Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,028)

Search Parameters:
Keywords = COVID-19 lung infection

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1001 KiB  
Review
Old and New Definitions of Acute Respiratory Distress Syndrome (ARDS): An Overview of Practical Considerations and Clinical Implications
by Cesare Biuzzi, Elena Modica, Noemi De Filippis, Daria Pizzirani, Benedetta Galgani, Agnese Di Chiaro, Daniele Marianello, Federico Franchi, Fabio Silvio Taccone and Sabino Scolletta
Diagnostics 2025, 15(15), 1930; https://doi.org/10.3390/diagnostics15151930 - 31 Jul 2025
Viewed by 324
Abstract
Lower respiratory tract infections remain a leading cause of morbidity and mortality among Intensive Care Unit patients, with severe cases often progressing to acute respiratory distress syndrome (ARDS). This life-threatening syndrome results from alveolar–capillary membrane injury, causing refractory hypoxemia and respiratory failure. Early [...] Read more.
Lower respiratory tract infections remain a leading cause of morbidity and mortality among Intensive Care Unit patients, with severe cases often progressing to acute respiratory distress syndrome (ARDS). This life-threatening syndrome results from alveolar–capillary membrane injury, causing refractory hypoxemia and respiratory failure. Early detection and management are critical to treat the underlying cause, provide protective lung ventilation, and, eventually, improve patient outcomes. The 2012 Berlin definition standardized ARDS diagnosis but excluded patients on non-invasive ventilation (NIV) or high-flow nasal cannula (HFNC) modalities, which are increasingly used, especially after the COVID-19 pandemic. By excluding these patients, diagnostic delays can occur, risking the progression of lung injury despite ongoing support. Indeed, sustained, vigorous respiratory efforts under non-invasive modalities carry significant potential for patient self-inflicted lung injury (P-SILI), underscoring the need to broaden diagnostic criteria to encompass these increasingly common therapies. Recent proposals expand ARDS criteria to include NIV and HFNCs, lung ultrasound, and the SpO2/FiO2 ratio adaptations designed to improve diagnosis in resource-limited settings lacking arterial blood gases or advanced imaging. However, broader criteria risk overdiagnosis and create challenges in distinguishing ARDS from other causes of acute hypoxemic failure. Furthermore, inter-observer variability in imaging interpretation and inconsistencies in oxygenation assessment, particularly when relying on non-invasive measurements, may compromise diagnostic reliability. To overcome these limitations, a more nuanced diagnostic framework is needed—one that incorporates individualized therapeutic strategies, emphasizes lung-protective ventilation, and integrates advanced physiological or biomarker-based indicators like IL-6, IL-8, and IFN-γ, which are associated with worse outcomes. Such an approach has the potential to improve patient stratification, enable more targeted interventions, and ultimately support the design and conduct of more effective interventional studies. Full article
Show Figures

Figure 1

36 pages, 1583 KiB  
Review
SARS-CoV-2 Pneumonia: Advances in Diagnosis and Treatment
by Olga Adriana Caliman-Sturdza, Iuliana Soldanescu and Roxana Elena Gheorghita
Microorganisms 2025, 13(8), 1791; https://doi.org/10.3390/microorganisms13081791 - 31 Jul 2025
Viewed by 324
Abstract
The development of severe SARS-CoV-2 pneumonia is characterized by extensive lung inflammation, which, in turn, leads to respiratory distress and a decline in blood oxygen levels. Hospital admission, along with intensive care or ventilator usage, becomes necessary because this condition leads to serious [...] Read more.
The development of severe SARS-CoV-2 pneumonia is characterized by extensive lung inflammation, which, in turn, leads to respiratory distress and a decline in blood oxygen levels. Hospital admission, along with intensive care or ventilator usage, becomes necessary because this condition leads to serious respiratory problems. This review aims to provide a comprehensive overview of the pathophysiological mechanisms, diagnostic methods, and current therapeutic options for pneumonia caused by the SARS-CoV-2 virus. The pathophysiological process of severe pneumonia due to SARS-CoV-2 infection is characterized by direct lung damage from viral replication, an excessive immune system response, inflammation, impaired gas exchange, and multi-organ failure. The coexistence of various medical conditions leads to substantial lung impairment, resulting in hypoxia and respiratory failure, which can ultimately lead to fatal outcomes. The diagnosis of severe SARS-CoV-2 pneumonia is made through a combination of clinical, radiologic, and laboratory findings. A multifaceted approach integrating antiviral therapy, corticosteroids, oxygen supplementation, ventilatory management, and immunomodulation is imperative to control inflammation and enhance clinical outcomes. Early intervention, meticulous monitoring, and personalized care are paramount for enhancing survival and mitigating complications in critically ill patients with COVID-19 pneumonia. Full article
(This article belongs to the Special Issue Editorial Board Members’ Collection Series: SARS-CoV-2 and COVID-19)
Show Figures

