1
|
Gulubova M, Merhar V, Chonov D, Mitev M, Pekova L, Ananiev J. Histopathological examination of lung from infant with lethal COVID-19 with special attention on pneumocytes type II and the immune infiltrate: a case study. Ital J Pediatr 2025; 51:174. [PMID: 40483512 PMCID: PMC12145646 DOI: 10.1186/s13052-025-01984-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND COVID-19 is a complex disease caused by SARS-CoV-2. The molecular and cellular mechanisms of the disease are unclear and their study is one of the greatest challenges for the modern science. Since the lung is the biggest target for SARS-CoV-2, the studies on cellular and molecular changes in this organ are essential to establish the pathogenesis of the disease. To date there is increasing number of reports on the lung pathology of fatal COVID-19 and the results are mainly obtained by autopsies of elderly patients, since this age group shows highest mortality. Little is known about the progression of the disease in children and especially newborn and infants and, to our knowledge, there are no reports on the lung features of fatal COVID-19 in this age group. METHODS In the present case study, we have investigated the lung morphological features in 11-months old infant who has died as a result of complications from COVID-19. Immunohistochemistry for immune cell markers and transmission electron microscopy for alveolocytes type II (ATII) are made. RESULTS Immediate cause of the death was acute respiratory failure resulting from bilateral interstitial pneumonia and subsequent acute cardiovascular failure. The histopathology shows lung edema, hyaline membranes, airway mucus plugging and interstitial inflammation. On cellular level we have observed a substantial increase in the number of ATII cells. ATII cells were marked with cytokeratin 19, TTF1 and napsin A. Transmission elec-tron microscopy reveals ongoing apoptosis in these cells with a typical chromatin clustering and condensation towards the inner nuclear membrane. Immunohisto-chemistry shows significant increase of CD68+ macro-phages in the alveoli, increase of IL-6 in immune and stromal cells, moderate elevation of FOXP3+ and IL-17+ cells and expression of CD4+ and CD8+ cells in alveolar walls. Immune cell interactions are discussed in the sense of ongoing cytokine storm. CONCLUSIONS Our findings highlight the complexity of COVID-19 lung affection, involving ATII cell hyperplasia, interstitial mononuclear cell infiltration and macrophages increase. The findings provide an additional knowledge on the pathophysiology of COVID-19 in the lung and can serve as a basis for investigation of molecular mechanisms of this disease.
Collapse
Affiliation(s)
- Maya Gulubova
- Department of Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria.
- Department of Anatomy, Histology, Embryology and Pathology, Medical Faculty, University "Prof. Dr. Assen Zlatarov", Burgas, Bulgaria.
- Clinics of Pathology, University Hospital "Prof. Dr. Stoyan Kirkovich" Stara, Zagora, Bulgaria.
- Medical Faculty, Trakia University,, Department of General and Clinical Pathology, Stara, Zagora, Bulgaria.
| | - Vesselina Merhar
- Department of Biology, Medical Genetics, Microbiology, Medical Faculty, University "Prof. Dr. Assen Zlatarov", Burgas, Bulgaria
| | - Dimitar Chonov
- Clinics of Pediatric Surgery, University Hospital "Prof. Dr. Stoyan Kirkovich" Stara, Zagora, Bulgaria
| | - Mitko Mitev
- Clinics of Rentgenology and Radiology, University Hospital, "Prof. Dr. Stoyan Kirkovich" Stara, Zagora, Bulgaria
| | - Lilia Pekova
- Clinics of Infectious Diseases, University Hospital "Prof. Dr. Stoyan Kirkovich", Stara Zagora, Bulgaria
| | - Julian Ananiev
- Department of Pathology, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
2
|
Peng W, Wei X, Wu Y, Shi C, Liu X, Wu J, Yang H, Rong N, Zhao B, Zhang G, Zhang W, Liu J, Liu J, Yang J. Dynamic Molecular Changes in Brain, Lung, and Heart of Hamsters Infected With SARS-CoV-2: Insights From a Severe and Recovery Phase Model. J Med Virol 2025; 97:e70410. [PMID: 40432336 DOI: 10.1002/jmv.70410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/13/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025]
Abstract
The Global pandemic of coronavirus disease 2019 was initiated by the emergence of severe acute respiratory syndrome coronavirus 2. In addition to conventional pulmonary lesions, a range of neurological injury symptoms have been identified in clinical practice, but the aetiology of neurological disorders linked to SARS-CoV-2 infection remains poorly understood. Syrian hamsters, which are highly susceptible to SARS-CoV-2 infection, exhibit a disease phenotype similar to that observed in human COVID-19 patients. In this study, a hamster model of COVID-19 infection was used to analyze molecular changes in different tissues at various time points post infection with distinct strains using proteomic and phosphoproteomic approaches. Multi-omics analysis showed that SARS-COV-2 infection triggers sustained downregulation of the abundance and phosphorylation levels of neuronal and synapse-associated proteins in the brain, suggesting that neuronal damage persists even during the recovery period. Additionally, infections with SARS-CoV-2 may contribute to the onset of long-term symptoms of COVID-19 by impacting energy metabolism, neurotransmitter release, and synaptic transmission pathways. This study provides a comprehensive molecular profile of hamsters infected with different SARS-CoV-2 strains in different tissues, offering foundational insights into the pathogenic mechanisms of COVID-19.
Collapse
Grants
- This study was supported by the National Key R&D Program of China (2023YFC2507102), the CAMS Innovation Fund for Medical Sciences (CIFMS) grant (2022-I2M-1-020, 2022-12M-CoV19-002, 2022-I2M-2-001, 2022-I2M-1-011, 2021-I2M-1-057, 2021-I2M-1-049, 2021-I2M-1-044, 2021-I2M-1-016, 2021-I2M-1-001 and 2022-I2M-CoV19-003), the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00008 and 22HHKYZX0034), State Key Laboratory Special Fund 2060204, and the National Natural Science Foundation of China (Grants 32070543 and 82341064).
Collapse
Affiliation(s)
- Wanjun Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Xiaohui Wei
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Chunmei Shi
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Xiaoyan Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Jing Wu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Hekai Yang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Na Rong
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Binbin Zhao
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Gengxin Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Wei Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Jiangfeng Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
| | - Jiangning Liu
- NHC Key Laboratory of Human Disease Comparative Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Laboratory Animal Science, Beijing, China
| | - Juntao Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Basic Medical Sciences, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Pathogen Biology, Beijing, China
| |
Collapse
|
3
|
Tao X, Wang Y, Jin J, Yan H, Yang H, Wan X, Li P, Xiao Y, Yu Q, Liu L, Liu Y, Han T, Zhang W. NSP6 regulates calcium overload-induced autophagic cell death and is regulated by KLHL22-mediated ubiquitination. J Adv Res 2025:S2090-1232(25)00350-9. [PMID: 40373961 DOI: 10.1016/j.jare.2025.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025] Open
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a substantial global threat. SARS-CoV-2 nonstructural proteins (NSPs) are essential for impeding the host replication mechanism while also assisting in the production and organization of new viral components. However, NSPs are not incorporated into viral particles, and their subsequent fate within host cells remains poorly understood. Additionally, their role in viral pathogenesis requires further investigation. OBJECTIVES This study aimed to discover the ultimate fate of NSP6 in host cells and to elucidate its role in viral pathogenesis. METHODS We investigated the effects of NSP6 on cell death and explored the underlying mechanism; moreover, we examined the degradation mechanism of NSP6 in human cells, along with analysing its correlation with coronavirus disease 2019 (COVID-19) severity in patient peripheral blood mononuclear cells (PBMCs). RESULTS NSP6 was demonstrated to induce cell death. Specifically, NSP6 interacted with EI24 autophagy-associated transmembrane protein (EI24) to increase intracellular Ca2+ levels, thereby enhancing the interactions between unc-51-like autophagy activating kinase 1 (ULK1) and RB1 inducible coiled-coil 1 (RB1CC1/FIP200), as well as beclin 1 (BECN1) and phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3). This cascade ultimately triggers autophagy, thus resulting in cell death. Additionally, we discovered that the homeostasis of the NSP6 protein was regulated by K48-linked ubiquitination. We identified kelch-like protein 22 (KLHL22) as the E3 ligase that was responsible for ubiquitinating and degrading NSP6, restoring intracellular calcium homeostasis and reversing NSP6-induced autophagic cell death. Moreover, NSP6 expression levels were observed to be positively associated with the severity of SARS-CoV-2-induced disease. CONCLUSION This study reveals that KLHL22-mediated ubiquitination controls NSP6 stability and that NSP6 induces autophagic cell death via calcium overload, highlighting its cytotoxic role and suggesting therapeutic strategies that target calcium signaling or promote NSP6 degradation as potential interventions against COVID-19.
Collapse
Affiliation(s)
- Xingyu Tao
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yanan Wang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Jiangbo Jin
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China
| | - Huilin Yan
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Hui Yang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Xiaorui Wan
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Ping Li
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yanghua Xiao
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Qi Yu
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Lingjiao Liu
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yang Liu
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China; Department of Clinical Microbiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China.
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China.
| | - Wei Zhang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China.
| |
Collapse
|
4
|
Hachimi A, El-Mansoury B, Merzouki M. Incidence, pathophysiology, risk factors, histopathology, and outcomes of COVID-19-induced acute kidney injury: A narrative review. Microb Pathog 2025; 202:107360. [PMID: 39894232 DOI: 10.1016/j.micpath.2025.107360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has led to a significant burden on global healthcare systems. COVID-19-induced acute kidney injury (AKI) is among one of the complications, that has emerged as a critical and frequent condition in COVID-19 patients. This AKI among COVID-19 patients is associated with poor outcomes, and high mortality rates, especially in those with severe AKI or requiring renal replacement therapy. COVID-19-induced AKI represents a significant complication with complex pathophysiology and multifactorial risk factors. Indeed, several pathophysiological mechanisms, including direct viral invasion of renal cells, systemic inflammation, endothelial and thrombotic abnormalities as well as nephrotoxic drugs and rhabdomyolysis are believed to underlie this condition. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include acute tubular necrosis, glomerular injury, and the presence of viral particles within renal tissue and urine. Identified risk factors for developing AKI vary among studies, depending on regions, underlying conditions, and the severity of the disease. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include show acute tubular necrosis, glomerular injury, and viral particles within renal tissue and urine. While, identified risk factors for developing AKI vary among studies, according to regions, underlying conditions, and the gravity of the disease. This narrative review aims to synthesize current knowledge on the incidence, pathophysiology, risk factors, histopathology, and outcomes of AKI induced by COVID-19.
Collapse
Affiliation(s)
- Abdelhamid Hachimi
- Medical ICU, Mohammed VI(th) University Hospital of Marrakech, Marrakech, Morocco; Morpho-Science Research Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco; Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Bilal El-Mansoury
- Nutritional Physiopathologies, Neuroscience and Toxicology Team, Laboratory of Anthropogenic, Biotechnology and Health, Faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco
| | - Mohamed Merzouki
- Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
5
|
Lloreta-Trull J, Marin-Corral J, Juanpere N, Pascual-Guardia S, Gimeno J, Naranjo D, Segalés L, Hernández S, Simón M, Serrano L, Casado B, Lloveras B, Gea J. Muscle disease in severe COVID-19 patients: a microangiopathic myopathy. Ultrastruct Pathol 2025; 49:296-305. [PMID: 40257175 DOI: 10.1080/01913123.2025.2488809] [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: 11/19/2024] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025]
Abstract
Patients surviving coronavirus disease of 2019 (COVID-19) often complain of skeletal muscle weakness that may be very limiting and long-lasting. There are almost no studies on the skeletal muscle of these patients, and electron microscopic data are scarce. We assessed the ultrastructural changes in the quadriceps of eight patients with COVID-19 and found a combination of features different from those reported in corticosteroid myopathy and acute relaxant-steroid myopathy. The most remarkable and constant changes involved the endothelial cells and consisted of massive amounts of pinocytotic vesicles, degenerative changes, platelet aggregates and, most characteristic of all, an increase in the external lamina thickness that seems to stem from reduplication due to successive bouts of endothelial cell damage and subsequent regeneration. Viral particles were not found in any of the cases. This distinct and quite common set of alterations defines the myopathy associated with infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This association seems to be the result of an inflammatory process that would arise in infected cells but could damage non-infected endomysial blood vessels, thus resulting in persistent changes of the microvasculature that would be related to long-standing myopathic clinical features.
Collapse
Affiliation(s)
- Josep Lloreta-Trull
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Judith Marin-Corral
- Intensive Care Department, Hospital del Mar., Critical Pathology Research Group (GREPAC), IMIM, Barcelona, Spain
| | - Nuria Juanpere
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Sergi Pascual-Guardia
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Department of Respiratory Medicine, Hospital del Mar-IMIM, Barcelona, Spain
| | - Javier Gimeno
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Dolores Naranjo
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Laura Segalés
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Silvia Hernández
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Mercedes Simón
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Laia Serrano
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Beatriz Casado
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Belén Lloveras
- Department of Pathology, Hospital del Mar-Parc de Salut Mar, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Joaquim Gea
- Department of Experimental and Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Department of Respiratory Medicine, Hospital del Mar-IMIM, Barcelona, Spain
| |
Collapse
|
6
|
Detrille A, Huvelle S, van Gils MJ, Geara T, Pascal Q, Snitselaar J, Bossevot L, Cavarelli M, Dereuddre-Bosquet N, Relouzat F, Contreras V, Chapon C, Caillé F, Sanders RW, Le Grand R, Naninck T. Whole-body visualization of SARS-CoV-2 biodistribution in vivo by immunoPET imaging in non-human primates. Nat Commun 2025; 16:2816. [PMID: 40118860 PMCID: PMC11928647 DOI: 10.1038/s41467-025-58173-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
The COVID-19 pandemic has caused at least 780 million cases globally. While available treatments and vaccines have reduced the mortality rate, spread and evolution of the virus are ongoing processes. Despite extensive research, the long-term impact of SARS-CoV-2 infection is still poorly understood and requires further investigation. Routine analysis provides limited access to the tissues of patients, necessitating alternative approaches to investigate viral dissemination in the organism. We address this issue by implementing a whole-body in vivo imaging strategy to longitudinally assess the biodistribution of SARS-CoV-2. We demonstrate in a COVID-19 non-human primate model that a single injection of radiolabeled [89Zr]COVA1-27-DFO human monoclonal antibody targeting a preserved epitope of the SARS-CoV-2 spike protein allows longitudinal tracking of the virus by positron emission tomography with computed tomography (PET/CT). Convalescent animals exhibit a persistent [89Zr]COVA1-27-DFO PET signal in the lungs, as well as in the brain, three months following infection. This imaging approach also allows viral detection in various organs, including the airways and kidneys, of exposed animals during the acute infection phase. Overall, the technology we developed offers a comprehensive assessment of SARS-CoV-2 distribution in vivo and provides a promising approach for the non-invasive study of long-COVID pathophysiology.
