, Teixeira Alves LG, Kulkarni U, Brdovčak MC, Materljan J, Eschke K, Chaudhry MZ, Hoffmann M, Bertoglio F, Ruschig M, Hust M, Šustić M, Krmpotić A, Jonjić S, Widera M, Ciesek S, Pöhlmann S, Landthaler M, Čičin-Šain L. A single-dose MCMV-based vaccine elicits long-lasting immune protection in mice against distinct SARS-CoV-2 variants. Front Immunol 2024; 15:1383086. [PMID: 39119342 PMCID: PMC11306140 DOI: 10.3389/fimmu.2024.1383086] [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: 02/06/2024] [Accepted: 06/11/2024] [Indexed: 08/10/2024] Open
Abstract
Current vaccines against COVID-19 elicit immune responses that are overall strong but wane rapidly. As a consequence, the necessary booster shots have contributed to vaccine fatigue. Hence, vaccines that would provide lasting protection against COVID-19 are needed, but are still unavailable. Cytomegaloviruses (CMVs) elicit lasting and uniquely strong immune responses. Used as vaccine vectors, they may be attractive tools that obviate the need for boosters. Therefore, we tested the murine CMV (MCMV) as a vaccine vector against COVID-19 in relevant preclinical models of immunization and challenge. We have previously developed a recombinant MCMV vaccine vector expressing the spike protein of the ancestral SARS-CoV-2 (MCMVS). In this study, we show that the MCMVS elicits a robust and lasting protection in young and aged mice. Notably, spike-specific humoral and cellular immunity was not only maintained but also even increased over a period of at least 6 months. During that time, antibody avidity continuously increased and expanded in breadth, resulting in neutralization of genetically distant variants, like Omicron BA.1. A single dose of MCMVS conferred rapid virus clearance upon challenge. Moreover, MCMVS vaccination controlled two variants of concern (VOCs), the Beta (B.1.135) and the Omicron (BA.1) variants. Thus, CMV vectors provide unique advantages over other vaccine technologies, eliciting broadly reactive and long-lasting immune responses against COVID-19.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- Mice
- COVID-19 Vaccines/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Muromegalovirus/immunology
- Muromegalovirus/genetics
- Female
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred BALB C
- Humans
- Genetic Vectors
- Immunity, Cellular
- Immunity, Humoral
- Disease Models, Animal
Collapse
Affiliation(s)
- Kristin Metzdorf
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Henning Jacobsen
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Yeonsu Kim
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | - Luiz Gustavo Teixeira Alves
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Upasana Kulkarni
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| | | | - Jelena Materljan
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Kathrin Eschke
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - M. Zeeshan Chaudhry
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center – Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Federico Bertoglio
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Maximilian Ruschig
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Michael Hust
- Department of Medical Biotechnology, Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Marko Šustić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- German Centre for Infection Research (DZIF), External Partner Site Frankfurt, Frankfurt, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center – Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of the Helmholtz Centre for Infection Medicine and the Hannover Medical School, Hannover, Germany
| |
Collapse
|
13
|
Suarez-Castillejo C, Calvo N, Preda L, Córdova Díaz R, Toledo-Pons N, Martínez J, Pons J, Vives-Borràs M, Pericàs P, Ramón L, Iglesias A, Cànaves-Gómez L, Valera Felices JL, Morell-García D, Núñez B, Sauleda J, Sala-Llinàs E, Alonso-Fernández A. Cardiopulmonary Complications after Pulmonary Embolism in COVID-19. Int J Mol Sci 2024; 25:7270. [PMID: 39000378 PMCID: PMC11242326 DOI: 10.3390/ijms25137270] [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: 05/03/2024] [Revised: 06/14/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
Although pulmonary embolism (PE) is a frequent complication in COVID-19, its consequences remain unknown. We performed pulmonary function tests, echocardiography and computed tomography pulmonary angiography and identified blood biomarkers in a cohort of consecutive hospitalized COVID-19 patients with pneumonia to describe and compare medium-term outcomes according to the presence of PE, as well as to explore their potential predictors. A total of 141 patients (56 with PE) were followed up during a median of 6 months. Post-COVID-19 radiological lung abnormalities (PCRLA) and impaired diffusing capacity for carbon monoxide (DLCOc) were found in 55.2% and 67.6% cases, respectively. A total of 7.3% had PE, and 6.7% presented an intermediate-high probability of pulmonary hypertension. No significant difference was found between PE and non-PE patients. Univariate analysis showed that age > 65, some clinical severity factors, surfactant protein-D, baseline C-reactive protein, and both peak red cell distribution width and Interleukin (IL)-10 were associated with DLCOc < 80%. A score for PCRLA prediction including age > 65, minimum lymphocyte count, and IL-1β concentration on admission was constructed with excellent overall performance. In conclusion, reduced DLCOc and PCRLA were common in COVID-19 patients after hospital discharge, but PE did not increase the risk. A PCRLA predictive score was developed, which needs further validation.
Collapse
Affiliation(s)
- Carla Suarez-Castillejo
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Néstor Calvo
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luminita Preda
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Rocío Córdova Díaz
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Nuria Toledo-Pons
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Joaquín Martínez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Pons
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Miquel Vives-Borràs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
| | - Pere Pericàs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luisa Ramón
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Amanda Iglesias
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Cànaves-Gómez
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jose Luis Valera Felices
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Morell-García
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Análisis Clínicos, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Belén Núñez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Sauleda
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ernest Sala-Llinàs
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Alonso-Fernández
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
14
|
Lubarsky D, Clark DE, Crum K, Karpinos A, Austin ED, Soslow JH. Quantifying the impact of post-acute sequelae of coronavirus on the cardiopulmonary endurance of athletes. Pulm Circ 2024; 14:e12413. [PMID: 39022309 PMCID: PMC11252491 DOI: 10.1002/pul2.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/24/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
Post-acute sequelae of Coronavirus (PASC), or Long COVID, has emerged as a critical health concern. The clinical manifestations of PASC have been described, but studies have not quantified the cardiopulmonary effects. The goal of this study was to quantify PASC cardiopulmonary changes among endurance athletes. Endurance athletes were recruited via social media; 45 met inclusion criteria, 32 had PASC and 13 were asymptomatic at 3 months (control). Comprehensive interviews were conducted to assess: cardiopulmonary symptoms at 3 months; quantitative and qualitative changes in cardiovascular endurance; exercise hours per week at baseline and 3 months; and Modified Oslo, Dyspnea, and EQ-5D-5L scales. All collected data was based on self-reported symptoms. Wilcoxon rank sum compared PASC with control to distinguish the effects of PASC vs effects of COVID infection/lockdown. PASC subjects were more likely to be female (Table). The most common 3-month symptoms in PASC were fatigue and shortness of breath. Based on self-reported data, subjects endorsed a median decrease of 27% in cardiopulmonary endurance levels compared with 0% in controls (p = 0.0019). PASC subjects exercised less hours and had worse self-reported health as compared with controls. PASC subjects also had significantly worse Modified Oslo, Dyspnea, and EQ-5D-5L scores. Of the 32 PASC patients, 10 (31%) reported a complete inability to engage in any cardiovascular endurance exercise at 3 months. PASC leads to a significant, quantifiable decrease in cardiopulmonary health and endurance.
Collapse
Affiliation(s)
- Daniel Lubarsky
- Department of PediatricsMonroe Carell Jr. Children's Hospital at Vanderbilt, and Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Daniel E. Clark
- Department of PediatricsStanford University School of MedicineStanfordCaliforniaUSA
| | - Kimberly Crum
- Department of PediatricsMonroe Carell Jr. Children's Hospital at Vanderbilt, and Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ashley Karpinos
- Department of Internal Medicine and Sports MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Eric D. Austin
- Department of PediatricsMonroe Carell Jr. Children's Hospital at Vanderbilt, and Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jonathan H. Soslow
- Department of PediatricsMonroe Carell Jr. Children's Hospital at Vanderbilt, and Vanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
15
|
Singh A, Chimata AV, Deshpande P, Bajpai S, Sangeeth A, Rajput M, Singh A. SARS-CoV2 Nsp3 protein triggers cell death and exacerbates amyloid β42-mediated neurodegeneration. Neural Regen Res 2024; 19:1385-1392. [PMID: 37905889 PMCID: PMC11467943 DOI: 10.4103/1673-5374.382989] [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: 04/04/2023] [Revised: 06/25/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
Infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) virus, responsible for the coronavirus disease 2019 (COVID-19) pandemic, induces symptoms including increased inflammatory response, severe acute respiratory syndrome (SARS), cognitive dysfunction like brain fog, and cardiovascular defects. Long-term effects of SARS-CoV2 COVID-19 syndrome referred to as post-COVID-19 syndrome on age-related progressive neurodegenerative disorders such as Alzheimer’s disease remain understudied. Using the targeted misexpression of individual SARS-CoV2 proteins in the retinal neurons of the Drosophila melanogaster eye, we found that misexpression of nonstructural protein 3 (Nsp3), a papain-like protease, ablates the eye and generates dark necrotic spots. Targeted misexpression of Nsp3 in the eye triggers reactive oxygen species production and leads to apoptosis as shown by cell death reporters, terminal deoxynucleotidyl transferase (TdT) dUTP Nick-end labeling (TUNEL) assay, and dihydroethidium staining. Furthermore, Nsp3 misexpression activates both apoptosis and autophagy mechanism(s) to regulate tissue homeostasis. Transient expression of SARS-CoV2 Nsp3 in murine neuroblastoma, Neuro-2a cells, significantly reduced the metabolic activity of these cells and triggers cell death. Misexpression of SARS-CoV2 Nsp3 in an Alzheimer’s disease transgenic fly eye model (glass multiple repeats [GMR]>amyloid β42) further enhances the neurodegenerative rough eye phenotype due to increased cell death. These findings suggest that SARS-CoV2 utilizes Nsp3 protein to potentiate cell death response in a neurodegenerative disease background that has high pre-existing levels of neuroinflammation and cell death.