Figure 1

24 pages, 3590 KiB  
Article
Mesocricetus auratus (Golden Syrian Hamster) Experimental Model of SARS-CoV-2 Infection Reveals That Lung Injury Is Associated with Phenotypic Differences Between SARS-CoV-2 Variants
by Daniela del Rosario Flores Rodrigues, Alexandre dos Santos da Silva, Arthur Daniel Rocha Alves, Bárbara Araujo Rossi, Richard de Almeida Lima, Sarah Beatriz Salvador Castro Faria, Oswaldo Gonçalves Cruz, Rodrigo Muller, Julio Scharfstein, Amanda Roberta Revoredo Vicentino, Aline da Rocha Matos, João Paulo Rodrigues dos Santos, Pedro Paulo Abreu Manso, Milla Bezerra Paiva, Debora Ferreira Barreto-Vieira, Gabriela Cardoso Caldas, Marcelo Pelajo Machado and Marcelo Alves Pinto
Viruses 2025, 17(8), 1048; https://doi.org/10.3390/v17081048 - 28 Jul 2025
Viewed by 470
Abstract
Despite the current level of public immunity to SARS-CoV-2, the early inflammatory events associated with respiratory distress in COVID-19 patients are not fully elucidated. Syrian golden hamsters, facultative hibernators, recapitulate the phenotype of SARS-CoV-2-induced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)—induced severe acute [...] Read more.
Despite the current level of public immunity to SARS-CoV-2, the early inflammatory events associated with respiratory distress in COVID-19 patients are not fully elucidated. Syrian golden hamsters, facultative hibernators, recapitulate the phenotype of SARS-CoV-2-induced severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)—induced severe acute lung injury seen in patients. In this study, we describe the predominance of the innate immune response in hamsters inoculated with four different SARS-CoV-2 variants, underscoring phenotypic differences among them. Severe inflammatory lung injury was chronologically associated with acute and significant weight loss, mainly in animals inoculated with A.2 and Delta variants. Omicron-infected animals had lower overall histopathology scores compared to other variants. We highlight the central role of endothelial injury and activation in the pathogenesis of experimental SARS-CoV-2 infection in hamsters, characterised by the presence of proliferative type I and type II pneumocytes with abundant surfactant expression, thereby maintaining hyperinflated alveolar fields. Additionally, there was evidence of intrapulmonary lymphatic vessel proliferation, which was accompanied by a lack of detectable microthrombosis in the lung parenchyma. However, white microthrombi were observed in lymphatic vessels. Our findings suggest that the physiological compensatory mechanisms that maintain respiratory homeostasis in Golden Syrian hamsters prevent severe respiratory distress and death after SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue Emerging Concepts in SARS-CoV-2 Biology and Pathology, 3rd Edition)
Show Figures

Figure 1

19 pages, 967 KiB  
Review
Hematologic and Immunologic Overlap Between COVID-19 and Idiopathic Pulmonary Fibrosis
by Gabriela Mara, Gheorghe Nini, Stefan Marian Frenț and Coralia Cotoraci
J. Clin. Med. 2025, 14(15), 5229; https://doi.org/10.3390/jcm14155229 - 24 Jul 2025
Viewed by 366
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral infections and chronic lung disease, this review offers a focused analysis of the shared hematologic and immunologic mechanisms between COVID-19 and IPF. Our aim is to better understand how SARS-CoV-2 infection may worsen disease progression in IPF and identify converging pathophysiological pathways that may inform clinical management. We conducted a narrative synthesis of the peer-reviewed literature from PubMed, Scopus, and Web of Science, focusing on clinical, experimental, and pathological studies addressing immune and coagulation abnormalities in both COVID-19 and IPF. Both diseases exhibit significant overlap in inflammatory and fibrotic signaling, particularly via the TGF-β, IL-6, and TNF-α pathways. COVID-19 amplifies coagulation disturbances and endothelial dysfunction already present in IPF, promoting microvascular thrombosis and acute exacerbations. Myeloid cell overactivation, impaired lymphocyte responses, and fibroblast proliferation are central to this shared pathophysiology. These synergistic mechanisms may accelerate fibrosis and increase mortality risk in IPF patients infected with SARS-CoV-2. This review proposes an integrative framework for understanding the hematologic and immunologic convergence of COVID-19 and IPF. Such insights are essential for refining therapeutic targets, improving prognostic stratification, and guiding early interventions in this high-risk population. Full article
(This article belongs to the Special Issue Chronic Lung Conditions: Integrative Approaches to Long-Term Care)
Show Figures