Collapse
Affiliation(s)
- Alexandra Detrille
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Steve Huvelle
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Orsay, France
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Tatiana Geara
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Quentin Pascal
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Jonne Snitselaar
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Catherine Chapon
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Fabien Caillé
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Orsay, France
| | - Rogier W Sanders
- Department of Medical Microbiology and Infection Prevention of the Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMRS1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Fontenay-aux-Roses, France.
| |
Collapse
|
7
|
Nankivell BJ, P'ng C, Tran T, Draper J, Ko D, Luu I, Basile K, Kable K, Sciberras F, Wong G, Kok J. The Effects of COVID-19 in Kidney Transplantation: Evidence From Tissue Pathology. Transplantation 2025; 109:352-361. [PMID: 39020461 DOI: 10.1097/tp.0000000000005121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
BACKGROUND The biological effects of SARS-CoV-2 infection in transplanted kidneys are uncertain with little pathological information. METHODS This single-center, prospective observational study evaluated kidney transplant biopsies from recipients of deceased donors with COVID-19, current recipients contracting SARS-CoV-2 Omicron variant in 2022, against prior BK virus (BKV) infection and uninfected (without SARS-CoV-2 or BKV) samples, as respective positive and negative comparators (n = 503 samples). RESULTS We demonstrated nonvirus tubular injury in implanted tissue from infected donors and prevalent recipients with mild acute COVID-19 and acute kidney injury, excluding direct viral infection as a cause of kidney damage. COVID particles were absent in 4116 ultrastructural images of 295 renal tubules from 4 patients with acute COVID-19. No viral cytopathic effect, viral allograft nephropathy, or SARS-CoV-2 RNA was detected in acute tissues, nor in 128 sequential samples from infected donors or recipients with COVID-19. Following recipient COVID-19 (mean 16.8 ± 12.0 wk post-infection), the biopsy-prevalence of rejection was 33.0% (n = 100 biopsies) versus 13.4% for contemporaneous uninfected controls (n = 337; P < 0.001). Prior COVID-19 was an independent risk factor for incident rejection using multivariable generalized estimating equation adjusted for competing risks (odds ratio, 2.195; 95% confidence interval, 1.189-4.052; P = 0.012). Landmark and matched-pair analyses confirmed an association of SARS-CoV-2 with subsequent transplant rejection, with a similar pattern following BKV infection. CONCLUSIONS Transplantation from COVID-19+ deceased donors yielded good recipient outcomes without evidence of viral tissue transmission. Acute kidney injury during COVID-19 was mediated by archetypical tubular injury and infection correlated with an increased risk of subsequent rejection.
Collapse
Affiliation(s)
- Brian J Nankivell
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Chow P'ng
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Thomas Tran
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Jenny Draper
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Danny Ko
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Ivan Luu
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kerri Basile
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kathy Kable
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | | | - Germaine Wong
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
8
|
Selvarajan S, John JS, Tharyan P, Kirubakaran R, Singh B, George B, Mathew JL, Rupali P. Therapeutic Versus Non-Therapeutic Dose Anticoagulation in COVID-19 Infection: A Systematic Review and Meta-analysis of Randomised Controlled Trials. EJHAEM 2025; 6:e1100. [PMID: 39935487 PMCID: PMC11811394 DOI: 10.1002/jha2.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 02/13/2025]
Abstract
Background Abnormal coagulation and thrombotic complications prompted many guidelines to recommend thromboprophylaxis for patients hospitalised with COVID-19, but the dose required for prophylaxis remains unclear. This systematic review (SR) analyses the safety and efficacy of therapeutic dose anticoagulation (TDA) versus non-therapeutic dose anticoagulation (NDA) in COVID-19 patients. Methods According to the Cochrane Handbook of Systematic Review of Interventions, we performed an SR. The protocol is registered in Prospero (CRD42021269197, date 12 August 2021). Results In this SR of 18 studies, TDA was shown to reduce all-cause mortality (risk ratio [RR] 0.83; 95% confidence interval [95% CI] 0.70, 0.99) in COVID-19 infection. TDA also reduced thrombosis (RR 0.55; 95% CI 0.48, 0.72) but increased major bleeding (RR 1.87; 95% CI 1.29, 2.69). A stratified analysis according to severity revealed that, in non-critical patients, TDA resulted in mortality benefit (RR 0.79; 95% CI 0.67, 0.94). In critical patients, TDA did not affect all-cause mortality (RR 1.03; 95% CI 0.89, 1.18) but reduced thrombosis (RR 0.65; 95% CI 0.48, 0.86) and increased major bleeding (RR 1.85; 95% CI 1.06, 3.23). Conclusion TDA significantly reduced all-cause mortality and thrombosis in non-critical COVID-19 patients at the expense of increased major bleeding. In critical COVID-19, this mortality benefit was not observed.
Collapse
Affiliation(s)
- Sushil Selvarajan
- Department of Clinical HaematologyChristian Medical CollegeVelloreIndia
| | - Jisha Sara John
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
| | - Prathap Tharyan
- Prof. BV Moses Centre for Evidence Informed HealthcareChristian Medical CollegeVelloreIndia
| | - Richard Kirubakaran
- Prof. BV Moses Centre for Evidence Informed HealthcareChristian Medical CollegeVelloreIndia
| | - Bhagteshwar Singh
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
- Department of Clinical Infection Microbiology and ImmunologyInstitute of Infection Veterinary & Ecological SciencesUniversity of LiverpoolLiverpoolUK
- Centre for Evidence Synthesis in Global Health, Department of Clinical SciencesLiverpool School of Tropical MedicineLiverpoolUK
| | - Biju George
- Department of Clinical HaematologyChristian Medical CollegeVelloreIndia
| | - Joseph L. Mathew
- Advanced Paediatrics CentrePostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - Priscilla Rupali
- Department of Infectious DiseasesChristian Medical CollegeVelloreIndia
| |
Collapse
|
9
|
Qi L, Deep A, Fox J, Yii M, Rahman M, Myint M, Myat H, Thet Z. A scoping review on adult patients with de novo glomerular diseases following COVID-19 infection or vaccine. Int Urol Nephrol 2025; 57:447-462. [PMID: 39225763 PMCID: PMC11772384 DOI: 10.1007/s11255-024-04189-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND There are increasing reports of glomerular disease (GD) following COVID-19 infection and vaccination. Current evidence on the possible link between COVID-19 infection or vaccination and GD is conflicting. OBJECTIVE The present study undertakes a scoping review of research to describe the relationship between COVID-19 infection and vaccination with GD and the common management strategies and overall outcomes of the disease to identify knowledge gaps and guide further research. ELIGIBILITY CRITERIA All original research studies published in English until 5th September 2022 were considered for inclusion in the review. Exclusion criteria were animal studies, autopsy studies, and data involving patients who were paediatric patients (< 16 years), were transplant recipients, had a recurrence of glomerular disease, had concomitant cancer or non-COVID-19 infection which may cause glomerular disease, or did not receive a renal biopsy. SOURCES OF EVIDENCE The five electronic databases searched were MEDLINE, PubMed, Scopus, EMBASE, and Cochrane. METHODS Two separate search strings related to COVID-19, and glomerular disease were combined using the Boolean operator 'AND'. Filters were used to limit publications to original research studies published in English. Search results from each database were imported into Covidence software ( www.covidence.org ) and used for de-duplication, article screening, and data extraction. Descriptive analyses were used to summarise demographics, diagnoses, and treatment outcomes. RESULTS After removing duplicates, 6853 titles and abstracts were screened. Of the 188 studies included, 106 studies described 341 patients with GD following COVID-19 infection and 82 described 146 patients with GD following a COVID-19 vaccination. IgA nephropathy was the most common GD pathology reported following COVID-19 vaccination with GD most common following mRNA vaccines. Collapsing focal segmental glomerulosclerosis was the most common GD following COVID-19 infection. Immunosuppressive treatment of GD was more common in the vaccine cohort than in the infection cohort. CONCLUSION Despite the significant number of COVID-19 infections and vaccinations around the world, our understanding of GD associated with COVID-19 infection and vaccination remains poor, and more research is needed to understand the possible relationship better.
Collapse
Affiliation(s)
- Liam Qi
- Department of Medicine, Prince Charles Hospital, Metro North Hospital and Health Service, Chermside, QLD, Australia
- The University of Queensland, Faculty of Medicine, Brisbane, QLD, Australia
| | - Aman Deep
- Department of Nephrology, Central Queensland Hospital and Health Service, Rockhampton, QLD, Australia
| | - Jordan Fox
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia
| | - Mark Yii
- Department of Medicine, Flinders Medical Centre, South Adelaide Local Health Network, Bedford Park, SA, Australia
| | - Muhammad Rahman
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia
| | - Mar Myint
- K & K Kidney Health, Rockhampton, QLD, Australia
| | - Htoo Myat
- Department of Medicine, Canberra Hospital, Canberra Hospital and Health Service, Garran, ACT, Australia
| | - Zaw Thet
- Department of Nephrology, Central Queensland Hospital and Health Service, Rockhampton, QLD, Australia.
- K & K Kidney Health, Rockhampton, QLD, Australia.
- The University of Queensland Rural Clinical School, Rockhampton, QLD, Australia.
- School of Medicine and Dentistry, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.
| |
Collapse
|
10
|
Anzalone N, Gerevini S, del Poggio A, Gaudino S, Causin F, Politi LS, Triulzi FM, Pero G, Pichiecchio A, Bastianello S, Baruzzi FM, Bianchini E, Foti G, Ricciardi GK, Sponza M, Menozzi R, Cosottini M, Chirico PD, Saba L, Gasparotti R. Neuroradiological manifestations in hospitalized patients with COVID-19: An Italian national multicenter study on behalf of AINR (Associazione Italiana di Neuroradiologia) and SIRM (Società Italiana di Radiologia Medica). Neuroradiol J 2025; 38:44-51. [PMID: 38897216 PMCID: PMC11562890 DOI: 10.1177/19714009241240312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
PURPOSE This multicentric study aims to characterize and assess the occurrence of neuroradiological findings among patients with SARS-CoV-2 infection during the first Italian wave of the pandemic outbreak. MATERIALS AND METHODS Patients' data were collected between May 2020 and June 2020. Clinical and laboratory data, chest imaging, brain CT, and MRI imaging were included. Acquired data were centralized and analyzed in two hospitals: ASST Spedali Civili, Brescia, and IRRCS San Raffaele Research Hospital, Milan, Italy. COVID-19 patients were classified into two different subgroups, vascular and nonvascular. The vascular pattern was further divided into ischemic and hemorrhagic stroke groups. RESULTS Four hundred and fifteen patients from 20 different Italian Centers were enrolled in the study. The most frequent symptom was focal neurological deficit, found in 143 patients (34.5%). The most frequent neuroradiological finding was ischemic stroke in 122 (29.4%) patients. Forty-four (10.6%) patients presented a cerebral hemorrhage. Forty-seven patients had non-stroke neuroimaging lesions (11.3%). The most common was PRES-like syndrome (28%), SWI hypointensities (22%), and encephalitis (19%). The stroke group had higher CAD risk (37.5% vs 20%, p = .016) and higher D-dimer levels (1875 ng/mL vs 451 ng/mL, p < .001) compared to the negative group. CONCLUSION Our study describes the biggest cohort study in Italy on brain imaging of COVID-19 patients and confirms that COVID-19 patients are at risk of strokes, possibly due to a pro-thrombotic microenvironment. Moreover, apart from stroke, the other neuroradiological patterns described align with the ones reported worldwide.
Collapse
Affiliation(s)
- Nicoletta Anzalone
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
- Vita-Salute San Raffaele University, Italy
| | | | - Anna del Poggio
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
| | - Simona Gaudino
- Radiology Department, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Italy
| | | | | | - Fabio Maria Triulzi
- Neuroradiology Department, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Italy
| | - Guglielmo Pero
- Neuroradiology Department, ASST Grande Ospedale Metropolitano Niguarda, Italy
| | | | | | | | - Elena Bianchini
- Neuroradiology Unit, Radiology Department, Ospedale Legnano, Italy
| | - Giovanni Foti
- Radiology Department, Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | | | - Massimo Sponza
- Neuroradiology Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Roberto Menozzi
- Neuroradiology Department, Azienda Ospedaliera Universitaria, Parma, Italy
| | - Mirco Cosottini
- Neuroradiology Department, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | - Luca Saba
- Radiology Department, Azienda Ospedaliero Universitaria, Cagliari, Italy
| | | |
Collapse
|
11
|
Yilmaz E, Yilmaz D, Yildiz CG, Cacan E. Upregulation of the MAP2K4 gene triggers endothelial-mesenchymal transition in COVID-19. Mol Biol Rep 2025; 52:180. [PMID: 39888478 DOI: 10.1007/s11033-025-10289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND SARS-CoV-2 infection is marked by an excessive inflammatory response, leading to elevated production of pro-inflammatory cytokines through activation of intracellular pathways like mitogen-activated protein kinase (MAPK). Viruses can use the MAPK signaling pathway to their advantage, but the relationship of this pathway to the severe SARS-CoV-2 period has not been fully elucidated. MAP2K4 is involved in the MAPK signaling pathway and affects cellular processes such as cell-cell junction, cell proliferation, differentiation and apoptosis. METHODS AND RESULTS In this study, we sought to determine the associated biomarkers that are involved in the MAP2K4 pathway and elucidate its possible roles in terms of some clinical features associated with COVID-19. We evaluated the expressions of MAP2K4, SNAI1, SLUG, ZEB1 and E-Cadherin. For this purpose, we prospectively recruited 66 individuals, 39 of whom were women and had a mean age of 65 years. The results revealed that MAP2K4 upregulation increased SNAI1 gene expression level whereas E- Cadherin level was decreased in SARS-CoV-2 positive participants. In addition, negative correlations were determined with PLT, Lymphocyte and CKMB and E- Cadherin levels in positive participants. We also observed a negative correlation between the MAP2K4 and AST, and a positive correlation between SLUG and BUN, ZEB1 and CK. CONCLUSIONS We conclude that SARS-CoV-2 infection triggers fibrosis by increasing MAP2K4 regulation. Additionally, this is the first study to demonstrate the possible contribution of MAP2K4 in influencing COVID-19 clinical features, which may be relevant for identifying COVID-19 positive participants with severe complications.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, Tokat, 66100, Türkiye
| | - Can Gokay Yildiz
- Department of Emergency Medicine, Tokat City Hospital, Tokat, 60200, Türkiye
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye.
| |
Collapse
|
12
|
Vanderheiden A, Diamond MS. Animal Models of Non-Respiratory, Post-Acute Sequelae of COVID-19. Viruses 2025; 17:98. [PMID: 39861887 PMCID: PMC11768974 DOI: 10.3390/v17010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Post-acute sequelae of COVID-19 (PASC) are a diverse set of symptoms and syndromes driven by dysfunction of multiple organ systems that can persist for years and negatively impact the quality of life for millions of individuals. We currently lack specific therapeutics for patients with PASC, due in part to an incomplete understanding of its pathogenesis, especially for non-pulmonary sequelae. Here, we discuss three animal models that have been utilized to investigate PASC: non-human primates (NHPs), hamsters, and mice. We focus on neurological, gastrointestinal, and cardiovascular PASC and highlight advances in mechanistic insight that have been made using these animal models, as well as discussing the sequelae that warrant continued and intensive research.