Collapse
Affiliation(s)
- Aditi Singh
- Department of Biology, University of Dayton, Dayton, OH, USA
| | | | | | - Soumya Bajpai
- Department of Biology, University of Dayton, Dayton, OH, USA
| | - Anjali Sangeeth
- Department of Biology, University of Dayton, Dayton, OH, USA
| | | | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, USA
- Premedical Program, University of Dayton, Dayton, OH, USA
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
- The Integrative Science and Engineering Center, University of Dayton, Dayton, OH, USA
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
16
|
Paes Leme AF, Yokoo S, Normando AGC, Ormonde JVS, Domingues RR, Cruz FF, Silva PL, Souza BSF, Dos Santos CC, Castro-Faria-Neto H, Martins CM, Lopes-Pacheco M, Rocco PRM. Proteomics of serum-derived extracellular vesicles are associated with the severity and different clinical profiles of patients with COVID-19: An exploratory secondary analysis. Cytotherapy 2024; 26:444-455. [PMID: 38363248 DOI: 10.1016/j.jcyt.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Coronavirus disease 2019 (COVID-19) is characterized by a broad spectrum of clinical manifestations with the potential to progress to multiple organ dysfunction in severe cases. Extracellular vesicles (EVs) carry a range of biological cargoes, which may be used as biomarkers of disease state. METHODS An exploratory secondary analysis of the SARITA-2 and SARITA-1 datasets (randomized clinical trials on patients with mild and moderate/severe COVID-19) was performed. Serum-derived EVs were used for proteomic analysis to identify enriched biological processes and key proteins, thus providing insights into differences in disease severity. Serum-derived EVs were separated from patients with COVID-19 by size exclusion chromatography and nanoparticle tracking analysis was used to determine particle concentration and diameter. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was performed to identify and quantify protein signatures. Bioinformatics and multivariate statistical analysis were applied to distinguish candidate proteins associated with disease severity (mild versus moderate/severe COVID-19). RESULTS No differences were observed in terms of the concentration and diameter of enriched EVs between mild (n = 14) and moderate/severe (n = 30) COVID-19. A total of 414 proteins were found to be present in EVs, of which 360 were shared while 48 were uniquely present in severe/moderate compared to mild COVID-19. The main biological signatures in moderate/severe COVID-19 were associated with platelet degranulation, exocytosis, complement activation, immune effector activation, and humoral immune response. Von Willebrand factor, serum amyloid A-2 protein, histone H4 and H2A type 2-C, and fibrinogen β-chain were the most differentially expressed proteins between severity groups. CONCLUSION Exploratory proteomic analysis of serum-derived EVs from patients with COVID-19 detected key proteins related to immune response and activation of coagulation and complement pathways, which are associated with disease severity. Our data suggest that EV proteins may be relevant biomarkers of disease state and prognosis.
Collapse
Affiliation(s)
- Adriana F Paes Leme
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Sami Yokoo
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Ana Gabriela C Normando
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - João Vitor S Ormonde
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Romenia Ramos Domingues
- Laboratório Nacional de Biociências - LNBio, Centro Nacional de Pesquisa em Energia e Materiais - CNPEM, Campinas, São Paulo, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno S F Souza
- Goncalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, Brazil; D'Or Institute for Research and Education (IDOR), Salvador, Bahia, Brazil; Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Bahia, Brazil
| | - Claudia C Dos Santos
- The Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Sciences and Interdepartmental Division of Critical Care, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
17
|
Riou M, Coste F, Meyer A, Enache I, Talha S, Charloux A, Reboul C, Geny B. Mechanisms of Pulmonary Vasculopathy in Acute and Long-Term COVID-19: A Review. Int J Mol Sci 2024; 25:4941. [PMID: 38732160 PMCID: PMC11084496 DOI: 10.3390/ijms25094941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.
Collapse
Affiliation(s)
- Marianne Riou
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Florence Coste
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Irina Enache
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Samy Talha
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Anne Charloux
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Cyril Reboul
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| |
Collapse
|
18
|
Canderan G, Muehling LM, Kadl A, Ladd S, Bonham C, Cross CE, Lima SM, Yin X, Sturek JM, Wilson JM, Keshavarz B, Bryant N, Murphy DD, Cheon IS, McNamara CA, Sun J, Utz PJ, Dolatshahi S, Irish JM, Woodfolk JA. Distinct Type 1 Immune Networks Underlie the Severity of Restrictive Lung Disease after COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587929. [PMID: 38617217 PMCID: PMC11014603 DOI: 10.1101/2024.04.03.587929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The variable etiology of persistent breathlessness after COVID-19 have confounded efforts to decipher the immunopathology of lung sequelae. Here, we analyzed hundreds of cellular and molecular features in the context of discrete pulmonary phenotypes to define the systemic immune landscape of post-COVID lung disease. Cluster analysis of lung physiology measures highlighted two phenotypes of restrictive lung disease that differed by their impaired diffusion and severity of fibrosis. Machine learning revealed marked CCR5+CD95+ CD8+ T-cell perturbations in mild-to-moderate lung disease, but attenuated T-cell responses hallmarked by elevated CXCL13 in more severe disease. Distinct sets of cells, mediators, and autoantibodies distinguished each restrictive phenotype, and differed from those of patients without significant lung involvement. These differences were reflected in divergent T-cell-based type 1 networks according to severity of lung disease. Our findings, which provide an immunological basis for active lung injury versus advanced disease after COVID-19, might offer new targets for treatment.
Collapse
|
19
|
Razzaque MS. Can adverse cardiac events of the COVID-19 vaccine exacerbate preexisting diseases? Expert Rev Anti Infect Ther 2024; 22:131-137. [PMID: 38284355 DOI: 10.1080/14787210.2024.2311837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/25/2024] [Indexed: 01/30/2024]
Abstract
INTRODUCTION SARS-CoV-2 infection and COVID-19 vaccination can both lead to serious cardiac conditions such as myocarditis, arrhythmia, acute myocardial infarction, and coagulopathy. Further studies are needed to better understand the risks and benefits of COVID-19 vaccination, and to determine the best course of action for individuals with preexisting heart conditions. AREAS COVERED The current knowledge and challenges in understanding vaccine-associated heart issues concerning the COVID-19 pandemic are briefly summarized, highlighting similar cardiac conditions caused by either SARS-CoV-2 infection or COVID-19 vaccination and the potential clinical impacts. EXPERT OPINION The short-term risks of severe cardiovascular side effects following COVID-19 vaccination are relatively low. However, further studies are needed to determine whether adverse vaccination events outweigh the long-term benefits in specific groups of individuals. Since cardiac inflammation, blood pressure dysregulation, coagulopathy, acute myocardial infarction, or arrhythmia could be the consequences of either SARS-CoV-2 infection or COVID-19 vaccination, clinical questions should be asked whether the COVID-19 vaccine worsens the condition in persons with preexisting heart diseases. It is important to carefully assess the potential risks and benefits of COVID-19 vaccination, especially for individuals with preexisting heart conditions, and to continue monitoring and studying the long-term effects of vaccination on cardiovascular health.
Collapse
Affiliation(s)
- Mohammed S Razzaque
- Professor of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| |
Collapse
|
20
|
Landrum KR, Gaynes BN, Akello H, Malava JK, Dussault JM, Hosseinipour MC, Udedi M, Masiye J, Zimba CC, Pence BW. The longitudinal association of stressful life events with depression remission among SHARP trial participants with depression and hypertension or diabetes in Malawi. PLoS One 2024; 19:e0298546. [PMID: 38408059 PMCID: PMC10896523 DOI: 10.1371/journal.pone.0298546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 01/15/2024] [Indexed: 02/28/2024] Open
Abstract
Depressive disorders are leading contributors to morbidity in low- and middle-income countries and are particularly prevalent among people with non-communicable diseases (NCD). Stressful life events (SLEs) are risk factors for, and can help identify those at risk of, severe depressive illness requiring more aggressive treatment. Yet, research on the impact of SLEs on the trajectory of depressive symptoms among NCD patients indicated for depression treatment is lacking, especially in low resource settings. This study aims to estimate the longitudinal association of SLEs at baseline with depression remission achievement at three, six, and 12 months among adults with either hypertension or diabetes and comorbid depression identified as being eligible for depression treatment. Participants were recruited from 10 NCD clinics in Malawi from May 2019-December 2021. SLEs were measured by the Life Events Survey and depression remission was defined as achieving a Patient Health Questionaire-9 (PHQ-9) score <5 at follow-up. The study population (n = 737) consisted predominately of females aged 50 or higher with primary education and current employment. At baseline, participants reported a mean of 3.5 SLEs in the prior three months with 90% reporting ≥1 SLE. After adjustment, each additional SLE was associated with a lower probability of achieving depression remission at three months (cumulative incidence ratio (CIR) 0.94; 95% confidence interval: 0.90, 0.98, p = 0.002), six months (0.95; 0.92, 0.98, p = 0.002) and 12 months (0.96; 0.94, 0.99, p = 0.011). Re-expressed per 3-unit change, the probability of achieving depression remission at three, six, and 12 months was 0.82, 0.86, and 0.89 times lower per 3 SLEs (the median number of SLEs). Among NCD patients identified as eligible for depression treatment, recent SLEs at baseline were associated with lower probability of achieving depression remission at three, six, and 12 months. Findings suggest that interventions addressing SLEs during integrated NCD and depression care interventions (e.g., teaching and practicing SLE coping strategies) may improve success of depression treatment among adult patient populations in low-resource settings and may help identify those at risk of severe and treatment resistant depression.