Figure 1

27 pages, 1201 KiB  
Review
Non-Viral Therapy in COVID-19: Where Are We Standing? How Our Experience with COVID May Help Us Develop Cell Therapies for Long COVID Patients
by Aitor Gonzaga, Gema Martinez-Navarrete, Loreto Macia, Marga Anton-Bonete, Gladys Cahuana, Juan R. Tejedo, Vanessa Zorrilla-Muñoz, Eduardo Fernandez-Jover, Etelvina Andreu, Cristina Eguizabal, Antonio Pérez-Martínez, Carlos Solano, Luis Manuel Hernández-Blasco and Bernat Soria
Biomedicines 2025, 13(8), 1801; https://doi.org/10.3390/biomedicines13081801 - 23 Jul 2025
Viewed by 467
Abstract
Objectives: COVID-19, caused by the SARS-CoV-2 virus, has infected over 777 million individuals and led to approximately 7 million deaths worldwide. Despite significant efforts to develop effective therapies, treatment remains largely supportive, especially for severe complications like acute respiratory distress syndrome (ARDS). [...] Read more.
Objectives: COVID-19, caused by the SARS-CoV-2 virus, has infected over 777 million individuals and led to approximately 7 million deaths worldwide. Despite significant efforts to develop effective therapies, treatment remains largely supportive, especially for severe complications like acute respiratory distress syndrome (ARDS). Numerous compounds from diverse pharmacological classes are currently undergoing preclinical and clinical evaluation, targeting both the virus and the host immune response. Methods: Despite the large number of articles published and after a preliminary attempt was published, we discarded the option of a systematic review. Instead, we have done a description of therapies with these results and a tentative mechanism of action. Results: Preliminary studies and early-phase clinical trials have demonstrated the potential of Mesenchymal Stem Cells (MSCs) in mitigating severe lung damage in COVID-19 patients. Previous research has shown MSCs to be effective in treating various pulmonary conditions, including acute lung injury, idiopathic pulmonary fibrosis, ARDS, asthma, chronic obstructive pulmonary disease, and lung cancer. Their ability to reduce inflammation and promote tissue repair supports their potential role in managing COVID-19-related complications. This review demonstrates the utility of MSCs in the acute phase of COVID-19 and postulates the etiopathogenic role of mitochondria in Long-COVID. Even more, their combination with other therapies is also analyzed. Conclusions: While the therapeutic application of MSCs in COVID-19 is still in early stages, emerging evidence suggests promising outcomes. As research advances, MSCs may become an integral part of treatment strategies for severe COVID-19, particularly in addressing immune-related lung injury and promoting recovery. However, a full pathogenic mechanism may explain or unify the complexity of signs and symptoms of Long COVID and Post-Acute Sequelae (PASC). Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