Collapse
Affiliation(s)
- Abigail Vanderheiden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
13
|
Gultom M, Lin L, Brandt CB, Milusev A, Despont A, Shaw J, Döring Y, Luo Y, Rieben R. Sustained Vascular Inflammatory Effects of SARS-CoV-2 Spike Protein on Human Endothelial Cells. Inflammation 2024:10.1007/s10753-024-02208-x. [PMID: 39739157 DOI: 10.1007/s10753-024-02208-x] [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: 08/30/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been associated with systemic inflammation and vascular injury, which contribute to the development of acute respiratory syndrome (ARDS) and the mortality of COVID-19 infection. Moreover, multiorgan complications due to persistent endothelial dysfunction have been suspected as the cause of post-acute sequelae of SARS-CoV-2 infection. Therefore, elucidation of the vascular inflammatory effect of SARS-CoV-2 will increase our understanding of how endothelial cells (ECs) contribute to the short- and long-term consequences of SARS-CoV-2 infection. Here, we investigated the interaction of SARS-CoV-2 spike protein with human ECs from aortic (HAoEC) and pulmonary microvascular (HPMC) origins, cultured under physiological flow conditions. We showed that the SARS-CoV-2 spike protein triggers prolonged expression of cell adhesion markers in both ECs, similar to the effect of TNF-α. SARS-CoV-2 spike treatment also led to the release of various cytokines and chemokines observed in severe COVID-19 patients. Moreover, increased binding of leucocytes to the endothelial surface and a procoagulant state of the endothelium were observed. Transcriptomic profiles of SARS-CoV-2 spike-activated HPMC and HAoEC showed prolonged upregulation of genes and pathways associated with responses to virus, cytokine-mediated signaling, pattern recognition, as well as complement and coagulation pathways. Our findings support experimental and clinical observations of the vascular consequences of SARS-CoV-2 infection and highlight the importance of EC protection as one of the strategies to mitigate the severe effects as well as the possible post-acute complications of COVID-19 disease.
Collapse
Affiliation(s)
- Mitra Gultom
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Camilla Blunk Brandt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anastasia Milusev
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Alain Despont
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Jane Shaw
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Department for Biomedical Research, University of Bern, Bern, Switzerland
- Department of Angiology, Inselspital, Bern University Hospital, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum Für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Robert Rieben
- Department for Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
14
|
Jensen TO, Harper K, Gupta S, Liu ST, Dharan NJ, Baker JV, Pett SL, Shaw-Saliba K, Esmail A, Ho MQ, Almasri E, Dewar RL, Lundgren J, Vock DM. Impact of Baseline SARS-CoV-2 Load in Plasma and Upper Airways on the Incidence of Acute Extrapulmonary Complications of COVID-19: A Multicentric, Prospective, Cohort Study. Clin Infect Dis 2024; 79:1394-1403. [PMID: 39271151 PMCID: PMC11650867 DOI: 10.1093/cid/ciae469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Extrapulmonary complications (EPCs) are common in patients hospitalized for coronavirus disease 2019 (COVID-19), but data on their clinical consequences and association with viral replication and systemic viral dissemination are lacking. METHODS Patients hospitalized for COVID-19 and enrolled in the Therapeutics for Inpatients with COVID-19 (TICO) platform trial at 114 international sites between August 2020 and November 2021 were included in a prospective cohort study. We categorized EPCs into 39 event types within 9 categories and estimated their frequency through day 28 and their association with clinical outcomes through day 90. We analyzed the association between baseline viral burden (plasma nucleocapsid antigen [N-Ag] level and upper airway viral load) and EPCs, adjusting for other baseline factors. RESULTS A total of 2625 trial participants were included in the study. Their median age was 57 years (interquartile range, 46-68 years), 57.7% were male, and 537 (20.5%) had ≥1 EPC. EPCs were associated with higher day-90 all-cause mortality rate (hazard ratio, 9.6 [95% confidence interval, 7.3-12.7]) after adjustment for other risk factors. The risk of EPCs increased with increasing baseline plasma N-Ag level (hazard ratio, 1.21 per log10 ng/L increase [95% confidence interval, 1.09-1.34]), and upper airway viral load (1.12 per log10 copies/mL increase [1.04-1.19), after adjustment for comorbid conditions, disease severity, inflammatory markers, and other baseline factors. Trial treatment allocation had no effect on EPC risk. CONCLUSIONS Systemic viral dissemination as evidenced by high plasma N-Ag level and high respiratory viral burden are associated with development of EPCs in COVID-19, which in turn are associated with higher 90-day mortality rates.
Collapse
Affiliation(s)
- Tomas O Jensen
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Katrina Harper
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shaili Gupta
- Department of Medicine, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sean T Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nila J Dharan
- Kirby Institute, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Jason V Baker
- Division of Infectious Diseases and International Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Infectious Diseases, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Sarah L Pett
- The Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom
| | - Kathryn Shaw-Saliba
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Aliasgar Esmail
- Division of Pulmonology, Department of Medicine, Centre for Lung Infection and Immunity, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Minh Q Ho
- Department of Infectious Diseases, Orlando VA Medical Center, Orlando, Florida, USA
| | - Eyad Almasri
- Department of Pulmonology, University of California San Francisco, Fresno, California, USA
| | - Robin L Dewar
- Virus Isolation and Serology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jens Lundgren
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - David M Vock
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Mata-Castro N, Castañeda-Vozmediano R, Perna C, Prada Puentes C, Sanz López L. Histological Findings of Resected Tracheal Ring in SARS-CoV-2-Positive and -Negative Tracheostomized Patients. Life (Basel) 2024; 14:1655. [PMID: 39768362 PMCID: PMC11679342 DOI: 10.3390/life14121655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION The aim of this study was to compare the histopathological findings in the resected tracheal ring of tracheotomized critically ill patients with or without severe SARS-CoV-2 infection. MATERIAL AND METHODS This is a prospective case-control study. The data collection period was between May 2020 and 2022. Eighty tracheostomies were performed on patients with long intubation, and the resected tracheal ring was examined by standard microscopy. Forty consecutive tracheotomies were carried out in COVID-19-positive and -negative patients. RESULTS The mean age was 67.1 (6.9 SD) years in the COVID-19 group and 67.8 (9.6 SD) in the control group (p = 0.3). The number of men in each group was 30 (75.0%) versus 27 (67.5%), respectively (p = 0.5). No relevant histological alterations were found in 82.5% of samples. Chronic subepithelial inflammation was found in 13.8% of cases. Two cases presented with vasculitis (2.5%), and one case presented with thrombotic microangiopathy (1.2%), all of them in the COVID-19 group. We found no statistically significant dependence between relevant histologic findings versus no alterations (X2 = 0.779, p= 0.377) and no significant risk indices (RR = 1.8, OR = 2.032, PAR = 44%). CONCLUSION There is no evidence of increased risk of histopathological findings in the resected tracheal ring of patients with long intubation and COVID-19 disease.
Collapse
Affiliation(s)
- Nieves Mata-Castro
- Department of ENT, 12 de Octubre University Hospital, 28041 Madrid, Spain;
| | | | - Cristian Perna
- Department of Pathology, Ramon y Cajal Hospital, 28034 Madrid, Spain;
- RYCIS, School of Medicine Universidad de Alcala, 28801 Madrid, Spain
| | - Carlos Prada Puentes
- Department of Pathological Anatomy, Torrejón University Hospital, 28850 Madrid, Spain;
| | - Lorena Sanz López
- School of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain;
- Department of ENT, Torrejón University Hospital, Universidad Francisco de Vitoria, 28850 Madrid, Spain
| |
Collapse
|
16
|
Bulterys PL, Xu G, Pinsky BA, Troxell ML, Menke JR, Berry GJ, Fernandez-Pol S, Hazard FK. The Histopathologic Features of Early COVID Pneumonia in a Pediatric Patient: New Insight into the Role of Macrophages. Int J Surg Pathol 2024; 32:1595-1601. [PMID: 39435671 DOI: 10.1177/10668969241236704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
A life-threatening complication of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is acute respiratory distress syndrome. Our understanding of the pathologic changes in coronavirus disease 2019 (COVID-19) is based almost exclusively on post-mortem analyses of adults. These studies established several hallmarks of SARS-CoV-2 lung infection, including diffuse alveolar damage, microvascular thrombi, and acute bronchopneumonia. We describe a fatal example of COVID pneumonia in a 9-year-old girl who presented with fever 10 months following the diagnosis of ALK-positive anaplastic large cell lymphoma (ALCL). A chest computed tomography scan revealed left upper lobe lung consolidation and nodular airspace disease, and an initial SARS-CoV-2 nasopharyngeal swab (RT-PCR) was negative. A subsequent lung biopsy performed due to concern for relapsed ALCL demonstrated sheets of intra-alveolar and interstitial macrophages, and macrophage-rich fibrinous exudates. Immunohistochemical and in-situ hybridization stains confirmed these macrophages as the predominant SARS-CoV-2-infected cell type. Subsequent RT-PCR testing of upper and lower respiratory tract samples was positive for SARS-CoV-2 infection. Whole genome sequencing confirmed the presence of the B.1.617.2 (Delta) variant. This biopsy illustrates the histopathologic features of early COVID pneumonia in antemortem lung tissue from a pediatric patient, and establishes macrophages as a potential source of SARS-CoV-2 amplification.
Collapse
Affiliation(s)
- Philip L Bulterys
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Guangwu Xu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin A Pinsky
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Megan L Troxell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua R Menke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gerald J Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Florette K Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
17
|
Yilmaz E, Yilmaz D, Cacan E. Severe and post-COVID-19 are associated with high expression of vimentin and reduced expression of N-cadherin. Sci Rep 2024; 14:29256. [PMID: 39587116 PMCID: PMC11589739 DOI: 10.1038/s41598-024-72192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/04/2024] [Indexed: 11/27/2024] Open
Abstract
SARS-CoV-2 penetrates human cells via its spike protein, which mainly interacts with ACE2 receptors, triggering viral replication and an exacerbated immune response characterized by a cytokine storm. Vimentin III, an intermediate filament protein predominantly found in mesenchymal cells, has garnered considerable attention in recent research due to its multifaceted biological roles and significance in the endothelial-mesenchymal transition (EndMT) during various fibrotic processes. However, the pathophysiological mechanisms linking vimentin to SARS-CoV-2 remain incompletely elucidated. In this study, we determined the expression profiles of vimentin in three cohorts: patients admitted to the intensive care unit with SARS-CoV-2 infection, individuals in the 6-12 month convalescent phase post-infection and COVID-19 negative controls. Our objective was to assess the association between peripheral blood biomarkers implicated in endothelial dysfunction and genes related to fibrosis. Serum levels of vimentin and N-cadherin were determined by ELISA, while vimentin gene expression was determined by qRT-PCR. In addition, we examined the correlation between clinical parameters and serum levels of vimentin and N-cadherin in severe COVID-19 patients and healthy counterparts. Our findings revealed elevated serum vimentin levels and increased gene expression in severe COVID-19 patients compared to healthy controls. Conversely, serum N-cadherin levels were diminished in both acute and convalescent stages of severe COVID-19 relative to healthy individuals. Notably, associations were observed between C-reactive protein, lactate dehydrogenase, lymphocyte count and vimentin levels in severe COVID-19 patients, indicative of endothelial dysfunction. Furthermore, our study identified vimentin and N-cadherin as potential diagnostic markers via ROC analysis. Overall, delineating the dysregulation of vimentin and N-cadherin due to SARS-CoV-2 infection in disease pathogenesis and tissue homeostasis offers novel insights for clinical management and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Tokat Gaziosmanpasa University, 60200, Tokat, Turkey
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, 66100, Yozgat, Turkey
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Tokat Gaziosmanpasa University, 60200, Tokat, Turkey.
| |
Collapse
|
18
|
Khanal R, Heinen N, Bogomolova A, Meister TL, Herrmann ST, Westhoven S, Nocke MK, Todt D, Jockenhövel F, Klein IM, Hartmann L, Vondran FWR, Steinmann E, Zimmer G, Ott M, Brown RJP, Sharma AD, Pfaender S. MicroRNAs modulate SARS-CoV-2 infection of primary human hepatocytes by regulating the entry factors ACE2 and TMPRSS2. Liver Int 2024; 44:2983-2995. [PMID: 39175256 DOI: 10.1111/liv.16079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIMS Severe acute respiratory syndrome coronavirus (SARS-CoV-2) preferentially infects the respiratory tract; however, several studies have implicated a multi-organ involvement. Hepatic dysfunctions caused by SARS-CoV-2 infection have been increasingly recognized and described to correlate with disease severity. To elucidate molecular factors that could contribute towards hepatic infection, we concentrated on microRNAs (miRNAs), a class of small non-coding RNAs that modulate various cellular processes and which are reported to be differentially regulated during liver injury. We aimed to study the infection of primary human hepatocytes (PHH) with SARS-CoV-2 and to evaluate the potential of miRNAs for modulating viral infection. METHODS We analysed liver autopsies from a coronavirus disease 19 (COVID-19)-positive cohort for the presence of viral RNA using Nanopore sequencing. PHH were used for the infection with SARS-CoV-2. The candidate miRNAs targeting angiotensin converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) were identified using in silico approaches. To discover the potential regulatory mechanism, transfection experiments, qRT-PCRs, western blots and luciferase reporter assays were performed. RESULTS We could detect SARS-CoV-2 RNA in COVID-19-positive liver autopsies. We show that PHH express ACE2 and TMPRSS2 and can be readily infected with SARS-CoV-2, resulting in robust replication. Transfection of selected miRNA mimics reduced SARS-CoV-2 receptor expression and SARS-CoV-2 burden in PHH. In silico and biochemical analyses supported a potential direct binding of miR-141-3p to the SARS-CoV-2 genome. CONCLUSION We confirm that PHH are susceptible to SARS-CoV-2 infection and demonstrate selected miRNAs targeting SARS-CoV-2 entry factors and/or the viral genome reduce viral loads. These data provide novel insights into hepatic susceptibility to SARS-CoV-2 and associated dysfunctions in COVID-19.