Collapse
Affiliation(s)
- Kelsey R. Landrum
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Bradley N. Gaynes
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | | | - Josée M. Dussault
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Mina C. Hosseinipour
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Michael Udedi
- Noncommunicable Diseases and Mental Health Unit, Malawi Ministry of Health, Lilongwe, Malawi
| | - Jones Masiye
- Noncommunicable Diseases and Mental Health Unit, Malawi Ministry of Health, Lilongwe, Malawi
| | | | - Brian W. Pence
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
21
|
Wu X, Xiang M, Jing H, Wang C, Novakovic VA, Shi J. Damage to endothelial barriers and its contribution to long COVID. Angiogenesis 2024; 27:5-22. [PMID: 37103631 PMCID: PMC10134732 DOI: 10.1007/s10456-023-09878-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 04/16/2023] [Indexed: 04/28/2023]
Abstract
The world continues to contend with COVID-19, fueled by the emergence of viral variants. At the same time, a subset of convalescent individuals continues to experience persistent and prolonged sequelae, known as long COVID. Clinical, autopsy, animal and in vitro studies all reveal endothelial injury in acute COVID-19 and convalescent patients. Endothelial dysfunction is now recognized as a central factor in COVID-19 progression and long COVID development. Different organs contain different types of endothelia, each with specific features, forming different endothelial barriers and executing different physiological functions. Endothelial injury results in contraction of cell margins (increased permeability), shedding of glycocalyx, extension of phosphatidylserine-rich filopods, and barrier damage. During acute SARS-CoV-2 infection, damaged endothelial cells promote diffuse microthrombi and destroy the endothelial (including blood-air, blood-brain, glomerular filtration and intestinal-blood) barriers, leading to multiple organ dysfunction. During the convalescence period, a subset of patients is unable to fully recover due to persistent endothelial dysfunction, contributing to long COVID. There is still an important knowledge gap between endothelial barrier damage in different organs and COVID-19 sequelae. In this article, we mainly focus on these endothelial barriers and their contribution to long COVID.
Collapse
Affiliation(s)
- Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, 150001, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, 150001, Harbin, China
| | - Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, 150001, Harbin, China
| | - Chengyue Wang
- Department of Hematology, The First Hospital, Harbin Medical University, 150001, Harbin, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, 150001, Harbin, China.
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, MA, Boston, USA.
| |
Collapse
|
22
|
Mohan M, Kothari A, Verhagen N, Shreenivas A, Radhakrishnan SV, Dhakal B, Figueroa-Castro C, Chhabra S, Janz S, Pasquini M, Hamadani M, Szabo A, D'Souza A. Blood and marrow transplant within 4 weeks of SARS-CoV-2 infection is associated with increased risk of mortality: a National COVID Cohort Collaborative (N3C) Study. Bone Marrow Transplant 2024; 59:121-124. [PMID: 37803198 DOI: 10.1038/s41409-023-02096-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 10/08/2023]
Affiliation(s)
- Meera Mohan
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Anai Kothari
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Aditya Shreenivas
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Binod Dhakal
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carlos Figueroa-Castro
- Division of Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Siegfried Janz
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marcello Pasquini
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mehdi Hamadani
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aniko Szabo
- Division of Biostatistics, Institute of Health and Safety, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anita D'Souza
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
23
|
Scheim DE, Vottero P, Santin AD, Hirsh AG. Sialylated Glycan Bindings from SARS-CoV-2 Spike Protein to Blood and Endothelial Cells Govern the Severe Morbidities of COVID-19. Int J Mol Sci 2023; 24:17039. [PMID: 38069362 PMCID: PMC10871123 DOI: 10.3390/ijms242317039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Consistent with well-established biochemical properties of coronaviruses, sialylated glycan attachments between SARS-CoV-2 spike protein (SP) and host cells are key to the virus's pathology. SARS-CoV-2 SP attaches to and aggregates red blood cells (RBCs), as shown in many pre-clinical and clinical studies, causing pulmonary and extrapulmonary microthrombi and hypoxia in severe COVID-19 patients. SARS-CoV-2 SP attachments to the heavily sialylated surfaces of platelets (which, like RBCs, have no ACE2) and endothelial cells (having minimal ACE2) compound this vascular damage. Notably, experimentally induced RBC aggregation in vivo causes the same key morbidities as for severe COVID-19, including microvascular occlusion, blood clots, hypoxia and myocarditis. Key risk factors for COVID-19 morbidity, including older age, diabetes and obesity, are all characterized by markedly increased propensity to RBC clumping. For mammalian species, the degree of clinical susceptibility to COVID-19 correlates to RBC aggregability with p = 0.033. Notably, of the five human betacoronaviruses, the two common cold strains express an enzyme that releases glycan attachments, while the deadly SARS, SARS-CoV-2 and MERS do not, although viral loads for COVID-19 and the two common cold infections are similar. These biochemical insights also explain the previously puzzling clinical efficacy of certain generics against COVID-19 and may support the development of future therapeutic strategies for COVID-19 and long COVID patients.
Collapse
Affiliation(s)
- David E Scheim
- US Public Health Service, Commissioned Corps, Inactive Reserve, Blacksburg, VA 24060, USA
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, P.O. Box 208063, New Haven, CT 06520, USA
| | | |
Collapse
|
24
|
Fetita C, Richeux J, Didier A, Maury M, Flint J, Brillet PY, Bergot E, Bernaudin JF, Justet A. Comparison between computerised lung SPECT-CT and noncontrast thoracic HRCT for quantitative analysis of post-acute COVID-19 pulmonary vascular pruning. ERJ Open Res 2023; 9:00076-2023. [PMID: 37868150 PMCID: PMC10588800 DOI: 10.1183/23120541.00076-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 08/09/2023] [Indexed: 10/24/2023] Open
Abstract
Computerised processing of images from routine noncontrast HRCT could be an efficient, costless and safe tool to investigate the vascular remodelling that occurs in the months after COVID-19 in a large number of patients https://bit.ly/3qAQZDW.
Collapse
Affiliation(s)
- Catalin Fetita
- SAMOVAR, Telecom SudParis, Institut Polytechnique de Paris, Evry, France
- These authors contributed equally
| | - Jean Richeux
- Service de Pneumologie, CHU de Caen, Caen, France
| | - Antoine Didier
- SAMOVAR, Telecom SudParis, Institut Polytechnique de Paris, Evry, France
| | - Mathilde Maury
- SAMOVAR, Telecom SudParis, Institut Polytechnique de Paris, Evry, France
| | - Jasper Flint
- Internal Medicine Department, Pulmonary Critical Care and Sleep Medicine Section, Yale School of Medicine, New Haven, CT, USA
| | - Pierre-Yves Brillet
- Service de Radiologie Hôpital Avicenne, AP-HP, Bobigny, France
- INSERM 1272 Université Sorbonne Paris-Nord, Bobigny, France
| | - Emmanuel Bergot
- Service de Pneumologie, CHU de Caen, Caen, France
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, Caen, France
| | - Jean-Francois Bernaudin
- INSERM 1272 Université Sorbonne Paris-Nord, Bobigny, France
- Faculté de Médecine Sorbonne Université Paris, Paris, France
- These authors contributed equally
| | - Aurélien Justet
- Service de Pneumologie, CHU de Caen, Caen, France
- Internal Medicine Department, Pulmonary Critical Care and Sleep Medicine Section, Yale School of Medicine, New Haven, CT, USA
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT, UMR6030, GIP Cyceron, Caen, France
- These authors contributed equally
| |
Collapse
|
25
|
Krivosikova L, Kuracinova T, Martanovic P, Hyblova M, Kaluzay J, Uhrinova A, Janega P, Babal P. Long COVID Complicated by Fatal Cytomegalovirus and Aspergillus Infection of the Lungs: An Autopsy Case Report. Viruses 2023; 15:1810. [PMID: 37766216 PMCID: PMC10535245 DOI: 10.3390/v15091810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
After the acute phase of COVID-19, some patients develop long COVID. This term is used for a variety of conditions with a complex, yet not fully elucidated etiology, likely including the prolonged persistence of the virus in the organism and progression to lung fibrosis. We present a unique autopsy case of a patient with severe COVID-19 with prolonged viral persistence who developed interstitial lung fibrosis complicated by a fatal combination of cytomegalovirus and Aspergillus infection. SARS-CoV-2 virus was detected at autopsy in the lungs more than two months after the acute infection, although tests from the nasopharynx were negative. Immune dysregulation after COVID-19 and the administration of corticoid therapy created favorable conditions for the cytomegalovirus and Aspergillus infection that were uncovered at autopsy. These pathogens may represent a risk for opportunistic infections, complicating not only the acute coronavirus infection but also long COVID, as was documented in the presented case.
Collapse
Affiliation(s)
- Lucia Krivosikova
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia; (L.K.); (T.K.); (P.J.)
| | - Tereza Kuracinova
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia; (L.K.); (T.K.); (P.J.)
| | - Peter Martanovic
- Health Care Surveillance Authority, 829 24 Bratislava, Slovakia;
| | | | - Jozef Kaluzay
- 4th Department of Internal Medicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia;
| | | | - Pavol Janega
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia; (L.K.); (T.K.); (P.J.)
- Centre of Experimental Medicine, Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, 813 71 Bratislava, Slovakia
| | - Pavel Babal
- Institute of Pathological Anatomy, Faculty of Medicine, Comenius University in Bratislava, 814 99 Bratislava, Slovakia; (L.K.); (T.K.); (P.J.)