16 pages, 720 KiB  
Article
Demographic and Clinical Profile of Patients with Osteogenesis Imperfecta Hospitalized Due to Coronavirus Disease (COVID)-19: A Case Series of 13 Patients from Brazil
by Luana Lury Morikawa, Luiz Felipe Azevedo Marques, Adriele Evelyn Ferreira Silva, Patrícia Teixeira Costa, Lucas Silva Mello, Andrea de Melo Alexandre Fraga and Fernando Augusto Lima Marson
Healthcare 2025, 13(15), 1779; https://doi.org/10.3390/healthcare13151779 - 23 Jul 2025
Viewed by 265
Abstract
Background: Osteogenesis imperfecta (OI) is a rare genetic connective tissue disorder characterized by bone fragility, most often caused by pathogenic variants in type I collagen genes. In this context, we aimed to describe the clinical and epidemiological characteristics of patients with OI who [...] Read more.
Background: Osteogenesis imperfecta (OI) is a rare genetic connective tissue disorder characterized by bone fragility, most often caused by pathogenic variants in type I collagen genes. In this context, we aimed to describe the clinical and epidemiological characteristics of patients with OI who were hospitalized for coronavirus disease (COVID)-19 in Brazil between 2020 and 2024. Methods: We conducted a retrospective descriptive analysis using data from the Brazilian Unified Health System (SUS, which stands for the Portuguese Sistema Único de Saúde) through the Open-Data-SUS platform. Patients with a confirmed diagnosis of OI and hospitalization due to COVID-19 were included. Descriptive statistical analysis was performed to evaluate demographic, clinical, and outcome-related variables. We included all hospitalized COVID-19 cases with a confirmed diagnosis of OI between 2020 and 2024. Results: Thirteen hospitalized patients with OI and COVID-19 were identified. Most were adults (9; 69.2%), male (7; 53.8%), self-identified as White (9; 69.2%), and all were residents of urban areas (13; 100.0%). The most frequent symptoms were fever (10; 76.9%), cough (9; 69.2%), oxygen desaturation (9; 69.2%), dyspnea (8; 61.5%), and respiratory distress (7; 53.8%). Two patients had heart disease, one had chronic lung disease, and one was obese. As for vaccination status, five patients (38.5%) had been vaccinated against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Four patients (30.8%) required admission to an intensive care unit (ICU), and six (46.2%) required noninvasive ventilatory support. Among those admitted to the ICU, only two required invasive mechanical ventilation. The clinical outcome was death in two cases (15.4%). Both patients were male, White, and had not been vaccinated against SARS-CoV-2. One was 47 years old, was not admitted to the ICU, but required noninvasive ventilation. Despite the underlying condition most patients had favorable outcomes, consistent with an international report. Conclusions: This is the first report to describe the clinical and epidemiological profile of patients with OI hospitalized for COVID-19 in Brazil, providing initial insights into how a rare bone disorder intersects with an acute respiratory infection. The generally favorable outcomes observed—despite the underlying skeletal fragility—suggest that individuals with OI are not necessarily at disproportionate risk of severe COVID-19, particularly when appropriately monitored. The occurrence of deaths only among unvaccinated patients underscores the critical role of SARS-CoV-2 vaccination in this population. Although pharmacological treatment data were unavailable, the potential protective effects of bisphosphonates and vitamin D merit further exploration. These findings support the need for early preventive strategies, systematic vaccination efforts, and dedicated clinical protocols for rare disease populations during infectious disease outbreaks. Full article
Show Figures

Figure 1

23 pages, 2202 KiB  
Article
Afucosylated IgG Promote Thrombosis in Mouse Injected with SARS-CoV-2 Spike Expressing Megakaryocytes
by Meryem Mabrouk, Farah Atifi, Hicham Wahnou, Afaf Allaoui, Nabil Zaid, Abdallah Naya, Ejaife O. Agbani, Loubna Khalki, Meriem Khyatti, Youssef Tijani, Khadija Akarid, Damien Arnoult, Haissam Abou-Saleh, Othman El Faqer, Salma Labied, Mounia Ammara, Fadila Guessous, Farid Jalali and Younes Zaid
Int. J. Mol. Sci. 2025, 26(14), 7002; https://doi.org/10.3390/ijms26147002 - 21 Jul 2025
Viewed by 531
Abstract
Despite the prevalence of fucosylated IgG in plasma, specific IgGs with low core fucosylation sporadically emerge in response to virus infections and blood cell alloantigens. This low fucosylation of IgG is implicated in the pathogenesis of SARS-CoV-2 and dengue infections. In COVID-19, the [...] Read more.
Despite the prevalence of fucosylated IgG in plasma, specific IgGs with low core fucosylation sporadically emerge in response to virus infections and blood cell alloantigens. This low fucosylation of IgG is implicated in the pathogenesis of SARS-CoV-2 and dengue infections. In COVID-19, the presence of IgGs with low core fucosylation (afucosylated IgGs) targeting spike protein predicts disease progression to a severe form and actively mediates this progression. This study reveals that SARS-CoV-2 infection of megakaryocytes promotes the generation of pathogenic afucosylated anti-spike IgGs, leading to outcomes, such as pulmonary vascular thrombosis, acute lung injury, and mortality in FcγRIIa-transgenic mice. Platelets from mice injected with virus-infected human megakaryocytes express significant activation biomarkers, indicating a direct link between the immune response and platelet activation. Mice injected with virus-infected human megakaryocytes demonstrate an elevated rate of thrombus formation induced by FeCl3 (4%) and a reduction in bleeding time, emphasizing the intricate interplay of viral infection, immune response, and hemostatic complications. Treatment with inhibitors targeting FcγRIIa, serotonin, or complement anaphylatoxins of mice injected with spike-expressing MKs successfully prevents observed platelet activation, thrombus formation, and bleeding abnormalities, offering potential therapeutic strategies for managing severe outcomes associated with afucosylated IgGs in COVID-19 and related disorders. Full article
(This article belongs to the Special Issue The Molecular Role of Platelets in Human Diseases)
Show Figures