Collapse
Affiliation(s)
- Rajendra Khanal
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Natalie Heinen
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Alexandra Bogomolova
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Toni L Meister
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Institute for Infection Research and Vaccine Development (IIRVD), Centre for Internal Medicine, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Centre for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Simon T Herrmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Saskia Westhoven
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Maximilian K Nocke
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Daniel Todt
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- European Virus Bioinformatics Center (EVBC), Jena, Germany
| | - Freya Jockenhövel
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Isabel M Klein
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Laura Hartmann
- Tissue Bank of the German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian W R Vondran
- Department of General, Visceral, Pediatric and Transplant Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Eike Steinmann
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Gert Zimmer
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard J P Brown
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Research Group Liver Regeneration & RNA Therapeutics, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Stephanie Pfaender
- Department for Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
- Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
- University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Menna C, Fiorelli S, Marinucci BT, Massullo D, D'Andrilli A, Ciccone AM, Andreetti C, Maurizi G, Vanni C, Siciliani A, Tiracorrendo M, Mancini M, Venuta F, Rendina EA, Ibrahim M. New perspectives on tracheal resection for COVID-19-related stenosis: A propensity score matching analysis. J Thorac Cardiovasc Surg 2024; 168:1385-1393. [PMID: 38555996 DOI: 10.1016/j.jtcvs.2024.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE The large number of patients with COVID-19 subjected to prolonged invasive mechanical ventilation has been expected to result in a significant increase in tracheal stenosis in the next years. The aim of this study was to evaluate and compare postoperative outcomes of patients who survived COVID-19 critical illness and underwent tracheal resection for postintubation/posttracheostomy tracheal stenosis with those of non-COVID-19 patients. METHODS It was single-center, retrospective study. All consecutive patients with post-intubation/posttracheostomy tracheal stenosis who underwent tracheal resection from February 2020 to March 2022 were enrolled. A total of 147 tracheal resections were performed: 24 were in post-COVID-19 patients and 123 were in non-COVID-19 patients. A 1:1 propensity score matching analysis was performed, considering age, gender, body mass index, and length of stenosis. After matching, 2 groups of 24 patients each were identified: a post-COVID-19 group and a non-COVID group. RESULTS No mortality after surgery was registered. Posttracheostomy etiology of stenosis resulted more frequently in post-COVID-19 patients (n = 20 in the post-COVID-19 group vs n = 11 in the non-COVID-19 group; P = .03), as well as intensive care unit admissions during the postoperative period (16 vs 9 patients; P = .04). Need for postoperative reintubation for glottic edema and respiratory failure was higher in the post-COVID-19 group (7 vs 2 postoperative reintubation procedures; P = .04). Postoperative dysphonia was observed in 11 (46%) patients in the post-COVID-19 group versus 4 (16%) patients in the non-COVID-19 group (P = .03). CONCLUSIONS Tracheal resection continues to be safe and effective in COVID-19-related tracheal stenosis scenarios. Intensive care unit admission rates and postoperative complications seem to be higher in post-COVID-19 patients who underwent tracheal resection compared with non-COVID-19 patients.
Collapse
Affiliation(s)
- Cecilia Menna
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.
| | - Silvia Fiorelli
- Division of Anesthesiology and Intensive Care, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | | | - Domenico Massullo
- Division of Anesthesiology and Intensive Care, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio D'Andrilli
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Anna Maria Ciccone
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Claudio Andreetti
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Giulio Maurizi
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Camilla Vanni
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Alessandra Siciliani
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Matteo Tiracorrendo
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Massimiliano Mancini
- Morphologic and Molecular Pathology Unit, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Federico Venuta
- Division of Thoracic Surgery, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Erino Angelo Rendina
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Mohsen Ibrahim
- Division of Thoracic Surgery, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
20
|
Yousef A, Solomon I, Cheng G, Makani S, Boys J, Weissbrod PA. COVID-19 Related Tracheal Stenosis Requiring Tracheal Resection: A Case Series. Ann Otol Rhinol Laryngol 2024; 133:879-885. [PMID: 39127881 PMCID: PMC12064082 DOI: 10.1177/00034894241266489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To characterize the preoperative and intraoperative findings of symptomatic tracheal stenosis associated with COVID-19 related respiratory failure requiring tracheal resection. METHOD We performed a retrospective review identifying all patients with a history of tracheal stenosis secondary to COVID-19 related respiratory failure who subsequently received a tracheal resection at our institution between January 2020 and June 2023. Clinical, radiological, pathological, and surgical characteristics were recorded to describe and characterize pre-operative and intraoperative findings associated with tracheal stenosis in the setting of a previous COVID-19 infection. RESULTS We retrospectively reviewed 11 patients with COVID-19 related tracheal stenosis that required open tracheal or cricotracheal resection. The mean age was 54.1. Patients were hospitalized for a mean of 49.5 days related to COVID-19 complications. Tracheotomy was completed in 10 patients (90.9%) during their initial hospitalization with COVID-19 related respiratory failure. Patients were intubated a mean of 18.6 days prior to tracheotomy completion. Ten patients (90.9%) underwent endoscopic operative interventions for their tracheal stenosis prior to open resection. Intraoperatively, the mean stenosis length was 3.33 cm. The mean tracheal resection length was 3.96 cm. Patients were hospitalized for a mean of 8.27 days post operatively with no significant post operative complications. CONCLUSIONS Symptomatic tracheal stenosis in the setting of prolonged intubation due to COVID-19 is an under-described etiology. This is one of the largest single institution retrospective reviews that identifies 11 patients with prolonged intubation who developed symptomatic tracheal stenosis refractory to conservative management and ultimately requiring tracheal resection.
Collapse
Affiliation(s)
- Andrew Yousef
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| | - Isaac Solomon
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| | - George Cheng
- Division of Pulmonology, Critical Care, Sleep Medicine & Physiology, University of California, San Diego
| | - Samir Makani
- Department of Pulmonology and Critical Care Medicine, Scripps Hospital Encinitas, Encinitas, California
| | - Joshua Boys
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego
| | - Philip A. Weissbrod
- Department of Otolaryngology-Head and Neck Surgery, University of California, San Diego, California, USA
| |
Collapse
|
21
|
Keiser PT, Zhang W, Ricca M, Wacquiez A, Grimins A, Cencic R, Patten JJ, Shah P, Padilha E, Connor JH, Pelletier J, Lyons SM, Saeed M, Brown LE, Porco JA, Davey RA. Amidino-rocaglates (ADRs), a class of synthetic rocaglates, are potent inhibitors of SARS-CoV-2 replication through inhibition of viral protein synthesis. Antiviral Res 2024; 230:105976. [PMID: 39117283 PMCID: PMC11434215 DOI: 10.1016/j.antiviral.2024.105976] [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: 04/09/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Coronaviruses are highly transmissible respiratory viruses that cause symptoms ranging from mild congestion to severe respiratory distress. The recent outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the need for new antivirals with broad-acting mechanisms to combat increasing emergence of new variants. Currently, there are only a few antivirals approved for treatment of SARS-CoV-2. Previously, the rocaglate natural product silvestrol and synthetic rocaglates such as CR-1-31b were shown to have antiviral effects by inhibiting eukaryotic translation initiation factor 4A1 (eIF4A) function and virus protein synthesis. In this study, we evaluated amidino-rocaglates (ADRs), a class of synthetic rocaglates with the most potent eIF4A-inhibitory activity to-date, for inhibition of SARS-CoV-2 infection. This class of compounds showed low nanomolar potency against multiple SARS-CoV-2 variants and in multiple cell types, including human lung-derived cells, with strong inhibition of virus over host protein synthesis and low cytotoxicity. The most potent ADRs were also shown to be active against two highly pathogenic and distantly related coronaviruses, SARS-CoV and MERS-CoV. Mechanistically, cells with mutations of eIF4A1, which are known to reduce rocaglate interaction displayed reduced ADR-associated loss of cellular function, consistent with targeting of protein synthesis. Overall, ADRs and derivatives may offer new potential treatments for SARS-CoV-2 with the goal of developing a broad-acting anti-coronavirus agent.
Collapse
Affiliation(s)
- Patrick T Keiser
- Department of Virology, Immunology, and Microbiology, Boston University Medical School, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA
| | - Wenhan Zhang
- Boston University Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Michael Ricca
- Boston University Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Alan Wacquiez
- National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA; Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Autumn Grimins
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Regina Cencic
- Department of Biochemistry, Department of Oncology and Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada, H3G 1Y6
| | - J J Patten
- Department of Virology, Immunology, and Microbiology, Boston University Medical School, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA
| | - Pranav Shah
- National Institutes of Health, National Center for Advancing Translational Sciences, Bethesda, MD, 20892, USA
| | - Elias Padilha
- National Institutes of Health, National Center for Advancing Translational Sciences, Bethesda, MD, 20892, USA
| | - John H Connor
- Department of Virology, Immunology, and Microbiology, Boston University Medical School, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA
| | - Jerry Pelletier
- Department of Biochemistry, Department of Oncology and Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada, H3G 1Y6
| | - Shawn M Lyons
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Mohsan Saeed
- National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA; Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA, 02118, USA
| | - Lauren E Brown
- Boston University Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - John A Porco
- Boston University Center for Molecular Discovery (BU-CMD), Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Robert A Davey
- Department of Virology, Immunology, and Microbiology, Boston University Medical School, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, MA, 02118, USA.
| |
Collapse
|
22
|
Pedreañez A, Mosquera-Sulbaran JA, Tene D. Role of the receptor for advanced glycation end products in the severity of SARS-CoV-2 infection in diabetic patients. Diabetol Int 2024; 15:732-744. [PMID: 39469543 PMCID: PMC11512988 DOI: 10.1007/s13340-024-00746-1] [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: 03/19/2024] [Accepted: 07/17/2024] [Indexed: 10/30/2024]
Abstract
Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, is a severe disease in older adults and in individuals with associated comorbidities such as diabetes mellitus. Patients with diabetes infected with SARS-CoV-2 are more likely to develop severe pneumonia, hospitalization, and mortality compared with infected non-diabetic patients. During diabetes, hyperglycemia contributes to the maintenance of a low-grade inflammatory state which has been implicated in the microvascular and macrovascular complications associated with this pathology. The receptor for advanced glycation end products (RAGE) is a multi-ligand pattern recognition receptor, expressed on a wide variety of cells, which participates as an important mediator of inflammatory responses in many diseases, including lung diseases. This review highlights the role of RAGE in the pathophysiology of COVID-19 with special emphasis on diabetic patients. These data could explain the severity of the disease, positioning it as a key therapeutic target in the clinical management of this infection.
Collapse
Affiliation(s)
- Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Apartado Postal: 23, Maracaibo 4001-A, Maracaibo, Zulia Venezuela
| | - Jesús A. Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas “Dr. Américo Negrette”, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Diego Tene
- Universidad Nacional del Chimborazo, Facultad de Ciencias de la Salud, Riobamba, Ecuador
| |
Collapse
|
23
|
Zhang X, Wen R, Chen H, Liu J, Wu Y, Xu M, Wang R, Zeng X. COVID-19 and diabetes research: Where are we now and what does the future hold? A bibliometric visualization analysis. Heliyon 2024; 10:e37615. [PMID: 39315181 PMCID: PMC11417241 DOI: 10.1016/j.heliyon.2024.e37615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Background & objective The extensive spread of Coronavirus disease 2019 (COVID-19) worldwide has caused a dramatic negative impact on many individuals' health. This study aims to systematically and comprehensively analyze the current status and possible future directions of diabetes mellitus (DM) and COVID-19 research. Methods We obtained publications about COVID-19 and DM from the Web of Science Core Collection (WoSCC) using the search terms "COVID-19″ and similar terms combined with "DM" and similar terms, with a date range of January 2020 to May 2024. And we used CiteSpace V 6.3.R2 to perform the bibliometric visualization analysis. Results The search enrolled 6266 publications. The USA is a country with the most publications; Harvard University was the most productive institution in this field. The highest-ranked journal was the PLOS ONE, and the most cited journal was Lancet. The 20 most cited journals have all been cited 28754 times, accounting for 28 % of the total cites; the range of those journals was 790-3197. Publications on COVID-19 and DM research exhibited a distinct trajectory, shifting from an initial emphasis on understanding the impact of diabetes on COVID-19 infection and its associated pathophysiological mechanisms to a focus on analyzing the differential responses of diverse patient populations. Subsequently, research has progressed to examine the effects of medications and vaccines, as well as the long-term consequences of COVID-19 in diabetic individuals. Throughout this research endeavor, the exploration of diverse therapeutic interventions, their efficacy, and ultimate outcomes have consistently remained a paramount focus. And " metabolic syndrome," " long COVID," and " gestational diabetes" are still likely to be the hotspots and frontiers of research in the future. Conclusions This bibliometric analysis related to DM in COVID-19 illuminates the current research situation and developmental trends, supporting researchers in the exploration of prospective directions for research.