- Medirex Group Academy, n.p.o., 949 05 Nitra, Slovakia;
| |
Collapse
|
26
|
Borek I, Birnhuber A, Voelkel NF, Marsh LM, Kwapiszewska G. The vascular perspective on acute and chronic lung disease. J Clin Invest 2023; 133:e170502. [PMID: 37581311 PMCID: PMC10425217 DOI: 10.1172/jci170502] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
The pulmonary vasculature has been frequently overlooked in acute and chronic lung diseases, such as acute respiratory distress syndrome (ARDS), pulmonary fibrosis (PF), and chronic obstructive pulmonary disease (COPD). The primary emphasis in the management of these parenchymal disorders has largely revolved around the injury and aberrant repair of epithelial cells. However, there is increasing evidence that the vascular endothelium plays an active role in the development of acute and chronic lung diseases. The endothelial cell network in the capillary bed and the arterial and venous vessels provides a metabolically highly active barrier that controls the migration of immune cells, regulates vascular tone and permeability, and participates in the remodeling processes. Phenotypically and functionally altered endothelial cells, and remodeled vessels, can be found in acute and chronic lung diseases, although to different degrees, likely because of disease-specific mechanisms. Since vascular remodeling is associated with pulmonary hypertension, which worsens patient outcomes and survival, it is crucial to understand the underlying vascular alterations. In this Review, we describe the current knowledge regarding the role of the pulmonary vasculature in the development and progression of ARDS, PF, and COPD; we also outline future research directions with the hope of facilitating the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Izabela Borek
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Norbert F. Voelkel
- Pulmonary Medicine Department, University of Amsterdam Medical Centers, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, German Lung Center (DZL), Cardiopulmonary Institute, Giessen, Germany
| |
Collapse
|
27
|
Yang K, Liu S, Yan H, Lu W, Shan X, Chen H, Bao C, Feng H, Liao J, Liang S, Xu L, Tang H, Yuan JXJ, Zhong N, Wang J. SARS-CoV-2 spike protein receptor-binding domain perturbates intracellular calcium homeostasis and impairs pulmonary vascular endothelial cells. Signal Transduct Target Ther 2023; 8:276. [PMID: 37452066 PMCID: PMC10349149 DOI: 10.1038/s41392-023-01556-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/09/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to the spike protein or receptor-binding domain (S-RBD) of SARS-CoV-2 significantly influences endothelial cells and induces pulmonary vascular endotheliopathy. In this study, angiotensin-converting enzyme 2 humanized inbred (hACE2 Tg) mice and cultured pulmonary vascular endothelial cells were used to investigate how spike protein/S-RBD impacts pulmonary vascular endothelium. Results show that S-RBD leads to acute-to-prolonged induction of the intracellular free calcium concentration ([Ca2+]i) via acute activation of TRPV4, and prolonged upregulation of mechanosensitive channel Piezo1 and store-operated calcium channel (SOCC) key component Orai1 in cultured human pulmonary arterial endothelial cells (PAECs). In mechanism, S-RBD interacts with ACE2 to induce formation of clusters involving Orai1, Piezo1 and TRPC1, facilitate the channel activation of Piezo1 and SOCC, and lead to elevated apoptosis. These effects are blocked by Kobophenol A, which inhibits the binding between S-RBD and ACE2, or intracellular calcium chelator, BAPTA-AM. Blockade of Piezo1 and SOCC by GsMTx4 effectively protects the S-RBD-induced pulmonary microvascular endothelial damage in hACE2 Tg mice via normalizing the elevated [Ca2+]i. Comparing to prototypic strain, Omicron variants (BA.5.2 and XBB) of S-RBD induces significantly less severe cell apoptosis. Transcriptomic analysis indicates that prototypic S-RBD confers more severe acute impacts than Delta or Lambda S-RBD. In summary, this study provides compelling evidence that S-RBD could induce persistent pulmonary vascular endothelial damage by binding to ACE2 and triggering [Ca2+]i through upregulation of Piezo1 and Orai1. Targeted inhibition of ACE2-Piezo1/SOCC-[Ca2+]i axis proves a powerful strategy to treat S-RBD-induced pulmonary vascular diseases.
Collapse
Affiliation(s)
- Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Han Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haixia Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pathology, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Changlei Bao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Liao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Escaffre O, Szaniszlo P, Törő G, Vilas CL, Servantes BJ, Lopez E, Juelich TL, Levine CB, McLellan SLF, Cardenas JC, Freiberg AN, Módis K. Hydrogen Sulfide Ameliorates SARS-CoV-2-Associated Lung Endothelial Barrier Disruption. Biomedicines 2023; 11:1790. [PMID: 37509430 PMCID: PMC10376201 DOI: 10.3390/biomedicines11071790] [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: 05/18/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have confirmed that lung microvascular endothelial injury plays a critical role in the pathophysiology of COVID-19. Our group and others have demonstrated the beneficial effects of H2S in several pathological processes and provided a rationale for considering the therapeutic implications of H2S in COVID-19 therapy. Here, we evaluated the effect of the slow-releasing H2S donor, GYY4137, on the barrier function of a lung endothelial cell monolayer in vitro, after challenging the cells with plasma samples from COVID-19 patients or inactivated SARS-CoV-2 virus. We also assessed how the cytokine/chemokine profile of patients' plasma, endothelial barrier permeability, and disease severity correlated with each other. Alterations in barrier permeability after treatments with patient plasma, inactivated virus, and GYY4137 were monitored and assessed by electrical impedance measurements in real time. We present evidence that GYY4137 treatment reduced endothelial barrier permeability after plasma challenge and completely reversed the endothelial barrier disruption caused by inactivated SARS-CoV-2 virus. We also showed that disease severity correlated with the cytokine/chemokine profile of the plasma but not with barrier permeability changes in our assay. Overall, these data demonstrate that treatment with H2S-releasing compounds has the potential to ameliorate SARS-CoV-2-associated lung endothelial barrier disruption.
Collapse
Affiliation(s)
- Olivier Escaffre
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Peter Szaniszlo
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabor Törő
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Caitlyn L. Vilas
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Brenna J. Servantes
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ernesto Lopez
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Terry L. Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Corri B. Levine
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Susan L. F. McLellan
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica C. Cardenas
- The Center for Translational Injury Research, Department of Surgery, UTHealth McGovern Medical School, Houston, TX 77030, USA
| | - Alexander N. Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, Sealy & Smith Foundation, University of Texas Medical Branch, Galveston, TX 77555, USA
- The Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Katalin Módis
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
29
|
Sjöland H, Lindgren M, Toska T, Hansson PO, Glise Sandblad K, Alex C, Björck L, Cronie O, Björk J, Lundberg CE, Adiels M, Rosengren A. Pulmonary embolism and deep venous thrombosis after COVID-19: long-term risk in a population-based cohort study. Res Pract Thromb Haemost 2023; 7:100284. [PMID: 37361398 PMCID: PMC10284449 DOI: 10.1016/j.rpth.2023.100284] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/23/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Background Venous thromboembolism (VTE) (pulmonary embolism (PE) or deep venous thrombosis (DVT)) is common during acute COVID-19. Long-term excess risk has not yet been established. Objective To study long-term VTE risk after COVID-19. Methods Swedish citizens aged 18-84 years, hospitalized and/or testing positive for COVID-19 between January 1, 2020, and September 11, 2021 (exposed), stratified by initial hospitalization, were compared to matched (1:5) non-exposed population-derived subjects without COVID-19. Outcomes were incident VTE, PE or DVT recorded within 60, 60-<180, and ≥180 days. Cox regression was used for evaluation and a model adjusted for age, sex, comorbidities and socioeconomic markers developed to control for confounders. Results Among exposed patients, 48,861 were hospitalized for COVID-19 (mean age 60.6 years) and 894,121 were without hospitalization (mean age 41.4 years). Among patients hospitalized for COVID-19, fully adjusted hazard ratios (HRs) during 60-<180 days were 6.05 (95% confidence interval (CI) 4.80─7.62) for PE and 3.97 (CI 2.96─5.33) for DVT, compared to non-exposed with corresponding estimates among COVID-19 without hospitalization 1.17 (CI 1.01─1.35) and 0.99 (CI 0.86─1.15), based on 475 and 2,311 VTE events, respectively. Long-term (≥180 days) HRs in patients hospitalized for COVID-19 were 2.01 (CI 1.51─2.68) for PE and 1.46 (CI 1.05─2.01) for DVT while non-hospitalized had similar risk to non-exposed, based on 467 and 2,030 VTE events, respectively. Conclusions Patients hospitalized for COVID-19 retained an elevated excess risk of VTE, mainly PE, after 180 days, while long-term risk of VTE in individuals with COVID-19 without hospitalization was similar to the non-exposed.
Collapse
Affiliation(s)
- Helen Sjöland
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Martin Lindgren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Triantafyllia Toska
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Per-Olof Hansson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Katarina Glise Sandblad
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Christian Alex
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Lena Björck
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| | - Ottmar Cronie
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Björk
- Division of Occupational and Environmental Medicine, Lund University, Lund, Sweden
- Clinical Studies Sweden, Forum South, Skåne University Hospital, Lund, Sweden
| | - Christina E. Lundberg
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Food and Nutrition, and Sport Science, University of Gothenburg, Gothenburg, Sweden
| | - Martin Adiels
- School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medicine Geriatrics and Emergency Medicine, Sahlgrenska University Hospital, Östra Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
30
|
Roozbehani M, Keyvani H, Razizadeh M, Yousefi P, Gholami A, Tabibzadeh A, Mousavizadeh L. LZTFL1 rs17713054 Polymorphism as an Indicator Allele for COVID-19 Severity. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY 2023; 38:124-128. [DOI: 10.3103/s0891416823020088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/14/2023] [Indexed: 01/02/2025]
|
31
|
Oliynyk OV, Rorat M, Strepetova OV, Dubrov SO, Guryanov VG, Oliynyk YV, Kulivets OS, Ślifirczyk A, Barg W. Efficacy of Sildenafil in Patients with Severe COVID-19 and Pulmonary Arterial Hypertension. Viruses 2023; 15:v15051157. [PMID: 37243243 DOI: 10.3390/v15051157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is common in severe coronavirus disease 2019 (COVID-19) and worsens the prognosis. Sildenafil, a phosphodiesterase-5 inhibitor, is approved for PAH treatment but little is known about its efficacy in cases of severe COVID-19 with PAH. This study aimed to investigate the clinical efficacy of sildenafil in patients with severe COVID-19 and PAH. Intensive care unit (ICU) patients were randomly assigned to receive sildenafil or a placebo, with 75 participants in each group. Sildenafil was administered orally at 0.25 mg/kg t.i.d. for one week in a placebo-controlled, double-blind manner as an add-on therapy alongside the patient's routine treatment. The primary endpoint was one-week mortality, and the secondary endpoints were the one-week intubation rate and duration of ICU stay. The mortality rate was 4% vs. 13.3% (p = 0.078), the intubation rate was 8% and 18.7% (p = 0.09), and the length of ICU stay was 15 vs. 19 days (p < 0.001) for the sildenafil and placebo groups, respectively. If adjusted for PAH, sildenafil treatment significantly reduced mortality and intubation risks: OR = 0.21 (95% CI: 0.05-0.89) and OR = 0.26 (95% CI: 0.08-0.86), respectively. Sildenafil demonstrated some clinical efficacy in patients with severe COVID-19 and PAH and should be considered as an add-on therapy in these patients.