Figure 1

19 pages, 2517 KiB  
Article
In Silico Analysis of Post-COVID-19 Condition (PCC) Associated SNP rs9367106 Predicts the Molecular Basis of Abnormalities in the Lungs and Brain Functions
by Amit K. Maiti
Int. J. Mol. Sci. 2025, 26(14), 6680; https://doi.org/10.3390/ijms26146680 - 11 Jul 2025
Viewed by 454
Abstract
Long- or post-COVID-19 syndrome, which is also designated by WHO as Post COVID-19 Condition (PCC), is characterized by the persistent symptoms that remain after recovery from SARS-CoV-2 infection. A worldwide consortium of Long COVID-19 Host Genetics Initiative (Long COVID-19 HGI) identified an SNP [...] Read more.
Long- or post-COVID-19 syndrome, which is also designated by WHO as Post COVID-19 Condition (PCC), is characterized by the persistent symptoms that remain after recovery from SARS-CoV-2 infection. A worldwide consortium of Long COVID-19 Host Genetics Initiative (Long COVID-19 HGI) identified an SNP rs9367106 (G>C; chr6:41,515,652, GRCh38, p = 1.76 × 10−10, OR = 1.63, 95% CI: 1.40–1.89) that is associated with PCC. Unraveling the functional significance of this SNP is of prime importance to understanding the development of the PCC phenotypes and their therapy. Here, in Silico, I explored how the risk allele of this SNP alters the functional mechanisms and molecular pathways leading to the development of PCC phenotypes. Bioinformatic methods include physical interactions using HI-C and Chia-PET analysis, Transcription Factors (TFs) binding ability, RNA structure modeling, epigenetic, and pathway analysis. This SNP resides within two long RNA genes, LINC01276 and FOXP4-AS1, and is located at ~31 kb upstream of a transcription factor FOXP4. This DNA region, including this SNP, physically interacts with FOXP4-AS1 and FOXP4, implying that this regulatory SNP could alter the normal cellular function of FOXP4-AS1 and FOXP4. Furthermore, rs9367106 is in eQTL with the FOXP4 gene in lung tissue. rs9367106 carrying DNA sequences act as distant enhancers and bind with several transcription factors (TFs) including YY1, PPAR-α, IK-1, GR-α, and AP2αA. The G>C transition extensively modifies the RNA structure that may affect the TF bindings and enhancer functions to alter the interactions and functions of these RNA molecules. This SNP also includes an ALU/SINE sequence and alteration of which by the G>C transition may prevent IFIH1/MDA5 activation, leading to suppression of host innate immune responses. LINC01276 targets the MED20 gene that expresses mostly in brain tissues, associated with sleep disorders and basal ganglia abnormalities similar to some of the symptoms of PCC phenotypes. Taken together, G>C transition of rs9367601 may likely alter the function of all three genes to explain the molecular basis of developing the long-term symptomatic abnormalities in the lungs and brain observed after COVID-19 recovery. Full article
(This article belongs to the Special Issue Genetic Variations in Human Diseases: 2nd Edition)
Show Figures

Figure 1

25 pages, 2374 KiB  
Article
Pulmonary Embolism in COVID-19: Trends from a Single-Center Study Across Ten Pandemic Waves in Romania
by Alexandra Herlo, Adelina Raluca Marinescu, Talida Georgiana Cut, Ruxandra Laza, Adina Maria Marza, Claudia Raluca Balasa Virzob, Cristian Iulian Oancea, Lucian-Flavius Herlo, Ioana-Melinda Luput-Andrica and Voichita Elena Lazureanu
Microorganisms 2025, 13(7), 1634; https://doi.org/10.3390/microorganisms13071634 - 10 Jul 2025
Viewed by 325
Abstract
The COVID-19 pandemic has significantly impacted global health, with pulmonary embolism (PE) emerging as a critical complication due to the hypercoagulable state induced by SARS-CoV-2 infection. Despite advancements in prevention and treatment, PE remains a major cause of morbidity and mortality in COVID-19 [...] Read more.
The COVID-19 pandemic has significantly impacted global health, with pulmonary embolism (PE) emerging as a critical complication due to the hypercoagulable state induced by SARS-CoV-2 infection. Despite advancements in prevention and treatment, PE remains a major cause of morbidity and mortality in COVID-19 patients. This study analyzes the trends, outcomes, and contributing factors of PE across ten pandemic waves in Romania, highlighting the evolving clinical burden and management approaches. This retrospective observational study was conducted on confirmed COVID-19 patients that also developed PE, who were admitted to “Victor Babeș” Hospital and Municipal Emergency Hospital in Timișoara, Romania. Data on demographics, clinical features, inflammatory markers, comorbidities, and treatment were collected from medical records. Statistical analyses, including ANOVA and Kaplan–Meier survival analysis, were conducted to evaluate trends and survival outcomes over time. The study included 166 patients, with a mean age of 67.26 ± 13.57 years. Mortality peaked at 50% in Wave 1, declined to 12% in Wave 7, and increased again to 28% in Wave 10. Intubation rates varied, with a high of 29% in Wave 6 and a low of 12% in Wave 8. Lung involvement was the most severe in Wave 4 (mean 0.54 ± 0.18) but improved in later waves, reaching a mean of 0.24 ± 0.12 in Wave 8. This study highlights the dynamic trends in PE during the COVID-19 pandemic in Romania. Improved clinical management, vaccination, and adaptive healthcare strategies contributed to better outcomes in later waves. Full article
(This article belongs to the Special Issue Infectious Disease Surveillance in Romania)
Show Figures