Collapse
Affiliation(s)
- Xunlan Zhang
- Zunyi Medical University, No.6 Xuefu West Road, Xinpu District, 563000, Zunyi City, China
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Ru Wen
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Hengzhi Chen
- Zunyi Medical University, No.6 Xuefu West Road, Xinpu District, 563000, Zunyi City, China
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Jian Liu
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Yu Wu
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Min Xu
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Rongpin Wang
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| | - Xianchun Zeng
- Department of Medical Imaging, Guizhou Provincial People Hospital, No.83, East Zhongshan Road, Nanming District, 550002, Guiyang City, China
| |
Collapse
|
24
|
Chen F, Jiang F, Ma J, Alghamdi MA, Zhu Y, Yong JWH. Intersecting planetary health: Exploring the impacts of environmental stressors on wildlife and human health. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116848. [PMID: 39116691 DOI: 10.1016/j.ecoenv.2024.116848] [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: 02/29/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
This comprehensive review articulates critical insights into the nexus of environmental stressors and their health impacts across diverse species, underscoring significant findings that reveal profound effects on both wildlife and human health systems. Central to our examination is the role of pollutants, climate variables, and pathogens in contributing to complex disease dynamics and physiological disruptions, with particular emphasis on immune and endocrine functions. This research brings to light emerging evidence on the severe implications of environmental pressures on a variety of taxa, including predatory mammals, raptorial birds, seabirds, fish, and humans, which are pivotal as indicators of broader ecosystem health and stability. We delve into the nuanced interplay between environmental degradation and zoonotic diseases, highlighting novel intersections that pose significant risks to biodiversity and human populations. The review critically evaluates current methodologies and advances in understanding the morphological, histopathological, and biochemical responses of these organisms to environmental stressors. We discuss the implications of our findings for conservation strategies, advocating for a more integrated approach that incorporates the dynamics of zoonoses and pollution control. This synthesis not only contributes to the academic discourse but also aims to influence policy by aligning with the Global Goals for Sustainable Development. It underscores the urgent need for sustainable interactions between humans and their environments, which are critical for preserving biodiversity and ensuring global health security. By presenting a detailed analysis of the interdependencies between environmental stressors and biological health, this review highlights significant gaps in current research and provides a foundation for future studies aimed at mitigating these pressing issues. Our study is significant as it proposes integrative and actionable strategies to address the challenges at the intersection of environmental change and public health, marking a crucial step forward in planetary health science.
Collapse
Affiliation(s)
- Fu Chen
- School of Public Administration, Hohai University, Nanjing 211100, China.
| | - Feifei Jiang
- School of Public Administration, Hohai University, Nanjing 211100, China.
| | - Jing Ma
- School of Public Administration, Hohai University, Nanjing 211100, China.
| | - Mohammed A Alghamdi
- Department of Laboratory & Blood Bank, Security Forces Hospital, Mecca, Saudi Arabia.
| | - Yanfeng Zhu
- School of Environment and Spatial Informatics, China University of Mining and Technology, Xuzhou 221000, China.
| | - Jean Wan Hong Yong
- Department of Biosystems and Technology, Swedish University of Agricultural Sciences, Alnarp 23456, Sweden.
| |
Collapse
|
25
|
Li S, Du Z, Ma H, Cai L, Liu X, He J. Mendelian randomization provides causal association between COVID-19 and thyroid cancer: insights from a multi-cancer analysis. Front Oncol 2024; 14:1419020. [PMID: 39319057 PMCID: PMC11419959 DOI: 10.3389/fonc.2024.1419020] [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: 04/17/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Since the onset of the COVID-19 pandemic, the SARS-CoV-2 virus has caused over 600 million confirmed infections and more than 6.8 million deaths worldwide, with ongoing implications for human health. COVID-19 has been extensively documented to have extrapulmonary manifestations due to the widespread expression of necessary ACE2 receptors in the human body. Nevertheless, the association between COVID-19 and cancer risk remains inadequately explored. This study employs Mendelian randomization (MR) methods to examine the causal relationship between genetic variations associated with COVID-19 and the risk of developing cancer. The findings indicate that COVID-19 has negligible impact on most cancer risks. Interestingly, a higher COVID-19 impact is associated with a decreased risk of thyroid cancer. In summary, our findings demonstrate a genetic correlation between COVID-19 and thyroid cancer, contributing to our understanding of the interplay between COVID-19 and cancer risk.
Collapse
Affiliation(s)
- Shuhong Li
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Zedong Du
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Hui Ma
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Liang Cai
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao Liu
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Jie He
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Hong S, Wu X, Feng H, Zhang Q, Wang X, Chang M, Chen X, Liu W. Risk factors for patients with tracheal stenosis: a systematic review and meta-analysis. J Int Med Res 2024; 52:3000605241275884. [PMID: 39263927 PMCID: PMC11402100 DOI: 10.1177/03000605241275884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
OBJECTIVE To systematically evaluate potential risk factors for tracheal stenosis and to provide a reference for the prevention and management of patients with this condition. METHODS Databases were searched to identify studies of the risk factors for tracheal stenosis, from their inception to October 2023, then a meta-analysis was performed. The study was registered with PROSPERO under the registration number CRD42023428906. RESULTS Ten studies of a total of 2525 patients were included. The meta-analysis showed that tracheotomy, diabetes, the duration of intubation, the duration of mechanical ventilation, respiratory tract infection, a high incision, and a ratio of intratracheal tube cuff diameter (C)/transverse diameter at the level of the clavicle (T) >150% were risk factors for the development of tracheal stenosis. CONCLUSION Measures such as shortening the duration of mechanical ventilation and intubation, reducing and avoiding tracheotomy after prolonged intubation, early tracheotomy in patients with obesity who require prolonged mechanical ventilation, appropriate choices of incision location and catheter, the maintenance of appropriate C/T, the prevention of respiratory infection, and the control of diabetes mellitus should limit the risk of tracheal stenosis.
Collapse
Affiliation(s)
- Siqi Hong
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaobing Wu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haihuan Feng
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qing Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaohan Wang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Minmin Chang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiuli Chen
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weijuan Liu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Xavier DM, Abreu RAL, Corrêa FG, Silva WT, Silva SN, Galvão EL, Junior MGDN. Effects of respiratory muscular training in post-covid-19 patients: a systematic review and meta-analysis of randomized controlled trials. BMC Sports Sci Med Rehabil 2024; 16:181. [PMID: 39192351 DOI: 10.1186/s13102-024-00954-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Post-Covid-19 syndrome is defined as non-self-sustaining signs and/or symptoms lasting more than 12 weeks, occurring during or after a Covid-19 infection. The primary outcome was the analysis of the respiratory muscle training (RMT) result in respiratory muscle strength, (maximum inspiratory pressure (MIP) e maximum expiratory pressure (MEP)); and the secondary results were the analysis of lung function, dyspnea, quality of life (QoL), fatigue and functional performance. METHODS The PICO description for this research was: P: patients diagnosed with post-Covid-19; I: RMT; C: Sham or simulated inspiratory or expiratory muscle training and usual care; O: MIP, MEP, Lung Function, level of dyspnea, QoL and functional performance. On January 15, 2024, the following databases were consulted: PubMed, Lilacs, Cochrane Library, PEDro and EMBASE. Randomized clinical trials were included without restrictions on year of publication or language. The data selection and extraction steps were carried out by two independent reviewers. RESULTS The search in the databases resulted in a total of 14,216 studies, and after the eligibility process, 7 studies were included with a sample of 527 patients. The MIP results suffered a statistically significant increase, that is, the RMT was favorable to improve the MIP (MD = 29.55cmH2O IC 95%: 7.56cmH2O to 51.54cmH2O, p = 0,00001). For the MEP outcome, the results were statistically significant in favor of RMT (MD = 10.93cmH2O CI 95%: 3.65cmH2O to 18.21cmH2O, p = 0.00001). We also noticed a significant improvement for the group that received the RMT in the distance covered in the 6-Minute Walk Test (6MWT) MD = 40.70 m CI 95%: 18.23 m to 65.17 m%, p = 0.01). CONCLUSION We noticed that RMT is being used in patients with respiratory diseases, including post-Covid-19. Our systematic review observed that this training provides an increase in inspiratory and expiratory muscle strength, a reduction in dyspnea levels, and an increase in the distance covered in the 6MWT and improved QoL in post-covid patients after intervention.
Collapse
Affiliation(s)
- Diego Mendes Xavier
- Department of Physiotherapy and Postgraduate Program in Rehabilitation and Functional Performance, Federal University of Vales do Jequitinhonha and Mucuri, Rodovia MG 367, Km 583, número 5000 Alto da Jacubá. CEP. 39100-000, Diamantina, 31330670, MG, Brazil.
| | | | - Fabiane Gontijo Corrêa
- Department of Physiotherapy and Postgraduate Program in Rehabilitation and Functional Performance, Federal University of Vales do Jequitinhonha and Mucuri, Rodovia MG 367, Km 583, número 5000 Alto da Jacubá. CEP. 39100-000, Diamantina, 31330670, MG, Brazil
| | - Whesley Tanor Silva
- Fundação Oswaldo Cruz, Instituto Nacional de Infectologia Evandro Chagas, Rio de Janeiro, Brasil
| | - Sarah Nascimento Silva
- Clinical Research and Public Policies in Infectious-Parasitic Diseases, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
| | - Endi Lanza Galvão
- Programa de Pós Graduação em Reabilitação e Desempenho Funcional, Department of Physiotherapy, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina Minas, Gerais, Brazil
| | | |
Collapse
|
28
|
Frangogiannis NG. The fate and role of the pericytes in myocardial diseases. Eur J Clin Invest 2024; 54:e14204. [PMID: 38586936 DOI: 10.1111/eci.14204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
The adult mammalian heart contains a large population of pericytes that play important roles in homeostasis and disease. In the normal heart, pericytes regulate microvascular permeability and flow. Myocardial diseases are associated with marked alterations in pericyte phenotype and function. This review manuscript discusses the role of pericytes in cardiac homeostasis and disease. Following myocardial infarction (MI), cardiac pericytes participate in all phases of cardiac repair. During the inflammatory phase, pericytes may secrete cytokines and chemokines and may regulate leukocyte trafficking, through formation of intercellular gaps that serve as exit points for inflammatory cells. Moreover, pericyte contraction induces microvascular constriction, contributing to the pathogenesis of 'no-reflow' in ischemia and reperfusion. During the proliferative phase, pericytes are activated by growth factors, such as transforming growth factor (TGF)-β and contribute to fibrosis, predominantly through secretion of fibrogenic mediators. A fraction of pericytes acquires fibroblast identity but contributes only to a small percentage of infarct fibroblasts and myofibroblasts. As the scar matures, pericytes form a coat around infarct neovessels, promoting stabilization of the vasculature. Pericytes may also be involved in the pathogenesis of chronic heart failure, by regulating inflammation, fibrosis, angiogenesis and myocardial perfusion. Pericytes are also important targets of viral infections (such as SARS-CoV2) and may be implicated in the pathogenesis of cardiac complications of COVID19. Considering their role in myocardial inflammation, fibrosis and angiogenesis, pericytes may be promising therapeutic targets in myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
29
|
Pechous RD, Malaviarachchi PA, Banerjee SK, Byrum SD, Alkam DH, Ghaffarieh A, Kurten RC, Kennedy JL, Zhang X. An ex vivo human precision-cut lung slice platform provides insight into SARS-CoV-2 pathogenesis and antiviral drug efficacy. J Virol 2024; 98:e0079424. [PMID: 38940558 PMCID: PMC11265413 DOI: 10.1128/jvi.00794-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has claimed millions of lives since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and lung disease appears the primary cause of death in COVID-19 patients. However, the underlying mechanisms of COVID-19 pathogenesis remain elusive, and there is no existing model where human disease can be faithfully recapitulated and conditions for the infection process can be experimentally controlled. Herein we report the establishment of an ex vivo human precision-cut lung slice (hPCLS) platform for studying SARS-CoV-2 pathogenicity and innate immune responses, and for evaluating the efficacy of antiviral drugs against SARS-CoV-2. We show that while SARS-CoV-2 continued to replicate during the course of infection of hPCLS, infectious virus production peaked within 2 days, and rapidly declined thereafter. Although most proinflammatory cytokines examined were induced by SARS-CoV-2 infection, the degree of induction and types of cytokines varied significantly among hPCLS from individual donors. Two cytokines in particular, IP-10 and IL-8, were highly and consistently induced, suggesting a role in the pathogenesis of COVID-19. Histopathological examination revealed focal cytopathic effects late in the infection. Transcriptomic and proteomic analyses identified molecular signatures and cellular pathways that are largely consistent with the progression of COVID-19 in patients. Furthermore, we show that homoharringtonine, a natural plant alkaloid derived from Cephalotoxus fortunei, not only inhibited virus replication but also production of pro-inflammatory cytokines, and thus ameliorated the histopathological changes caused by SARS-CoV-2 infection, demonstrating the usefulness of the hPCLS platform for evaluating antiviral drugs. IMPORTANCE Here, established an ex vivo human precision-cut lung slice platform for assessing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, viral replication kinetics, innate immune response, disease progression, and antiviral drugs. Using this platform, we identified early induction of specific cytokines, especially IP-10 and IL-8, as potential predictors for severe coronavirus disease 2019 (COVID-19), and uncovered a hitherto unrecognized phenomenon that while infectious virus disappears at late times of infection, viral RNA persists and lung histopathology commences. This finding may have important clinical implications for both acute and post-acute sequelae of COVID-19. This platform recapitulates some of the characteristics of lung disease observed in severe COVID-19 patients and is therefore a useful platform for understanding mechanisms of SARS-CoV-2 pathogenesis and for evaluating the efficacy of antiviral drugs.