Collapse
Affiliation(s)
- Oleksandr Valentynovych Oliynyk
- Department of Anaesthesiology and Intensive Care, Bogomolets National Medical University, 01601 Kyiv, Ukraine
- Department of Emergency Medicine, Rzeszow University, 35-310 Rzeszow, Poland
| | - Marta Rorat
- Department of Forensic Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Olena Vadymivna Strepetova
- Department of Anaesthesiology and Intensive Care, Bogomolets National Medical University, 01601 Kyiv, Ukraine
- Commercial Hospital "Manufaktura", 08173 Kyiv, Ukraine
- Commercial Hospital "Raiering", 02121 Kyiv, Ukraine
| | | | | | | | | | - Anna Ślifirczyk
- Nursing Department, Siedlce University of Natural Sciences and Humanities, 08-110 Siedlce, Poland
| | - Wojciech Barg
- Department of Human Physiology, Rzeszow University, 35-310 Rzeszow, Poland
| |
Collapse
|
32
|
Shaikh SB, Goracci C, Tjitropranoto A, Rahman I. Impact of aging on immune function in the pathogenesis of pulmonary diseases: potential for therapeutic targets. Expert Rev Respir Med 2023; 17:351-364. [PMID: 37078192 PMCID: PMC10330361 DOI: 10.1080/17476348.2023.2205127] [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: 10/06/2022] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION Several immunological alterations that occur during pulmonary diseases often mimic alterations observed in the aged lung. From the molecular perspective, pulmonary diseases and aging partake in familiar mechanisms associated with significant dysregulation of the immune systems. Here, we summarized the findings of how aging alters immunity to respiratory conditions to identify age-impacted pathways and mechanisms that contribute to the development of pulmonary diseases. AREAS COVERED The current review examines the impact of age-related molecular alterations in the aged immune system during various lung diseases, such as COPD, IPF, Asthma, and alongside many others that could possibly improve on current therapeutic interventions. Moreover, our increased understanding of this phenomenon may play a primary role in shaping immunomodulatory strategies to boost outcomes in the elderly. Here, the authors present new insights into the context of lung-related diseases and describe the alterations in the functioning of immune cells during various pulmonary conditions altered with age. EXPERT OPINION The expert opinion provided the concepts on how aging alters immunity during pulmonary conditions, and suggests the associated mechanisms during the development of lung diseases. As a result, it becomes important to comprehend the complex mechanism of aging in the immune lung system.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Chiara Goracci
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ariel Tjitropranoto
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
33
|
Pizzuto DA, Buonsenso D, Morello R, De Rose C, Valentini P, Fragano A, Baldi F, Di Giuda D. Lung perfusion assessment in children with long-COVID: A pilot study. Pediatr Pulmonol 2023. [PMID: 37097045 DOI: 10.1002/ppul.26432] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/09/2023] [Accepted: 04/16/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND There is increasing evidence that chronic endotheliopathy can play a role in patients with Post-Covid Condition (PCC, or Long Covid) by affecting peripheral vascularization. This pilot study aimed at assessing lung perfusion in children with Long-COVID with 99m Tc-MAA SPECT/CT. MATERIALS AND METHODS lung 99m Tc-MAA SPECT/CT was performed in children with Long-COVID and a pathological cardiopulmonary exercise testing (CPET). Intravenous injections were performed on patients in the supine position immediately before the planar scan according to the EANM guidelines for lung scintigraphy in children, followed by lung SPECT/CT acquisition. Reconstructed studies were visually analyzed. RESULTS Clinical and biochemical data were collected during acute infection and follow-up in 14 children (6 females, mean age: 12.6 years) fulfilling Long-COVID diagnostic criteria and complaining of chronic fatigue and postexertional malaise after mild efforts, documented by CPET. Imaging results were compared with clinical scenarios during acute infection and follow-up. Six out of 14 (42.8%) children showed perfusion defects on 99m Tc-MAA SPECT/CT scan, without morphological alterations on coregistered CT. CONCLUSIONS This pilot investigation confirmed previous data suggesting that a small subgroup of children can develop lung perfusion defects after severe acute respiratory syndrome coronavirus 2 infection. Larger cohort studies are needed to confirm these preliminary results, providing also a better understanding of which children may deserve this test and how to manage those with lung perfusion defects.
Collapse
Affiliation(s)
- Daniele Antonio Pizzuto
- Department of Radiology, Radiotherapy and Hematology, Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
- GlobalHealth Research Institute, Istituto di Igiene, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rosa Morello
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Cristina De Rose
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Piero Valentini
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - A Fragano
- Department of Radiology, Radiotherapy and Hematology, Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Fabiana Baldi
- Division of Respiratory Medicine, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Daniela Di Giuda
- Department of Radiology, Radiotherapy and Hematology, Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- University Department of Radiological Sciences and Hematology, Section of Nuclear Medicine, Uniiversità Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
34
|
Wang D, Gomes MT, Mo Y, Prohaska CC, Zhang L, Chelvanambi S, Clauss MA, Zhang D, Machado RF, Gao M, Bai Y. Human Endogenous Retrovirus, SARS-CoV-2, and HIV Promote PAH via Inflammation and Growth Stimulation. Int J Mol Sci 2023; 24:7472. [PMID: 37108634 PMCID: PMC10138839 DOI: 10.3390/ijms24087472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/05/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a pulmonary vascular disease characterized by the progressive elevation of pulmonary arterial pressures. It is becoming increasingly apparent that inflammation contributes to the pathogenesis and progression of PAH. Several viruses are known to cause PAH, such as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), human endogenous retrovirus K(HERV-K), and human immunodeficiency virus (HIV), in part due to acute and chronic inflammation. In this review, we discuss the connections between HERV-K, HIV, SARS-CoV-2, and PAH, to stimulate research regarding new therapeutic options and provide new targets for the treatment of the disease.
Collapse
Affiliation(s)
- Desheng Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Marta T. Gomes
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Yanfei Mo
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Clare C. Prohaska
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Lu Zhang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Sarvesh Chelvanambi
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthias A. Clauss
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Dongfang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Mingqi Gao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yang Bai
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
35
|
Chiou WC, Huang GJ, Chang TY, Hsia TL, Yu HY, Lo JM, Fu PK, Huang C. Ovatodiolide inhibits SARS-CoV-2 replication and ameliorates pulmonary fibrosis through suppression of the TGF-β/TβRs signaling pathway. Biomed Pharmacother 2023; 161:114481. [PMID: 36906971 PMCID: PMC9998303 DOI: 10.1016/j.biopha.2023.114481] [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: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection continues to pose threats to public health. The clinical manifestations of lung pathology in COVID-19 patients include sustained inflammation and pulmonary fibrosis. The macrocyclic diterpenoid ovatodiolide (OVA) has been reported to have anti-inflammatory, anti-cancer, anti-allergic, and analgesic activities. Here, we investigated the pharmacological mechanism of OVA in suppressing SARS-CoV-2 infection and pulmonary fibrosis in vitro and in vivo. Our results revealed that OVA was an effective SARS-CoV-2 3CLpro inhibitor and showed remarkable inhibitory activity against SARS-CoV-2 infection. On the other hand, OVA ameliorated pulmonary fibrosis in bleomycin (BLM)-induced mice, reducing inflammatory cell infiltration and collagen deposition in the lung. OVA decreased the levels of pulmonary hydroxyproline and myeloperoxidase, as well as lung and serum TNF-ɑ, IL-1β, IL-6, and TGF-β in BLM-induced pulmonary fibrotic mice. Meanwhile, OVA reduced the migration and fibroblast-to-myofibroblast conversion of TGF-β1-induced fibrotic human lung fibroblasts. Consistently, OVA downregulated TGF-β/TβRs signaling. In computational analysis, OVA resembles the chemical structures of the kinase inhibitors TβRI and TβRII and was shown to interact with the key pharmacophores and putative ATP-binding domains of TβRI and TβRII, showing the potential of OVA as an inhibitor of TβRI and TβRII kinase. In conclusion, the dual function of OVA highlights its potential for not only fighting SARS-CoV-2 infection but also managing injury-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei-Chung Chiou
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan.
| | - Guan-Jhong Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung City 404333, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung City 413305, Taiwan.
| | - Tein-Yao Chang
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 237010, Taiwan.
| | - Tzu-Lan Hsia
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan.
| | - Hao-You Yu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan.
| | - Jir-Mehng Lo
- Industrial Technology Research Institute, Biomedical Technology and Device Research Laboratories, Hsinchu City 310401, Taiwan.
| | - Pin-Kuei Fu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung City 402010, Taiwan; Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407219, Taiwan; Integrated Care Center of Interstitial Lung Disease, Taichung Veterans General Hospital, Taichung City 407219, Taiwan; College of Human Science and Social Innovation, Hungkuang University, Taichung City 433304, Taiwan.
| | - Cheng Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei City 112304, Taiwan.
| |
Collapse
|
36
|
Coman AE, Ceasovschih A, Petroaie AD, Popa E, Lionte C, Bologa C, Haliga RE, Cosmescu A, Slănină AM, Bacușcă AI, Șorodoc V, Șorodoc L. The Significance of Low Magnesium Levels in COVID-19 Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020279. [PMID: 36837480 PMCID: PMC9965430 DOI: 10.3390/medicina59020279] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/22/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
Magnesium is the fourth most common mineral in the human body and the second richest intracellular cation. This element is necessary for many physiological reactions, especially in the cardiovascular and respiratory systems. COVID-19 is an infectious disease caused by SARS-CoV-2. The majority of people who become ill as a result of COVID-19 have mild-to-moderate symptoms and recover without specific treatment. Moreover, there are people who develop severe forms of COVID-19, which require highly specialized medical assistance. Magnesium deficiency may play a role in the pathophysiology of infection with SARS-CoV-2. The primary manifestation of COVID-19 remains respiratory, but the virus can spread to other organs and tissues, complicating the clinical picture and culminating in multiorgan failure. The key mechanisms involved in the disease include direct viral cytotoxicity, endothelial dysfunction, and exaggerated release of inflammatory cytokines. The aim of this review was to summarize the available data regarding the role of magnesium in COVID-19 patients and its particularities in different clinical settings.