Figure 1

29 pages, 14985 KiB  
Article
Spatiotemporal Characterization of Changes in the Respiratory Tract and the Nervous System, Including the Eyes in SARS-CoV-2-Infected K18-hACE2 Mice
by Malgorzata Rosiak, Tom Schreiner, Georg Beythien, Eva Leitzen, Anastasiya Ulianytska, Lisa Allnoch, Kathrin Becker, Lukas M. Michaely, Sandra Lockow, Sabrina Clever, Christian Meyer zu Natrup, Asisa Volz, Wolfgang Baumgärtner, Malgorzata Ciurkiewicz, Kirsten Hülskötter and Katharina M. Gregor
Viruses 2025, 17(7), 963; https://doi.org/10.3390/v17070963 - 9 Jul 2025
Viewed by 554
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is known to affect multiple organ systems, including the respiratory tract and nervous and ocular systems. This retrospective study aimed to characterize the spatiotemporal distribution of viral antigen [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is known to affect multiple organ systems, including the respiratory tract and nervous and ocular systems. This retrospective study aimed to characterize the spatiotemporal distribution of viral antigen and associated pathological changes in the nose, lungs, brain, and eyes of K18-hACE2 mice intranasally infected with SARS-CoV-2. Using histology and immunohistochemistry, tissues were examined at 3, 6, and 7/8 days post-infection (dpi). In addition, lung and brain tissues were analyzed by means of RT-qPCR to determine viral RNA titers. Viral antigen was most pronounced in the nose, brain, and lung at 3, 6, and 7/8 dpi, respectively, whereas viral antigen was detected at 6 and 7/8 dpi in the retina. Quantitative PCR confirmed increasing viral RNA levels in both lung and brain, peaking at 7/8 dpi. Nasal and lung inflammation mirrored viral antigen distribution and localization. In the brain, the predominantly basal viral spread correlated with lymphohistiocytic meningoencephalitis, neuronal vacuolation, and altered neurofilament immunoreactivity. Retinal ganglion cells showed viral antigen expression without associated lesions. Microglial activation was evident in both the optic chiasm and the brain. These findings highlight the K18-hACE2 model’s utility for studying extrapulmonary SARS-CoV-2 pathogenesis. Understanding the temporal and spatial dynamics of viral spread enhances insights into SARS-CoV-2 neurotropism and its clinical manifestations. Full article
(This article belongs to the Section Coronaviruses)
Show Figures