Collapse
Affiliation(s)
- Roger D. Pechous
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Priyangi A. Malaviarachchi
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Srijon K. Banerjee
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Duah H. Alkam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Alireza Ghaffarieh
- Department of Ophthalmology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Richard C. Kurten
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Lung Cell Biology Laboratory, Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Lung Cell Biology Laboratory, Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
30
|
Rossi PAQ, Gomes R, Nascimento Salazar TCD, Lustosa Barros EM, Vasconcelos S, da Silva AS, Pereira EM, Melo VB, Fonseca MHG, Teixeira CR, Furtado GP, Pontes LQ, Khouri R, Vasconcelos B, Almeida SSD, Werneck GL, Rossi FE, Santos MAPD. Physical Activity at Different Life Stages and Its Consequence on the Initial Immunization and Inflammatory Response Against COVID-19. J Phys Act Health 2024; 21:717-725. [PMID: 38663845 DOI: 10.1123/jpah.2023-0370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/16/2024] [Accepted: 03/08/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND To evaluate the influence of previous physical activity (PA) during childhood, adolescence, and current PA practice on the production of antibodies and inflammatory response between the first and second doses of the COVID-19 vaccine. METHODS Fifty-nine men and 56 women were evaluated before the first vaccine, and 12 weeks later, blood samples were taken to quantify production of anti-severe acute respiratory syndrome coronavirus-2 immunoglobulin G antibodies and cytokines. Previous PA during childhood and adolescence was self-referred, and current PA was assessed using the International Physical Activity Questionnaire. RESULTS A positive and significant association was observed only between PA practice during adolescence and an increase in antibody production in adulthood (β = 2012.077, 95% confidence interval, 257.7953-3766.358, P = .025). Individuals who practiced PA during adolescence showed higher production of antibodies between the first and second vaccine dose compared to nonpractitioners (P = .025) and those that accumulated ≥150 minutes per week of current moderate-vigorous PA (MVPA), and presented higher antibody production in relation to who did <150 minutes per week of MVPA (P = .046). Individuals that were practitioners during childhood produced higher G-CSF (P = .047), and those that accumulated ≥150 minutes per week of current MVPA demonstrated lower IP-10 levels (P = .033). However, PA practitioners during adolescence presented higher G-CSF (P = .025), IL-17 (P = .038), IL-1RA (P = .005), IL-1β (P = .020), and IL-2 (P = .026) levels. CONCLUSION Our results suggest that adults that accumulated at least 150 minutes of MVPA per week or practiced PA during adolescence developed an improved immune and inflammatory response against COVID-19 vaccination.
Collapse
Affiliation(s)
- Priscila Almeida Queiroz Rossi
- Postgraduation Student in Science and Health, Federal University of Piaui, Teresina, PI, Brazil
- Department of Biophysics and Physiology, Nucleus of Study in Physiology Applied to Performance and Health (NEFADS), Federal University of Piaui, Teresina, PI, Brazil
| | - Regis Gomes
- Biotecnologia, Escritório Técnico da Fiocruz Ceará, Eusébio, CE, Brazil
| | - Teresa Cristina do Nascimento Salazar
- Postgraduation Student in Science and Health, Federal University of Piaui, Teresina, PI, Brazil
- Department of Biophysics and Physiology, Nucleus of Study in Physiology Applied to Performance and Health (NEFADS), Federal University of Piaui, Teresina, PI, Brazil
| | - Esmeralda Maria Lustosa Barros
- Department of Biophysics and Physiology, Nucleus of Study in Physiology Applied to Performance and Health (NEFADS), Federal University of Piaui, Teresina, PI, Brazil
| | | | | | | | - Vitoria Braga Melo
- Unidade de Apoio ao Diagnóstico de Covid-19 (UNADIG), Fiocruz Ceará, Eusébio, CE, Brazil
| | | | | | | | | | - Ricardo Khouri
- Gonçalo Moniz Institute, Fiocruz Bahia, Salvador, BA, Brazil
- Faculty of Medicine, Federal University of Bahia, Salvador,BA, Brazil
| | - Beatriz Vasconcelos
- Gonçalo Moniz Institute, Fiocruz Bahia, Salvador, BA, Brazil
- Faculty of Medicine, Federal University of Bahia, Salvador,BA, Brazil
| | - Sandro Soares de Almeida
- Department of Obstetrician, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, Brazil
- Faculdade Anhanguera de Guarulhos, Guarulhos,SP, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Fabrício Eduardo Rossi
- Department of Physical Education, Immunometabolism of Skeletal Muscle and Exercise Research Group, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
- Graduate Program in Science and Health, Federal University of Piauí (UFPI), Teresina, PI, Brazil
- Graduate Program in Movement Science, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - Marcos Antonio Pereira Dos Santos
- Department of Biophysics and Physiology, Nucleus of Study in Physiology Applied to Performance and Health (NEFADS), Federal University of Piaui, Teresina, PI, Brazil
| |
Collapse
|
31
|
Rodriguez-Espada A, Salgado-de la Mora M, Rodriguez-Paniagua BM, Limon-de la Rosa N, Martinez-Gutierrez MI, Pastrana-Brandes S, Navarro-Alvarez N. Histopathological impact of SARS-CoV-2 on the liver: Cellular damage and long-term complications. World J Gastroenterol 2024; 30:2866-2880. [PMID: 38947288 PMCID: PMC11212712 DOI: 10.3748/wjg.v30.i22.2866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/08/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the highly pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily impacts the respiratory tract and can lead to severe outcomes such as acute respiratory distress syndrome, multiple organ failure, and death. Despite extensive studies on the pathogenicity of SARS-CoV-2, its impact on the hepatobiliary system remains unclear. While liver injury is commonly indicated by reduced albumin and elevated bilirubin and transaminase levels, the exact source of this damage is not fully understood. Proposed mechanisms for injury include direct cytotoxicity, collateral damage from inflammation, drug-induced liver injury, and ischemia/hypoxia. However, evidence often relies on blood tests with liver enzyme abnormalities. In this comprehensive review, we focused solely on the different histopathological manifestations of liver injury in COVID-19 patients, drawing from liver biopsies, complete autopsies, and in vitro liver analyses. We present evidence of the direct impact of SARS-CoV-2 on the liver, substantiated by in vitro observations of viral entry mechanisms and the actual presence of viral particles in liver samples resulting in a variety of cellular changes, including mitochondrial swelling, endoplasmic reticulum dilatation, and hepatocyte apoptosis. Additionally, we describe the diverse liver pathology observed during COVID-19 infection, encompassing necrosis, steatosis, cholestasis, and lobular inflammation. We also discuss the emergence of long-term complications, notably COVID-19-related secondary sclerosing cholangitis. Recognizing the histopathological liver changes occurring during COVID-19 infection is pivotal for improving patient recovery and guiding decision-making.
Collapse
Affiliation(s)
- Alfonso Rodriguez-Espada
- Department of Molecular Biology, Universidad Panamericana School of Medicine, Campus México, Mexico 03920, Mexico
| | - Moises Salgado-de la Mora
- Department of Internal Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico 14080, Mexico
| | | | - Nathaly Limon-de la Rosa
- Department of Surgery, University of Colorado Anschutz Medical Campus, Denver, CO 80045, United States
| | | | - Santiago Pastrana-Brandes
- Department of Molecular Biology, Universidad Panamericana School of Medicine, Campus México, Mexico 03920, Mexico
| | - Nalu Navarro-Alvarez
- Department of Molecular Biology, Universidad Panamericana School of Medicine, Campus México, Mexico 03920, Mexico
- Department of Surgery, University of Colorado Anschutz Medical Campus, Denver, CO 80045, United States
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico 14080, Mexico
| |
Collapse
|
32
|
Wu TTH, Travaglini KJ, Rustagi A, Xu D, Zhang Y, Andronov L, Jang S, Gillich A, Dehghannasiri R, Martínez-Colón GJ, Beck A, Liu DD, Wilk AJ, Morri M, Trope WL, Bierman R, Weissman IL, Shrager JB, Quake SR, Kuo CS, Salzman J, Moerner W, Kim PS, Blish CA, Krasnow MA. Interstitial macrophages are a focus of viral takeover and inflammation in COVID-19 initiation in human lung. J Exp Med 2024; 221:e20232192. [PMID: 38597954 PMCID: PMC11009983 DOI: 10.1084/jem.20232192] [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: 11/28/2023] [Revised: 02/09/2024] [Accepted: 03/04/2024] [Indexed: 04/11/2024] Open
Abstract
Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.
Collapse
Affiliation(s)
- Timothy Ting-Hsuan Wu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Kyle J. Travaglini
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Arjun Rustagi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Yue Zhang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Leonid Andronov
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - SoRi Jang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Astrid Gillich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| | - Roozbeh Dehghannasiri
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Giovanny J. Martínez-Colón
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aimee Beck
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Dan Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Aaron J. Wilk
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Winston L. Trope
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rob Bierman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph B. Shrager
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Stephen R. Quake
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Christin S. Kuo
- Department of Pediatrics, Pulmonary Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia Salzman
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - W.E. Moerner
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Catherine A. Blish
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Mark A. Krasnow
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA, USA
| |
Collapse
|
33
|
Greenwald MA, Namin S, Zajdowicz J, Jones AL, Fritts L, Kuehnert MJ, Miller CJ, Ray G. Testing of tissue specimens obtained from SARS-CoV-2 nasopharyngeal swab-positive donors. Cell Tissue Bank 2024; 25:583-604. [PMID: 37995051 PMCID: PMC11143015 DOI: 10.1007/s10561-023-10119-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023]
Abstract
Risk for transmission of SARS-CoV-2 through allogeneic human tissue transplantation is unknown. To further evaluate the risk of virus transmission, tissues were obtained from deceased donors who had tested positive for SARS-CoV-2 RNA via nasopharyngeal swab. This study evaluated an array of human tissues recovered for transplantation, including bone, tendon, skin, fascia lata, vascular tissues, and heart valves. Tissue samples and plasma or serum samples, if available, were tested for viral RNA (vRNA) using a real time PCR system for the presence of virus RNA. All samples were tested in quadruplicate for both subgenomic (sgRNA) and genomic (gRNA) RNA encoding the SARS-CoV-2 nucleocapsid gene. Amplification of a cellular housekeeping gene served as the positive control for every sample. A total of 47 tissue samples from 17 donors were tested for SARS-CoV-2 RNA. Four donors had plasma or serum available for paired testing. SARS-CoV-2 RNA was not detected from any tissue or plasma/serum sample tested. Based on these findings, risk of transmission through the transplantation of tissue types studied from SARS-CoV-2 infected donors is likely to be low.
Collapse
Affiliation(s)
- Melissa A Greenwald
- Donor Alliance, Denver, CO, USA.
- Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | | | | | | | | | - Matthew J Kuehnert
- MTF Biologics, Edison, NJ, USA
- Hackensack Meridian School of Medicine, Hackensack, NJ, USA
| | | | | |
Collapse
|
34
|
Xiong X, Zheng Z, Liu C, Wang X, Luo S, Xie Q, Liu Y, Chen Q, Zheng M. Unveiling the metabolic and coagulation disruptions in SARS-CoV-2-associated acute macular neuroretinopathy: A case-control study. J Med Virol 2024; 96:e29714. [PMID: 38837795 DOI: 10.1002/jmv.29714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/20/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
SARS-CoV-2 infection has been associated with the increased incidence of acute macular neuroretinopathy (AMN), an infrequent ocular disorder. However, the precise mechanisms underpinning AMN in the context of SARS-CoV-2 infection (AMN-SARS-CoV-2) remain elusive. In this case-control study, 14 patients diagnosed with AMN-SARS-CoV-2 between 2022/12 and 2023/3 were enrolled and compared with 14 SARS-CoV-2-infected individuals without AMN, who served as controls (SARS-CoV-2-no AMN). Metabolomic profiling using ultrahigh-performance liquid chromatography-online electrospray mass spectrometry revealed significant alterations in serum metabolites in AMN-SARS-CoV-2 patients. Coagulation abnormalities were observed in AMN-SARS-CoV-2 patients, and their relationship with metabolic disorders was studied. Finally, a predictive model for AMN-SARS-CoV-2 was established. Seventy-six upregulated and 42 downregulated metabolites were identified in AMN-SARS-CoV-2 cases. Notably, arginine metabolism within the urea cycle was significantly altered, evidenced by variations in ornithine, citrulline, l-proline, and ADAM levels, correlating with abnormal coagulation markers like platelet crit, fibrinogen degradation product, and fibrinogen. Additionally, increased arginase 1 (AGR1) activity within the urea cycle and reduced nitric oxide synthase activity were observed in AMN-SARS-CoV-2. The integration of urea cycle metabolite levels with coagulation parameters yielded a robust discriminatory model for AMN-SARS-CoV-2, as evidenced by an area under the curve of 0.96. The findings of the present study enhance our comprehension of the underlying metabolic mechanisms associated with AMN-SARS-CoV-2 and offer potential diagnostic markers for this uncommon ocular disorder within the context of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xiaojing Xiong
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Zheng
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunlin Liu
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyu Wang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuai Luo
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinqin Xie
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Liu
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingwei Chen
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minming Zheng
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Borczuk AC. Pathology of COVID-19 Lung Disease. Surg Pathol Clin 2024; 17:203-214. [PMID: 38692805 DOI: 10.1016/j.path.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
The pathology of severe COVID-19 lung injury is predominantly diffuse alveolar damage, with other reported patterns including acute fibrinous organizing pneumonia, organizing pneumonia, and bronchiolitis. Lung injury was caused by primary viral injury, exaggerated immune responses, and superinfection with bacteria and fungi. Although fatality rates have decreased from the early phases of the pandemic, persistent pulmonary dysfunction occurs and its pathogenesis remains to be fully elucidated.