Collapse
Affiliation(s)
- Adorata Elena Coman
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Alexandr Ceasovschih
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (A.C.); (C.L.)
| | - Antoneta Dacia Petroaie
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Elena Popa
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Cătălina Lionte
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (A.C.); (C.L.)
| | - Cristina Bologa
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Raluca Ecaterina Haliga
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Adriana Cosmescu
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Ana Maria Slănină
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Agnes Iacinta Bacușcă
- Preventive Medicine and Interdisciplinarity Department, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania
| | - Victorița Șorodoc
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laurențiu Șorodoc
- 2nd Internal Medicine Department, Sf. Spiridon Clinical Emergency Hospital, 700111 Iasi, Romania
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
37
|
Wang R, Loscalzo J. Uncovering common pathobiological processes between COVID-19 and pulmonary arterial hypertension by integrating Omics data. Pulm Circ 2023; 13:e12191. [PMID: 36721384 PMCID: PMC9880519 DOI: 10.1002/pul2.12191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/15/2022] [Accepted: 01/01/2023] [Indexed: 01/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which led to the current pandemic. Many factors, including age and comorbidities, influence the severity and mortality of COVID-19. SARS-CoV-2 infection can cause pulmonary vascular dysfunction. The COVID-19 case-fatality rate in patients with pulmonary arterial hypertension (PAH) is higher in comparison with the general population. In this study, we aimed to identify pathobiological processes common to COVID-19 and PAH by utilizing the human protein-protein interactome and whole-genome transcription data from peripheral blood mononuclear cells (PBMCs) and from lung tissue. We found that there are significantly more interactions between SARS-CoV-2 targets and PAH disease proteins than expected by chance, suggesting that the PAH disease module is in the neighborhood of SARS-CoV-2 targets in the human interactome. In addition, SARS-CoV-2 infection-induced changes in gene expression significantly overlap with PAH-induced gene expression changes in both tissues, indicating SARS-CoV-2 and PAH may share common transcriptional regulators. We identified many upregulated genes and downregulated genes common to COVID-19 and PAH. Interestingly, we observed different co-regulation patterns and dysfunctional signaling pathways in PBMCs versus lung tissue. Endophenotype enrichment analysis revealed that genes regulating fibrosis, inflammation, hypoxia, oxidative stress, immune response, and thromboembolism are significantly enriched in the COVID-19-PAH co-expression modules. We examined the network proximity of the targets of repositioned drugs for COVID-19 to the co-expression modules in PBMCs and lung tissue, and identified 42 drugs that can be potentially used for COVID-19 patients with PAH as a comorbidity. The uncovered common pathobiological pathways are crucial for discovering therapeutic targets and designing tailored treatments for COVID-19 patients who also have PAH.
Collapse
Affiliation(s)
- Rui‐Sheng Wang
- Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
38
|
Farkas D, Bogamuwa S, Piper B, Newcomb G, Gunturu P, Bednash JS, Londino JD, Elhance A, Nho R, Mejia OR, Yount JS, Horowitz JC, Goncharova EA, Mallampalli RK, Robinson RT, Farkas L. A role for Toll-like receptor 3 in lung vascular remodeling associated with SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.524586. [PMID: 36747676 PMCID: PMC9900759 DOI: 10.1101/2023.01.25.524586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cardiovascular sequelae of severe acute respiratory syndrome (SARS) coronavirus-2 (CoV-2) disease 2019 (COVID-19) contribute to the complications of the disease. One potential complication is lung vascular remodeling, but the exact cause is still unknown. We hypothesized that endothelial TLR3 insufficiency contributes to lung vascular remodeling induced by SARS-CoV-2. In the lungs of COVID-19 patients and SARS-CoV-2 infected Syrian hamsters, we discovered thickening of the pulmonary artery media and microvascular rarefaction, which were associated with decreased TLR3 expression in lung tissue and pulmonary artery endothelial cells (ECs). In vitro , SARS-CoV-2 infection reduced endothelial TLR3 expression. Following infection with mouse-adapted (MA) SARS-CoV-2, TLR3 knockout mice displayed heightened pulmonary artery remodeling and endothelial apoptosis. Treatment with the TLR3 agonist polyinosinic:polycytidylic acid reduced lung tissue damage, lung vascular remodeling, and endothelial apoptosis associated with MA SARS-CoV-2 infection. In conclusion, repression of endothelial TLR3 is a potential mechanism of SARS-CoV-2 infection associated lung vascular remodeling and enhancing TLR3 signaling is a potential strategy for treatment.
Collapse
|
39
|
Farmakis IT, Giannakoulas G. Management of COVID-19 in Patients with Pulmonary Arterial Hypertension. Heart Fail Clin 2023; 19:107-114. [PMID: 36435565 PMCID: PMC9364740 DOI: 10.1016/j.hfc.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we discuss the evidence regarding the course and management of COVID-19 in patients with pulmonary arterial hypertension (PAH), the challenges in PAH management during the pandemic and, lastly, the long-term complications of COVID-19 in relation to pulmonary vascular disease. The inherent PAH disease characteristics, as well as age, comorbidities, and the patient's functional status act synergistically to define the prognosis of COVID-19 in patients with PAH. Management of COVID-19 should follow the general guidelines, while PAH-targeted therapies should be continued. The pandemic has caused a shift toward telemedicine in the chronic care of patients with PAH. Whether COVID-19 could predispose to the development of chronic pulmonary hypertension is a subject of future investigation.
Collapse
Affiliation(s)
- Ioannis T. Farmakis
- Department of Cardiology, AHEPA University Hospital, Stilp. Kiriakidi 1, Thessaloniki 54637, Greece,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langebeckstr. 1, 55131, Mainz, Germany
| | - George Giannakoulas
- Department of Cardiology, AHEPA University Hospital, Stilp. Kiriakidi 1, Thessaloniki 54637, Greece,Corresponding author. Aristotle University of Thessaloniki, AHEPA Hospital, Cardiology Department, Stilp. Kiriakidi 1, Thessaloniki 54637, Greece
| |
Collapse
|
40
|
Eroume À Egom E, Shiwani HA, Nouthe B. From acute SARS-CoV-2 infection to pulmonary hypertension. Front Physiol 2022; 13:1023758. [PMID: 36601347 PMCID: PMC9806360 DOI: 10.3389/fphys.2022.1023758] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
As the world progressively recovers from the acute stages of the coronavirus disease 2019 (COVID-19) pandemic, we may be facing new challenges regarding the long-term consequences of COVID-19. Accumulating evidence suggests that pulmonary vascular thickening may be specifically associated with COVID-19, implying a potential tropism of severe acute respiratory syndrome coronavirus 2 (SARS-COV-2) virus for the pulmonary vasculature. Genetic alterations that may influence the severity of COVID-19 are similar to genetic drivers of pulmonary arterial hypertension. The pathobiology of the COVID-19-induced pulmonary vasculopathy shares many features (such as medial hypertrophy and smooth muscle cell proliferation) with that of pulmonary arterial hypertension. In addition, the presence of microthrombi in the lung vessels of individuals with COVID-19 during the acute phase, may predispose these subjects to the development of chronic thromboembolic pulmonary hypertension. These similarities raise the intriguing question of whether pulmonary hypertension (PH) may be a long-term sequela of SARS-COV-2 infection. Accumulating evidence indeed support the notion that SARS-COV-2 infection is indeed a risk factor for persistent pulmonary vascular defects and subsequent PH development, and this could become a major public health issue in the future given the large number of individuals infected by SARS-COV-2 worldwide. Long-term studies assessing the risk of developing chronic pulmonary vascular lesions following COVID-19 infection is of great interest for both basic and clinical research and may inform on the best long-term management of survivors.
Collapse
Affiliation(s)
- Emmanuel Eroume À Egom
- Institut du Savoir Montfort (ISM), University of Ottawa, Ottawa, ON, Canada,CIEL, Centre d’Innovation et de Commercialisation en Recherche Clinique et Bio-Médicale Immânow’EL, Béatitude/Nkolbisson, Yaoundé, Cameroon,Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon,*Correspondence: Emmanuel Eroume À Egom,
| | - Haaris A. Shiwani
- Burnley General Hospital, East Lancashire Hospitals NHS Trust, Burnley, United Kingdom
| | - Brice Nouthe
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
41
|
Kim J, Rustam S, Mosquera JM, Randell SH, Shaykhiev R, Rendeiro AF, Elemento O. Unsupervised discovery of tissue architecture in multiplexed imaging. Nat Methods 2022; 19:1653-1661. [PMID: 36316562 PMCID: PMC11102857 DOI: 10.1038/s41592-022-01657-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022]
Abstract
Multiplexed imaging and spatial transcriptomics enable highly resolved spatial characterization of cellular phenotypes, but still largely depend on laborious manual annotation to understand higher-order patterns of tissue organization. As a result, higher-order patterns of tissue organization are poorly understood and not systematically connected to disease pathology or clinical outcomes. To address this gap, we developed an approach called UTAG to identify and quantify microanatomical tissue structures in multiplexed images without human intervention. Our method combines information on cellular phenotypes with the physical proximity of cells to accurately identify organ-specific microanatomical domains in healthy and diseased tissue. We apply our method to various types of images across healthy and disease states to show that it can consistently detect higher-level architectures in human tissues, quantify structural differences between healthy and diseased tissue, and reveal tissue organization patterns at the organ scale.