Graphical abstract

18 pages, 24095 KiB  
Article
Genome-Wide Association Study of COVID-19 Breakthrough Infections and Genetic Overlap with Other Diseases: A Study of the UK Biobank
by Yaning Feng, Kenneth Chi-Yin Wong, Wai Kai Tsui, Ruoyu Zhang, Yong Xiang and Hon-Cheong So
Int. J. Mol. Sci. 2025, 26(13), 6441; https://doi.org/10.3390/ijms26136441 - 4 Jul 2025
Viewed by 483
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to substantial health and financial burdens worldwide, and vaccines provide hope for reducing the burden of this pandemic. However, vaccinated people remain at risk for SARS-CoV-2 infection. Genome-wide association studies (GWASs) may identify potential genetic [...] Read more.
The coronavirus disease 2019 (COVID-19) pandemic has led to substantial health and financial burdens worldwide, and vaccines provide hope for reducing the burden of this pandemic. However, vaccinated people remain at risk for SARS-CoV-2 infection. Genome-wide association studies (GWASs) may identify potential genetic factors involved in the development of COVID-19 breakthrough infections (BIs); however, very few or no GWASs have been conducted for COVID-19 BI thus far. We conducted a GWAS and detailed bioinformatics analysis on COVID-19 BIs in a European population via the UK Biobank (UKBB). We conducted a series of analyses at different levels, including SNP-based, gene-based, pathway, and transcriptome-wide association analyses, to investigate genetic factors associated with COVID-19 BIs and hospitalized infections. The polygenic risk score (PRS) and Hoeffding’s test were performed to reveal the genetic relationships between BIs and other medical conditions. Two independent loci (LD-clumped at r2 = 0.01) reached genome-wide significance (p < 5 × 10−8), including rs36170929, which mapped to LOC102725191/VWDE, and rs28645263, which mapped to RETREG1. A pathway enrichment analysis highlighted pathways such as viral myocarditis, Rho-selective guanine exchange factor AKAP13 signaling, and lipid metabolism. The PRS analyses revealed significant genetic overlap between COVID-19 BIs and heart failure and between HbA1c and type 1 diabetes. Genetic dependence was also observed between COVID-19 BIs and asthma, lung abnormalities, schizophrenia, and type 1 diabetes on the basis of Hoeffding’s test. This GWAS revealed two significant loci that may be associated with COVID-19 BIs and a number of genes and pathways that may be involved in BIs. Genetic overlap with other diseases was identified. Further studies are warranted to replicate these findings and elucidate the mechanisms involved. Full article
Show Figures

Figure 1

16 pages, 2743 KiB  
Article
Evidence Generation for a Host-Response Biosignature of Respiratory Disease
by Kelly E. Dooley, Michael Morimoto, Piotr Kaszuba, Margaret Krasne, Gigi Liu, Edward Fuchs, Peter Rexelius, Jerry Swan, Krzysztof Krawiec, Kevin Hammond, Stuart C. Ray, Ryan Hafen, Andreas Schuh and Nelson L. Shasha Jumbe
Viruses 2025, 17(7), 943; https://doi.org/10.3390/v17070943 - 2 Jul 2025
Viewed by 536
Abstract
Background: In just twenty years, three dangerous human coronaviruses—SARS-CoV, MERS-CoV, and SARS-CoV-2 have exposed critical gaps in early detection of emerging viral threats. Current diagnostics remain pathogen-focused, often missing the earliest phase of infection. A virus-agnostic, host-based diagnostic capable of detecting responses to [...] Read more.
Background: In just twenty years, three dangerous human coronaviruses—SARS-CoV, MERS-CoV, and SARS-CoV-2 have exposed critical gaps in early detection of emerging viral threats. Current diagnostics remain pathogen-focused, often missing the earliest phase of infection. A virus-agnostic, host-based diagnostic capable of detecting responses to viral intrusion is urgently needed. Methods: We hypothesized that the lungs act as biomechanical instruments, with infection altering tissue tension, wave propagation, and flow dynamics in ways detectable through subaudible vibroacoustic signals. In a matched case–control study, we enrolled 19 RT-PCR-confirmed COVID-19 inpatients and 16 matched controls across two Johns Hopkins hospitals. Multimodal data were collected, including passive vibroacoustic auscultation, lung ultrasound, peak expiratory flow, and laboratory markers. Machine learning models were trained to identify host-response biosignatures from anterior chest recordings. Results: 19 COVID-19 inpatients and 16 matched controls (mean BMI 32.4 kg/m2, mean age 48.6 years) were successfully enrolled to the study. The top-performing, unoptimized, vibroacoustic-only model achieved an AUC of 0.84 (95% CI: 0.67–0.92). The host-covariate optimized model achieved an AUC of 1.0 (95% CI: 0.94–1.0), with 100% sensitivity (95% CI: 82–100%) and 99.6% specificity (95% CI: 85–100%). Vibroacoustic data from the anterior chest alone reliably distinguished COVID-19 cases from controls. Conclusions: This proof-of-concept study demonstrates that passive, noninvasive vibroacoustic biosignatures can detect host response to viral infection in a hospitalized population and supports further testing of this modality in broader populations. These findings support the development of scalable, host-based diagnostics to enable early, agnostic detection of future pandemic threats (ClinicalTrials.gov number: NCT04556149). Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