Collapse
Affiliation(s)
- Alain C Borczuk
- Department of Pathology, Northwell Health, 2200 Northern Boulevard Suite 104, Greenvale, NY 11548, USA.
| |
Collapse
|
36
|
Lenz C, Slack MPE, Shea KM, Reinert RR, Taysi BN, Swerdlow DL. Long-Term effects of COVID-19: a review of current perspectives and mechanistic insights. Crit Rev Microbiol 2024; 50:315-328. [PMID: 37074754 DOI: 10.1080/1040841x.2023.2190405] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/25/2023] [Indexed: 04/20/2023]
Abstract
Although SARS-CoV-2, responsible for COVID-19, is primarily a respiratory infection, a broad spectrum of cardiac, pulmonary, neurologic, and metabolic complications can occur. More than 50 long-term symptoms of COVID-19 have been described, and as many as 80% of patients may develop ≥1 long-term symptom. To summarize current perspectives of long-term sequelae of COVID-19, we conducted a PubMed search describing the long-term cardiovascular, pulmonary, gastrointestinal, and neurologic effects post-SARS-CoV-2 infection and mechanistic insights and risk factors for the above-mentioned sequelae. Emerging risk factors of long-term sequelae include older age (≥65 years), female sex, Black or Asian race, Hispanic ethnicity, and presence of comorbidities. There is an urgent need to better understand ongoing effects of COVID-19. Prospective studies evaluating long-term effects of COVID-19 in all body systems and patient groups will facilitate appropriate management and assess burden of care. Clinicians should ensure patients are followed up and managed appropriately, especially those in at-risk groups. Healthcare systems worldwide need to develop approaches to follow-up and support patients recovering from COVID-19. Surveillance programs can enhance prevention and treatment efforts for those most vulnerable.
Collapse
Affiliation(s)
| | - Mary P E Slack
- Griffith University, School of Medicine and Dentistry, Griffith University Gold Coast campus, Queensland, Australia
| | | | | | | | | |
Collapse
|
37
|
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: 7] [Impact Index Per Article: 7.0] [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
|
38
|
Abolhasani FS, Moein M, Rezaie N, Sheikhimehrabadi P, Shafiei M, Afkhami H, Modaresi M. Occurrence of COVID-19 in cystic fibrosis patients: a review. Front Microbiol 2024; 15:1356926. [PMID: 38694803 PMCID: PMC11061495 DOI: 10.3389/fmicb.2024.1356926] [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: 12/16/2023] [Accepted: 03/11/2024] [Indexed: 05/04/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic ailment caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. This autosomal recessive disorder is characterized by diverse pathobiological abnormalities, such as the disorder of CFTR channels in mucosal surfaces, caused by inadequate clearance of mucus and sputum, in addition to the malfunctioning of mucous organs. However, the primary motive of mortality in CF patients is pulmonary failure, which is attributed to the colonization of opportunistic microorganisms, formation of resistant biofilms, and a subsequent decline in lung characteristics. In December 2019, the World Health Organization (WHO) declared the outbreak of the radical coronavirus disease 2019 (COVID-19) as a worldwide public health crisis, which unexpectedly spread not only within China but also globally. Given that the respiration system is the primary target of the COVID-19 virus, it is crucial to investigate the impact of COVID-19 on the pathogenesis and mortality of CF patients, mainly in the context of acute respiratory distress syndrome (ARDS). Therefore, the goal of this review is to comprehensively review the present literature on the relationship between cystic fibrosis, COVID-19 contamination, and development of ARDS. Several investigations performed during the early stages of the virus outbreak have discovered unexpected findings regarding the occurrence and effectiveness of COVID-19 in individuals with CF. Contrary to initial expectancies, the rate of infection and the effectiveness of the virus in CF patients are lower than those in the overall population. This finding may be attributed to different factors, including the presence of thick mucus, social avoidance, using remedies that include azithromycin, the fairly younger age of CF patients, decreased presence of ACE-2 receptors, and the effect of CFTR channel disorder on the replication cycle and infectivity of the virus. However, it is important to notice that certain situations, which include undergoing a transplant, can also doubtlessly boost the susceptibility of CF patients to COVID-19. Furthermore, with an increase in age in CF patients, it is vital to take into account the prevalence of the SARS-CoV-2 virus in this population. Therefore, ordinary surveillance of CF patients is vital to evaluate and save the population from the capability of transmission of the virus given the various factors that contribute to the spread of the SARS-CoV-2 outbreak in this precise organization.
Collapse
Affiliation(s)
- Fatemeh Sadat Abolhasani
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masood Moein
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Rezaie
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, School of Medicine, Shahed University, Tehran, Iran
| | - Mohammadreza Modaresi
- Pediatric Pulmonary Disease and Sleep Medicine Research Center, Pediatric Center of Excellence, Children's Medical Center, Tehran, Iran
- Cystic Fibrosis Research Center, Iran CF Foundation (ICFF), Tehran, Iran
| |
Collapse
|
39
|
Alfieri L, Franceschetti L, Frisoni P, Bonato O, Radaelli D, Bonuccelli D, D’Errico S, Neri M. Cardiac SARS-CoV-2 Infection, Involvement of Cytokines in Postmortem Immunohistochemical Study. Diagnostics (Basel) 2024; 14:787. [PMID: 38667433 PMCID: PMC11049034 DOI: 10.3390/diagnostics14080787] [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: 02/29/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
In the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, significant attention was given to pulmonary manifestations. However, cardiac involvement is increasingly recognized as a critical factor influencing the prognosis, leading to myocardial damage, heart failure, acute coronary syndromes, potentially lethal arrhythmic events, and sudden cardiac death. Despite these findings, there is a lack of studies detailing the necroscopic, macroscopic, and microscopic cardiac changes associated with SARS-CoV-2. This study aimed to investigate the presence of SARS-CoV-2 viral proteins in cardiac tissue using immunohistochemical techniques to assess viral tropism. The analysis of cardiac tissue samples from deceased subjects, in different stages of conservation, confirmed to be positive for SARS-CoV-2 via reverse transcriptase-polymerase chain reaction (RT-PCR), showed immunopositivity for the SARS-CoV-2-NP viral antigen in 33% of cases. Notably, the presence of leukocyte infiltrates sufficient for diagnosing lymphocytic myocarditis was not observed. The central proinflammatory cytokines involved in the pathogenetic mechanism of coronavirus disease 19 (COVID-19) were researched using the immunohistochemical method. A significant increase in cytokine expression was detected, indicating myocardial involvement and dysfunction during SARS-CoV-2 infection. These findings suggest that the immunohistochemical detection of SARS-CoV-2 viral antigens and inflammatory cytokine expression in cardiac tissue could be crucial for a proper forensic assessment of the cause of death, even in sudden cardiac death.
Collapse
Affiliation(s)
- Letizia Alfieri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Lorenzo Franceschetti
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133 Milano, Italy
| | - Paolo Frisoni
- Unit of Legal Medicine, AUSL Romagna, G.B. Morgagni-L. Pierantoni Hospital, 47100 Forlì, Italy;
| | - Omar Bonato
- Unit of Legal Medicine, AULSS 5 Polesana, 45100 Rovigo, Italy;
| | - Davide Radaelli
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (D.R.); (S.D.)
| | - Diana Bonuccelli
- Department of Legal Medicine, Territorial Unit USL Toscana Nord-Ovest, 55100 Lucca, Italy;
| | - Stefano D’Errico
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (D.R.); (S.D.)
| | - Margherita Neri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| |
Collapse
|
40
|
Gandikota C, Vaddadi K, Sivasami P, Huang C, Liang Y, Pushparaj S, Deng X, Channappanava R, Metcalf JP, Liu L. The use of human iPSC-derived alveolar organoids to explore SARS-CoV-2 variant infections and host responses. J Med Virol 2024; 96:e29579. [PMID: 38572923 PMCID: PMC11603130 DOI: 10.1002/jmv.29579] [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: 10/19/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) primarily targets the respiratory system. Physiologically relevant human lung models are indispensable to investigate virus-induced host response and disease pathogenesis. In this study, we generated human induced pluripotent stem cell (iPSC)-derived alveolar organoids (AOs) using an established protocol that recapitulates the sequential steps of in vivo lung development. AOs express alveolar epithelial type II cell protein markers including pro-surfactant protein C and ATP binding cassette subfamily A member 3. Compared to primary human alveolar type II cells, AOs expressed higher mRNA levels of SARS-CoV-2 entry factors, angiotensin-converting enzyme 2 (ACE2), asialoglycoprotein receptor 1 (ASGR1) and basigin (CD147). Considering the localization of ACE2 on the apical side in AOs, we used three AO models, apical-in, sheared and apical-out for SARS-CoV-2 infection. All three models of AOs were robustly infected with the SARS-CoV-2 irrespective of ACE2 accessibility. Antibody blocking experiment revealed that ASGR1 was the main receptor for SARS-CoV2 entry from the basolateral in apical-in AOs. AOs supported the replication of SARS-CoV-2 variants WA1, Alpha, Beta, Delta, and Zeta and Omicron to a variable degree with WA1 being the highest and Omicron being the least. Transcriptomic profiling of infected AOs revealed the induction of inflammatory and interferon-related pathways with NF-κB signaling being the predominant host response. In summary, iPSC-derived AOs can serve as excellent human lung models to investigate infection of SARS-CoV-2 variants and host responses from both apical and basolateral sides.
Collapse
Affiliation(s)
- Chaitanya Gandikota
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Kishore Vaddadi
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Pulavendran Sivasami
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Chaoqun Huang
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Yurong Liang
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Samuel Pushparaj
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| | - Xufang Deng
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Rudragouda Channappanava
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, Oklahoma
| | - Jordan P. Metcalf
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Lin Liu
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma
| |
Collapse
|
41
|
Violi F, Harenberg J, Pignatelli P, Cammisotto V. COVID-19 and Long-COVID Thrombosis: From Clinical and Basic Science to Therapeutics. Thromb Haemost 2024; 124:286-296. [PMID: 37967846 DOI: 10.1055/s-0043-1776713] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Coronavirus infectious disease-19 (COVID-19) is a pandemic characterized by serious lung disease and thrombotic events in the venous and circulation trees, which represent a harmful clinical sign of poor outcome. Thrombotic events are more frequent in patients with severe disease requiring intensive care units and are associated with platelet and clotting activation. However, after resolution of acute infection, patients may still have clinical sequelae, the so-called long-COVID-19, including thrombotic events again in the venous and arterial circulation. The mechanisms accounting for thrombosis in acute and long COVID-19 have not been fully clarified; interactions of COVID-19 with angiotensin converting enzyme 2 or toll-like receptor family or infection-induced cytokine storm have been suggested to be implicated in endothelial cells, leucocytes, and platelets to elicit clotting activation in acute as well in chronic phase of the disease. In acute COVID-19, prophylactic or full doses of anticoagulants exert beneficial effects even if the dosage choice is still under investigation; however, a residual risk still remains suggesting a need for a more appropriate therapeutic approach. In long COVID-19 preliminary data provided useful information in terms of antiplatelet treatment but definition of candidates for thrombotic prophylaxis is still undefined.
Collapse
Affiliation(s)
- Francesco Violi
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Via Orazio, Naples, Italy
| | - Job Harenberg
- Medical Faculty Mannheim, Ruprecht-karls University Heidelberg, Heidelberg, Germany
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Via Orazio, Naples, Italy
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
42
|
Schädler J, Azeke AT, Ondruschka B, Steurer S, Lütgehetmann M, Fitzek A, Möbius D. Concordance between MITS and conventional autopsies for pathological and virological diagnoses. Int J Legal Med 2024; 138:431-442. [PMID: 37837537 PMCID: PMC10861633 DOI: 10.1007/s00414-023-03088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 10/16/2023]
Abstract
In pandemics or to further study highly contagious infectious diseases, new strategies are needed for the collection of post-mortem tissue samples to identify the pathogen as well as its morphological impact. In this study, an ultrasound-guided minimally invasive tissue sampling (MITS) protocol was developed and validated for post-mortem use. The histological and microbiological qualities of post-mortem specimens were evaluated and compared between MITS and conventional autopsy (CA) in a series of COVID-19 deaths. Thirty-six ultrasound-guided MITS were performed. In five cases more, specimens for histological and virological examination were also obtained and compared during the subsequently performed CA. Summary statistics and qualitative interpretations (positive, negative) were calculated for each organ tissue sample from MITS and CA, and target genes were determined for both human cell count (beta-globin) and virus (SARS-CoV-2 specific E gene). There are no significant differences between MITS and CA with respect to the detectability of viral load in individual organs, which is why MITS can be of utmost importance and an useful alternative, especially during outbreaks of infectious diseases.
Collapse
Affiliation(s)
- Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Akhator Terence Azeke
- Department of Anatomic Pathology, Irrua Specialist Teaching Hospital, Irrua, Nigeria
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology, and Hygiene, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Fitzek
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dustin Möbius
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
43
|
Mao Y, Chen Y, Li Y, Ma L, Wang X, Wang Q, He A, Liu X, Dong T, Gao W, Xu Y, Liu L, Ren L, Liu Q, Zhou P, Hu B, Zhou Y, Tian R, Shi ZL. Deep spatial proteomics reveals region-specific features of severe COVID-19-related pulmonary injury. Cell Rep 2024; 43:113689. [PMID: 38241149 DOI: 10.1016/j.celrep.2024.113689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/23/2023] [Accepted: 01/02/2024] [Indexed: 01/21/2024] Open
Abstract
As a primary target of severe acute respiratory syndrome coronavirus 2, lung exhibits heterogeneous histopathological changes following infection. However, comprehensive insight into their protein basis with spatial resolution remains deficient, which hinders further understanding of coronavirus disease 2019 (COVID-19)-related pulmonary injury. Here, we generate a region-resolved proteomic atlas of hallmark pathological pulmonary structures by integrating histological examination, laser microdissection, and ultrasensitive proteomics. Over 10,000 proteins are quantified across 71 post-mortem specimens. We identify a spectrum of pathway dysregulations in alveolar epithelium, bronchial epithelium, and blood vessels compared with non-COVID-19 controls, providing evidence for transitional-state pneumocyte hyperplasia. Additionally, our data reveal the region-specific enrichment of functional markers in bronchiole mucus plugs, pulmonary fibrosis, airspace inflammation, and alveolar type 2 cells, uncovering their distinctive features. Furthermore, we detect increased protein expression associated with viral entry and inflammatory response across multiple regions, suggesting potential therapeutic targets. Collectively, this study provides a distinct perspective for deciphering COVID-19-caused pulmonary dysfunction by spatial proteomics.