Collapse
Affiliation(s)
- Junbum Kim
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Samir Rustam
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Scott H Randell
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Renat Shaykhiev
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - André F Rendeiro
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
42
|
Chazova IE, Martynyuk TV, Gorbachevskii SV, Gramovich VV, Danilov NM, Panchenko EP, Chernyavskiy AM, Shmalts AA, Yavelov IS. ["Guiding lights" for the diagnosis of chronic thromboembolic pulmonary hypertension in the flow of patients with pulmonary embolism]. TERAPEVT ARKH 2022; 94:1052-1056. [PMID: 36286754 DOI: 10.26442/00403660.2022.09.201836] [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: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
On December 13, 2021, an expert council was held to determine the position of experts of different specialties regarding the reasons for the low level of diagnosis of chronic thromboembolic pulmonary hypertension (CTEPH) in real clinical practice in a pandemic of a new coronavirus infection and possible ways to improve detection in patients with pulmonary embolism (PE) ) in history. The reasons for the low level of diagnosis of CTEPH are the insufficient level of knowledge of specialists, especially primary care physicians; lack of clear regulatory documents and expert centers for the management of this category of patients. Primary diagnosis of CTEPH in a pandemic can be strengthened through the widespread use of telemedicine for consultations of primary care physicians with specialists from expert centers; to maximize the role of echocardiography and computed tomography (CT) as differential diagnostic tools for dyspnea, in particular in patients with COVID-19. To increase the detection rate of CTEPH, diagnostic vigilance is required in patients with risk factors and episodes of venous thromboembolism. To improve the screening of CTEPH, it is necessary to create an algorithm for monitoring patients who have had PE; provide educational activities, including through the media; create materials for patients with accessible information. The regulatory documents should designate the circle of responsible specialists who will be engaged in long-term monitoring of patients with PE. Educational programs are needed for primary care physicians, cardiologists, and other physicians who come into the field of view of patients with CTEPH; introduction of a program to create expert centers for monitoring and managing patients with the possibility of performing ventilation-perfusion lung scintigraphy, cardiopulmonary stress test, CT, right heart catheterization. It seems important to build cooperation with the Ministry of Health of Russia in order to create special protocols, procedures for managing patients with PE and CTEPH.
Collapse
Affiliation(s)
- I E Chazova
- Chazov National Medical Research Center of Cardiology
| | - T V Martynyuk
- Chazov National Medical Research Center of Cardiology
- The Russian National Research Medical University named after N.I. Pirogov
| | - S V Gorbachevskii
- Bakoulev Scientific Center for Cardiovascular Surgery
- Russian Medical Academy of Continuous Professional Education
| | - V V Gramovich
- Chazov National Medical Research Center of Cardiology
| | - N M Danilov
- Chazov National Medical Research Center of Cardiology
| | - E P Panchenko
- Chazov National Medical Research Center of Cardiology
| | | | - A A Shmalts
- Bakoulev Scientific Center for Cardiovascular Surgery
- Russian Medical Academy of Continuous Professional Education
| | - I S Yavelov
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
43
|
Chavda VP, Patel AB, Pandya A, Vora LK, Patravale V, Tambuwala ZM, Aljabali AAA, Serrano-Aroca Á, Mishra V, Tambuwala MM. Co-infection associated with SARS-CoV-2 and their management. Future Sci OA 2022; 8:FSO819. [PMID: 36788985 PMCID: PMC9912272 DOI: 10.2144/fsoa-2022-0011] [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: 03/02/2022] [Accepted: 10/18/2022] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 was discovered in Wuhan, China and quickly spread throughout the world. This deadly virus moved from person to person, resulting in severe pneumonia, fever, chills and hypoxia. Patients are still experiencing problems after recovering from COVID-19. This review covers COVID-19 and associated issues following recovery from COVID-19, as well as multiorgan damage risk factors and treatment techniques. Several unusual illnesses, including mucormycosis, white fungus infection, happy hypoxia and other systemic abnormalities, have been reported in recovered individuals. In children, multisystem inflammatory syndrome with COVID-19 (MIS-C) is identified. The reasons for this might include uncontrollable steroid usage, reduced immunity, uncontrollable diabetes mellitus and inadequate care following COVID-19 recovery.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Aayushi B Patel
- Pharmacy Section, LM College of Pharmacy, Ahmedabad, Gujarat, 380058, India
| | - Anjali Pandya
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Vandana Patravale
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Zara M Tambuwala
- College of Science, University of Lincoln, Brayford Campus, Lincoln, LN6 7TS, UK
| | - Alaa AA Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Faculty of Pharmacy, Irbid, 566, Jordan
| | - Ángel Serrano-Aroca
- Biomaterials & Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia, 46001, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Murtaza M Tambuwala
- Lincoln Medical School University of Lincoln, Brayford Campus, Lincoln, LN6 7TS, UK
| |
Collapse
|
44
|
Hinojosa W, Cristo‐Ropero MJ, Cruz‐Utrilla A, Segura de la Cal T, López‐Medrano F, Salguero‐Bodes R, Pérez‐Olivares C, Navarro B, Ochoa N, Arribas Ynsurriaga F, Escribano‐Subias P. The impact of COVID-19 pandemic on pulmonary hypertension: What have we learned? Pulm Circ 2022; 12:e12142. [PMID: 36238968 PMCID: PMC9537953 DOI: 10.1002/pul2.12142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022] Open
Abstract
The coronavirus 2019 disease (COVID-19) pandemic threatened the Spanish health-care system. Patients with demanding conditions such as precapillary pulmonary hypertension (PH) faced a potentially severe infection, while their usual access to medical care was restricted. This prospective, unicentric study assessed the impact of COVID-19 on PH patients' outcomes and the operational changes in the PH network. Sixty-three PH patients (41 pulmonary arterial hypertension [PAH]; 22 chronic thromboembolic pulmonary hypertension [CTEPH]) experienced COVID-19. Overall mortality was 9.5% without differences when stratifying by hemodynamics or PAH-risk score. Patients who died were older (73.6 ± 5 vs. 52.2 ± 15.4; p = 0.001), with more comorbidities (higher Charlson index: 4.17 ± 2.48 vs. 1.14 ± 1.67; p = 0.0002). Referrals to the PH expert center decreased compared to the previous 3 years (123 vs. 160; p = 0.002). The outpatient activity shifted toward greater use of telemedicine. Balloon pulmonary angioplasty activity could be maintained after the first pandemic wave and lockdown while pulmonary thromboendarterectomy procedures decreased (19 vs. 36; p = 0.017). Pulmonary transplantation activity remained similar. The COVID-19 mortality in PAH/CTEPH patients was not related to hemodynamic severity or risk stratification, but to comorbidities. The pandemic imposed structural changes but a planned organization and resource reallocation made it possible to maintain PH patients' care.
Collapse
Affiliation(s)
- Williams Hinojosa
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | | | - Alejandro Cruz‐Utrilla
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | - Teresa Segura de la Cal
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | - Francisco López‐Medrano
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Infectious Diseases UnitHospital Universitario 12 de OctubreMadridSpain
| | - Rafael Salguero‐Bodes
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Medicine Department, School of MedicineComplutense UniversityMadridSpain
| | - Carmen Pérez‐Olivares
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | - Begoña Navarro
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | - Nuria Ochoa
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| | - Fernando Arribas Ynsurriaga
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Medicine Department, School of MedicineComplutense UniversityMadridSpain
| | - Pilar Escribano‐Subias
- Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
- Servicio Madrileño de SaludCentro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
- Pulmonary Hypertension Multidisciplinary Unit, Cardiology DepartmentHospital Universitario 12 de OctubreMadridSpain
| |
Collapse
|
45
|
Rao S, Lee GM, Razzaghi H, Lorman V, Mejias A, Pajor NM, Thacker D, Webb R, Dickinson K, Bailey LC, Jhaveri R, Christakis DA, Bennett TD, Chen Y, Forrest CB. Clinical Features and Burden of Postacute Sequelae of SARS-CoV-2 Infection in Children and Adolescents. JAMA Pediatr 2022; 176:1000-1009. [PMID: 35994282 PMCID: PMC9396470 DOI: 10.1001/jamapediatrics.2022.2800] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/08/2022] [Indexed: 01/20/2023]
Abstract
Importance The postacute sequelae of SARS-CoV-2 infection (PASC) has emerged as a long-term complication in adults, but current understanding of the clinical presentation of PASC in children is limited. Objective To identify diagnosed symptoms, diagnosed health conditions, and medications associated with PASC in children. Design, Setting and Participants This retrospective cohort study used electronic health records from 9 US children's hospitals for individuals younger than 21 years who underwent antigen or reverse transcriptase-polymerase chain reaction (RT-PCR) testing for SARS-CoV-2 between March 1, 2020, and October 31, 2021, and had at least 1 encounter in the 3 years before testing. Exposures SARS-CoV-2 positivity by viral test (antigen or RT-PCR). Main Outcomes and Measures Syndromic (symptoms), systemic (conditions), and medication PASC features were identified in the 28 to 179 days following the initial test date. Adjusted hazard ratios (aHRs) were obtained for 151 clinically predicted PASC features by contrasting viral test-positive groups with viral test-negative groups using proportional hazards models, adjusting for site, age, sex, testing location, race and ethnicity, and time period of cohort entrance. The incidence proportion for any syndromic, systemic, or medication PASC feature was estimated in the 2 groups to obtain a burden of PASC estimate. Results Among 659 286 children in the study sample, 348 091 (52.8%) were male, and the mean (SD) age was 8.1 (5.7) years. A total of 59 893 (9.1%) tested positive by viral test for SARS-CoV-2, and 599 393 (90.9%) tested negative. Most were tested in outpatient testing facility settings (322 813 [50.3%]) or office settings (162 138 [24.6%]). The most common syndromic, systemic, and medication features were loss of taste or smell (aHR, 1.96; 95% CI, 1.16-3.32), myocarditis (aHR, 3.10; 95% CI, 1.94-4.96), and cough and cold preparations (aHR, 1.52; 95% CI, 1.18-1.96), respectively. The incidence of at least 1 systemic, syndromic, or medication feature of PASC was 41.9% (95% CI, 41.4-42.4) among viral test-positive children vs 38.2% (95% CI, 38.1-38.4) among viral test-negative children, with an incidence proportion difference of 3.7% (95% CI, 3.2-4.2). A higher strength of association for PASC was identified in those cared for in the intensive care unit during the acute illness phase, children younger than 5 years, and individuals with complex chronic conditions. Conclusions and Relevance In this large-scale, exploratory study, the burden of pediatric PASC that presented to health systems was low. Myocarditis was the most commonly diagnosed PASC-associated condition. Acute illness severity, young age, and comorbid complex chronic disease increased the risk of PASC.
Collapse
Affiliation(s)
- Suchitra Rao
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora
| | - Grace M. Lee
- Department of Pediatrics (Infectious Diseases), Stanford University School of Medicine, Stanford, California
| | - Hanieh Razzaghi
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Vitaly Lorman
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Asuncion Mejias
- Division of Infectious Diseases, Department of Pediatrics, Nationwide Children’s Hospital and The Ohio State University, Columbus
| | - Nathan M. Pajor
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Deepika Thacker
- Division of Cardiology, Nemours Children’s Health, Wilmington, Delaware
| | - Ryan Webb
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kimberley Dickinson
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - L. Charles Bailey
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ravi Jhaveri
- Division of Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Dimitri A. Christakis
- Center for Child Health, Behavior and Development, Seattle Children’s Hospital, Seattle, Washington
- Editor, JAMA Pediatrics
| | - Tellen D. Bennett
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics, the Perelman School of Medicine, University of Pennsylvania, Pennsylvania
| | - Christopher B. Forrest
- Applied Clinical Research Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
46
|
Mickael C, Lee MH, Graham BB. The COVID-19 pandemic and pulmonary arterial hypertension in Italy: adaptation, outcomes and valuable lessons learned. Eur Respir J 2022; 60:2200796. [PMID: 36202406 PMCID: PMC9924355 DOI: 10.1183/13993003.00796-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 11/05/2022]
Abstract
This editorial summarises the key findings of R. Badagliacca and co-workers regarding the changes in care and clinical outcomes of PAH patients during the initial COVID-19 pandemic in Italy https://bit.ly/3ytgIjk
Collapse
Affiliation(s)
- Claudia Mickael
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael H Lee
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Brian B Graham
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
47
|
Moraes ECDS, Martins-Gonçalves R, da Silva LR, Mandacaru SC, Melo RM, Azevedo-Quintanilha I, Perales J, Bozza FA, Souza TML, Castro-Faria-Neto HC, Hottz ED, Bozza PT, Trugilho MRO. Proteomic Profile of Procoagulant Extracellular Vesicles Reflects Complement System Activation and Platelet Hyperreactivity of Patients with Severe COVID-19. Front Cell Infect Microbiol 2022; 12:926352. [PMID: 35937696 PMCID: PMC9354812 DOI: 10.3389/fcimb.2022.926352] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background Extracellular vesicles (EVs) are a valuable source of biomarkers and display the pathophysiological status of various diseases. In COVID-19, EVs have been explored in several studies for their ability to reflect molecular changes caused by SARS-CoV-2. Here we provide insights into the roles of EVs in pathological processes associated with the progression and severity of COVID-19. Methods In this study, we used a label-free shotgun proteomic approach to identify and quantify alterations in EV protein abundance in severe COVID-19 patients. We isolated plasma extracellular vesicles from healthy donors and patients with severe COVID-19 by size exclusion chromatography (SEC). Then, flow cytometry was performed to assess the origin of EVs and to investigate the presence of circulating procoagulant EVs in COVID-19 patients. A total protein extraction was performed, and samples were analyzed by nLC-MS/MS in a Q-Exactive HF-X. Finally, computational analysis was applied to signify biological processes related to disease pathogenesis. Results We report significant changes in the proteome of EVs from patients with severe COVID-19. Flow cytometry experiments indicated an increase in total circulating EVs and with tissue factor (TF) dependent procoagulant activity. Differentially expressed proteins in the disease groups were associated with complement and coagulation cascades, platelet degranulation, and acute inflammatory response. Conclusions The proteomic data reinforce the changes in the proteome of extracellular vesicles from patients infected with SARS-CoV-2 and suggest a role for EVs in severe COVID-19.
Collapse
Affiliation(s)
- Emilly Caroline dos Santos Moraes
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Samuel Coelho Mandacaru
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory Protein Chemistry and Biochemistry and Laboratory of Gene Biology, Department of Cell Biology, University of Brasília, Brasília, Brazil
| | | | - Jonas Perales
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- D’Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Thiago Moreno Lopes Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Eugenio D. Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Monique R. O. Trugilho
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Uusküla A, Jürgenson T, Pisarev H, Kolde R, Meister T, Tisler A, Suija K, Kalda R, Piirsoo M, Fischer K. Long-term mortality following SARS-CoV-2 infection: A national cohort study from Estonia. THE LANCET REGIONAL HEALTH. EUROPE 2022; 18:100394. [PMID: 35505834 PMCID: PMC9051903 DOI: 10.1016/j.lanepe.2022.100394] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND The objective of this study was to describe 12-month mortality following SARS-CoV-2 infection compared with a reference population with no history of SARS-CoV-2. METHODS Nationwide cohort study using electronic health care data on SARS-CoV-2 RNA positive cases (n= 66,287) and reference group subjects (n=254,969) with linkage to SARS-CoV-2 testing and death records. FINDINGS People infected with SARS-COV-2 had more than three times the risk of dying over the following year compared with those who remained uninfected (aHR 3·1, 95%CI 2·9-3·3). Short-term mortality (up to 5 weeks post-infection) was significantly higher among COVID-19 group (1623·0/10 000) than in the reference group (118/10 000). For COVID-19 cases aged 60 years or older, increased mortality persisted until the end of the first year after infection, and was related to increased risk for cardiovascular (aHR 2·1, 95%CI 1·8-2·3), cancer (aHR 1·5, 95%CI 1·2-1·9), respiratory system diseases (aHR 1·9, 95%CI 1·2-3·0), and other causes of death (aHR 1·8, 95%CI 1·4-2·2). INTERPRETATION Increased risk of death from SARS-CoV-2 is not limited to the acute illness: SARS-CoV-2 infection carries a substantially increased mortality in the following 12 months. This excess death mainly occurs in older people and is driven by broad array of causes of death. FUNDING Research was carried out with the support of Estonian Research Council (grants PRG1197, PRG198), European Regional Development Fund (RITA 1/02-120) and European Social Fund via IT Academy program.
Collapse
Affiliation(s)
- Anneli Uusküla
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
- Corresponding author at: Anneli Uusküla PhD, Department of Family Medicine and Public Health, University of Tartu, Tartu 50411, Estonia, +372 737 4195.
| | - Tuuli Jürgenson
- Institute of Mathematics and Statistics, University of Tartu, Estonia
- Institute of Genomics, University of Tartu, Estonia
| | - Heti Pisarev
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Raivo Kolde
- Institute of Computer Science, University of Tartu, Estonia
| | - Tatjana Meister
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Anna Tisler
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Kadri Suija
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Ruth Kalda
- Department of Family Medicine and Public Health, University of Tartu, Tartu, Estonia
| | - Marko Piirsoo
- Institute of Technology, University of Tartu, Estonia
| | | |
Collapse
|
49
|
Usami O. Two Cases of COVID-19-Related Deaths Unaccounted for: A Call for Action. Cureus 2022; 14:e26238. [PMID: 35911326 PMCID: PMC9312909 DOI: 10.7759/cureus.26238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) high-risk survivors experience long-term COVID-19 symptoms. Hence, these individuals require early and ubiquitous respiratory rehabilitation to avoid malnutrition. We report the case of a 93-year-old woman who recovered from moderate II severity (pneumonia requiring oxygen). The patient, after prolonged hospitalization, demonstrated low severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infectivity and showed no COVID-19 respiratory symptoms for more than 72 hours. Subsequently, the patient became debilitated and lost her appetite without dysphagia, dysgeusia, and smell disorder, developed nosocomial pneumonia as a sequela of acute COVID-19 and died. We also report the second case of an 84-year-old man diagnosed with moderate II COVID-19 severity. After recovery, the patient was frail due to the previous onset of COVID-19 and worsened during his stay at home, losing appetite without dysphagia, dysgeusia, and smell disorder, and dying of senility as the official cause. Recovered COVID-19 appears to be a health risk by malnutrition without anorexia and depression, among other conditions. A proven rehabilitation program for each phase of the disease is required for better lung function and nutritional status.
Collapse
|
50
|
Monro M. A long COVID patient and their experience of osteopathic care: A case report. INT J OSTEOPATH MED 2022. [DOI: 10.1016/j.ijosm.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|