Collapse
Affiliation(s)
- Yiheng Mao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ying Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Li
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Longda Ma
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi Wang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qi Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China; University of Chinese Academy of Sciences, Beijing, China
| | - An He
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xi Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China; University of Chinese Academy of Sciences, Beijing, China
| | - Tianyi Dong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China; University of Chinese Academy of Sciences, Beijing, China
| | - Weina Gao
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yanfen Xu
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liang Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liang Ren
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Liu
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Zhou
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou 510005, China
| | - Ben Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ruijun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Zheng-Li Shi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430030, China.
| |
Collapse
|
44
|
Musigk N, Suwalski P, Golpour A, Fairweather D, Klingel K, Martin P, Frustaci A, Cooper LT, Lüscher TF, Landmesser U, Heidecker B. The inflammatory spectrum of cardiomyopathies. Front Cardiovasc Med 2024; 11:1251780. [PMID: 38464847 PMCID: PMC10921946 DOI: 10.3389/fcvm.2024.1251780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/29/2024] [Indexed: 03/12/2024] Open
Abstract
Infiltration of the myocardium with various cell types, cytokines and chemokines plays a crucial role in the pathogenesis of cardiomyopathies including inflammatory cardiomyopathies and myocarditis. A more comprehensive understanding of the precise immune mechanisms involved in acute and chronic myocarditis is essential to develop novel therapeutic approaches. This review offers a comprehensive overview of the current knowledge of the immune landscape in cardiomyopathies based on etiology. It identifies gaps in our knowledge about cardiac inflammation and emphasizes the need for new translational approaches to improve our understanding thus enabling development of novel early detection methods and more effective treatments.
Collapse
Affiliation(s)
- Nicolas Musigk
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Phillip Suwalski
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Ainoosh Golpour
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
- Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, United States
| | - Karin Klingel
- Cardiopathology Institute for Pathology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Pilar Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Centro de Investigación Biomédica en Red Cardiovascular (CIBER-CV, ISCIII), Madrid, Spain
| | | | - Leslie T. Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Thomas F. Lüscher
- GZO-Zurich Regional Health Centre, Wetzikon & Cardioimmunology, Centre for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Royal Brompton & Harefield Hospitals and National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Ulf Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| | - Bettina Heidecker
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
| |
Collapse
|
45
|
Song Y, Lou L, Zhang K. A review of the clinical characteristics and management of immunosuppressed patients living with HIV or solid organ transplants infected with SARS-CoV-2 omicron variants. Front Public Health 2024; 12:1327093. [PMID: 38454994 PMCID: PMC10917969 DOI: 10.3389/fpubh.2024.1327093] [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: 10/24/2023] [Accepted: 02/13/2024] [Indexed: 03/09/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) omicron strain was first detected in South Africa in November 2021. Although clinical responses to SARS-CoV-2 depend on host immunity, it remains uncertain how immunosuppression affects subsequent coronavirus disease 2019-related (COVID-19-related) incidence, severity, and mortality, especially with respect to the omicron strain. Conversely, immunosuppressants are often thought to predispose to infection. To explore the associations between host immunity and infection with SARS-CoV-2 omicron variants, here we discuss two groups of immunosuppressed patients: organ transplant recipients, who generally receive exogenous immunosuppressants, and Human Immunodeficiency Virus (HIV)-infected patients, who often have disease-related immunosuppression. In summarizing the clinical features and prognoses of HIV-infected patients and human organ transplant recipients infected with SARS-CoV-2 omicron variants, we provide new insights into the pathogenesis of omicron SARS-CoV-2 and provide a framework for the management of these patients now and in the future.
Collapse
Affiliation(s)
- Yan Song
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
| | - Lixin Lou
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
| | - Kaiyu Zhang
- Department of Infectious Diseases, The First Hospital of Jilin University, Jilin, China
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
46
|
Kumar S, Granados J, Aceves M, Peralta J, Leandro AC, Thomas J, Williams-Blangero S, Curran JE, Blangero J. Pre-Infection Innate Immunity Attenuates SARS-CoV-2 Infection and Viral Load in iPSC-Derived Alveolar Epithelial Type 2 Cells. Cells 2024; 13:369. [PMID: 38474333 PMCID: PMC10931100 DOI: 10.3390/cells13050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/05/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
A large portion of the heterogeneity in coronavirus disease 2019 (COVID-19) susceptibility and severity of illness (SOI) remains poorly understood. Recent evidence suggests that SARS-CoV-2 infection-associated damage to alveolar epithelial type 2 cells (AT2s) in the distal lung may directly contribute to disease severity and poor prognosis in COVID-19 patients. Our in vitro modeling of SARS-CoV-2 infection in induced pluripotent stem cell (iPSC)-derived AT2s from 10 different individuals showed interindividual variability in infection susceptibility and the postinfection cellular viral load. To understand the underlying mechanism of the AT2's capacity to regulate SARS-CoV-2 infection and cellular viral load, a genome-wide differential gene expression analysis between the mock and SARS-CoV-2 infection-challenged AT2s was performed. The 1393 genes, which were significantly (one-way ANOVA FDR-corrected p ≤ 0.05; FC abs ≥ 2.0) differentially expressed (DE), suggest significant upregulation of viral infection-related cellular innate immune response pathways (p-value ≤ 0.05; activation z-score ≥ 3.5), and significant downregulation of the cholesterol- and xenobiotic-related metabolic pathways (p-value ≤ 0.05; activation z-score ≤ -3.5). Whilst the effect of post-SARS-CoV-2 infection response on the infection susceptibility and postinfection viral load in AT2s is not clear, interestingly, pre-infection (mock-challenged) expression of 238 DE genes showed a high correlation with the postinfection SARS-CoV-2 viral load (FDR-corrected p-value ≤ 0.05 and r2-absolute ≥ 0.57). The 85 genes whose expression was negatively correlated with the viral load showed significant enrichment in viral recognition and cytokine-mediated innate immune GO biological processes (p-value range: 4.65 × 10-10 to 2.24 × 10-6). The 153 genes whose expression was positively correlated with the viral load showed significant enrichment in cholesterol homeostasis, extracellular matrix, and MAPK/ERK pathway-related GO biological processes (p-value range: 5.06 × 10-5 to 6.53 × 10-4). Overall, our results strongly suggest that AT2s' pre-infection innate immunity and metabolic state affect their susceptibility to SARS-CoV-2 infection and viral load.
Collapse
Affiliation(s)
- Satish Kumar
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Jose Granados
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Miriam Aceves
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Juan Peralta
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Ana C. Leandro
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - John Thomas
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (J.G.); (M.A.); (J.T.)
| | - Sarah Williams-Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Joanne E. Curran
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| | - John Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (J.P.); (A.C.L.); (S.W.-B.); (J.E.C.); (J.B.)
| |
Collapse
|
47
|
Tandon P, Abrams ND, Avula LR, Carrick DM, Chander P, Divi RL, Dwyer JT, Gannot G, Gordiyenko N, Liu Q, Moon K, PrabhuDas M, Singh A, Tilahun ME, Satyamitra MM, Wang C, Warren R, Liu CH. Unraveling Links between Chronic Inflammation and Long COVID: Workshop Report. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:505-512. [PMID: 38315950 DOI: 10.4049/jimmunol.2300804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
As COVID-19 continues, an increasing number of patients develop long COVID symptoms varying in severity that last for weeks, months, or longer. Symptoms commonly include lingering loss of smell and taste, hearing loss, extreme fatigue, and "brain fog." Still, persistent cardiovascular and respiratory problems, muscle weakness, and neurologic issues have also been documented. A major problem is the lack of clear guidelines for diagnosing long COVID. Although some studies suggest that long COVID is due to prolonged inflammation after SARS-CoV-2 infection, the underlying mechanisms remain unclear. The broad range of COVID-19's bodily effects and responses after initial viral infection are also poorly understood. This workshop brought together multidisciplinary experts to showcase and discuss the latest research on long COVID and chronic inflammation that might be associated with the persistent sequelae following COVID-19 infection.
Collapse
Affiliation(s)
- Pushpa Tandon
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Natalie D Abrams
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Leela Rani Avula
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | | | - Preethi Chander
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Rao L Divi
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Johanna T Dwyer
- Office of Dietary Supplements, National Institutes of Health, Bethesda, MD
| | - Gallya Gannot
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | | | - Qian Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kyung Moon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mercy PrabhuDas
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anju Singh
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Mulualem E Tilahun
- National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Merriline M Satyamitra
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chiayeng Wang
- National Cancer Institute, National Institutes of Health, Rockville, MD
| | - Ronald Warren
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christina H Liu
- National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Kesika P, Thangaleela S, Sisubalan N, Radha A, Sivamaruthi BS, Chaiyasut C. The Role of the Nuclear Factor-Kappa B (NF-κB) Pathway in SARS-CoV-2 Infection. Pathogens 2024; 13:164. [PMID: 38392902 PMCID: PMC10892479 DOI: 10.3390/pathogens13020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
COVID-19 is a global health threat caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is associated with a significant increase in morbidity and mortality. The present review discusses nuclear factor-kappa B (NF-κB) activation and its potential therapeutical role in treating COVID-19. COVID-19 pathogenesis, the major NF-κB pathways, and the involvement of NF-κB in SARS-CoV-2 have been detailed. Specifically, NF-κB activation and its impact on managing COVID-19 has been discussed. As a central player in the immune and inflammatory responses, modulating NF-κB activation could offer a strategic avenue for managing SARS-CoV-2 infection. Understanding the NF-κB pathway's role could aid in developing treatments against SARS-CoV-2. Further investigations into the intricacies of NF-κB activation are required to reveal effective therapeutic strategies for managing and combating the SARS-CoV-2 infection and COVID-19.
Collapse
Affiliation(s)
- Periyanaina Kesika
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (N.S.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Subramanian Thangaleela
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Natarajan Sisubalan
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (N.S.)
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Arumugam Radha
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | | | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
49
|
Gawaz A, Schindler M, Hagelauer E, Blanchard G, Riel S, Vollert A, Gilliet M, Unterluggauer L, Stary G, Pospischil I, Hoetzenecker W, Fehrenbacher B, Schaller M, Guenova E, Forchhammer S. SARS-CoV-2-Induced Vasculitic Skin Lesions Are Associated with Massive Spike Protein Depositions in Autophagosomes. J Invest Dermatol 2024; 144:369-377.e4. [PMID: 37580012 DOI: 10.1016/j.jid.2023.07.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023]
Abstract
In patients infected with severe acute respiratory syndrome coronavirus 2, vasculopathic changes of the skin are associated with a severe prognosis. However, the pathogenesis of this vasculopathy is not conclusively clarified. In this study, 25 prospectively collected skin samples from patients with COVID-19-related skin lesions were examined for vasculopathic changes and, in case of vasculitis, were further analyzed with electron microscopy and immunohistochemistry. Vasculopathy was observed in 76% of all COVID-19-related inflammatory skin lesions. Visual endothelial changes without manifest leukocytoclastic vasculitis were found in 60% of the COVID-19-related skin lesions, whereas leukocytoclastic vasculitis was diagnosed in 16%. In the cases of vasculitis, there were extensive spike protein depositions in microvascular endothelial cells that colocalized with the autophagosome proteins LC3B and LC3C. The autophagy protein complex LC3-associated endocytosis in microvascular endothelial cells seems to be an important pathogenic factor for severe acute respiratory syndrome coronavirus 2-related vasculitis in the skin. On ultrastructural morphology, the vasculitic process was dominated by intracellular vesicle formation and endothelial cell disruption. Direct presence of severe acute respiratory syndrome coronavirus 2 particles in the skin was not observed. Therefore, our results suggest that instead of direct viral infection, dermal vasculitic lesions in COVID-19 are caused by severe acute respiratory syndrome coronavirus 2 spike protein deposition followed by endothelial damage with activation of autophagy.
Collapse
Affiliation(s)
- Andrea Gawaz
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Elena Hagelauer
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Gabriela Blanchard
- Department of Dermatology, Lausanne University Hospital (CHUV), Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Simon Riel
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Anneli Vollert
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Michel Gilliet
- Department of Dermatology, Lausanne University Hospital (CHUV), Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Isabella Pospischil
- Department of Dermatology, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | - Wolfram Hoetzenecker
- Department of Dermatology, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | | | - Martin Schaller
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Emmanuella Guenova
- Department of Dermatology, Lausanne University Hospital (CHUV), Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Department of Dermatology, Hospital 12 de Octubre, Medical school, University Complutense, Madrid, Spain.
| | | |
Collapse
|
50
|
Angiola F, Franchetti G, Cestonaro C, Agnolucci J, Giordano R, Viel G. Dying at home during the SARS-CoV-2 endemic: The importance of defining the exact mechanism of death. Leg Med (Tokyo) 2024; 66:102361. [PMID: 38039658 DOI: 10.1016/j.legalmed.2023.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/08/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023]
Abstract
INTRODUCTION Coronavirus Disease 2019 (COVID-19) has become endemic in Europe thanks to the presence of less deadly and more infectious variants and to the existence of a significant portion of unvaccinated people among the general population. SARS-Cov-2 related deaths are probably going to fade in the next years, but Covid-19 should still be considered a potential cause of death in the out-of-hospital setting in the next future. MATERIAL AND METHODS Three (3) cases of unexpected death at home are here presented. Each case has been investigated with the same methodological approach: death scene investigation (DSI), complete autopsy with histology, immunohistochemistry, RNA in situ hybridization for SARS-CoV-2 spike protein in lung tissue, toxicology and microbiology. RESULTS AND DISCUSSION All three cases had a COVID + post-mortem nasopharyngeal swab. Histology and immunohistochemistry revealed a SARS-CoV-2 lung involvement in only two of the cases (Cases 2 and 3), while a septic bacterial pneumonia was found in Case 1, where RNA-in situ hybridization for viral spike protein showed no reactivity in pneumocytes. The integration of all postmortem evidence allowed to attribute a different role of SARS-Cov-2 in the determinism of the death. CONCLUSION In the current post-pandemic context, SARS-CoV-2 remains a possible cause of death when investigating out-of-hospital unexpected deaths. Since a positive post-mortem swab does not automatically imply a COVID-19-related death, histology and immunohistochemistry are helpful for identifying SARS-CoV-2 lung involvement and, therefore, its potential active role in the determinism of death.
Collapse
Affiliation(s)
- Francesco Angiola
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Giorgia Franchetti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Clara Cestonaro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Jacopo Agnolucci
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Renzo Giordano
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Guido Viel
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy.
| |
Collapse
|