18 pages, 797 KiB  
Review
Impact of Invasive Mechanical Ventilation on the Lung Microbiome
by Jose Luis Estela-Zape, Valeria Sanclemente-Cardoza, Maria Alejandra Espinosa-Cifuentes and Leidy Tatiana Ordoñez-Mora
Adv. Respir. Med. 2025, 93(4), 23; https://doi.org/10.3390/arm93040023 - 1 Jul 2025
Viewed by 575
Abstract
The lung microbiota is integral to maintaining microenvironmental homeostasis, influencing immune regulation, host defense against pathogens, and overall respiratory health. The dynamic interplay among the lung microbiota emphasizes their significance in shaping the respiratory milieu and potential impact on diverse pulmonary affections. This [...] Read more.
The lung microbiota is integral to maintaining microenvironmental homeostasis, influencing immune regulation, host defense against pathogens, and overall respiratory health. The dynamic interplay among the lung microbiota emphasizes their significance in shaping the respiratory milieu and potential impact on diverse pulmonary affections. This investigation aimed to identify the effects of invasive mechanical ventilation on the lung microbiome. Materials and Methods: A systematic review was conducted with registration number CRD42023461618, based on a search of PubMed, SCOPUS, and Web of Science databases, in line with the PRISMA guidelines. To achieve this, “(mechanical ventilation) AND (microbiota)” was used as the search term, replicable across all databases. The closing date of the search was 12 March 2025, and the evidence was scored using the MINORS scale. Results: A total of 16 studies were included, with patients aged 13.6 months to 76 years, predominantly male (64.2%). Common ICU admission diagnoses requiring invasive mechanical ventilation (IMV) included pneumonia, acute respiratory failure, and COVID-19. IMV was associated with reduced lung microbiota diversity and an increased prevalence of pathogenic bacteria, including Prevotella, Streptococcus, Staphylococcus, Pseudomonas, and Acinetobacter. The most frequently used antibiotics were cephalosporins, aminoglycosides, and penicillins. IMV-induced pulmonary dysbiosis correlated with higher infection risk and mortality, particularly in pneumonia and COVID-19 cases. Factors such as antimicrobial therapy, enteral nutrition, and systemic inflammation contributed to these alterations. Conclusions: Invasive mechanical ventilation has been associated with the development of alterations in the respiratory microbiome, resulting in reduced diversity of lung microorganisms. Full article
Show Figures

Figure 1

15 pages, 539 KiB  
Article
Effect of COVID-19 Disease on Serum Vitamin D Status in Children with Asthma—A Retrospective Study
by Jaqueline Abdul-Razzak, Mihaela Ionescu, Radu Diaconu, Alexandru Dan Popescu, Elena Carmen Niculescu, Ileana Octavia Petrescu, Cristina Elena Singer, Lucrețiu Radu, Liliana Anghelina and Cristian Gheonea
J. Clin. Med. 2025, 14(13), 4525; https://doi.org/10.3390/jcm14134525 - 26 Jun 2025
Viewed by 466
Abstract
Background/Objectives: Vitamin D is known to decrease the risk of contracting respiratory infections and developing exacerbations for children with asthma. This research evaluates the alterations in serum vitamin D concentrations and examines lung function in children with asthma, as indicated by clinical [...] Read more.
Background/Objectives: Vitamin D is known to decrease the risk of contracting respiratory infections and developing exacerbations for children with asthma. This research evaluates the alterations in serum vitamin D concentrations and examines lung function in children with asthma, as indicated by clinical symptoms and paraclinical results, after experiencing SARS-CoV-2 infection or other acute respiratory infections. Material and Method: This retrospective study included 145 children with asthma. For each patient, the following variables were acquired: demographic data, serum vitamin D levels, GINA asthma control levels, the fraction of exhaled nitric oxide (FeNO), pulmonary function tests parameters, data related to allergies, and the presence of exacerbations. Children were divided into two groups, according to the presence or absence of SARS-CoV-2 infection or other acute respiratory infections. Variables were statistically processed in SPSS. Results: In total, 93 children with asthma with SARS-CoV-2 infection or other acute respiratory infections and 52 children with asthma without SARS-CoV-2 infection or other acute respiratory infections were included in the study. Median serum vitamin D values were statistically significantly lower in children with a variable airflow limitation, compared to children with normal values (p = 0.004), as well as for children with partially controlled asthma, relative to children with well controlled asthma (p < 0.0005). Similarly, children with acute respiratory infections/COVID-19 disease had lower median values of serum vitamin D, compared to children without acute respiratory infections/COVID-19 disease (p < 0.0005). A decrease in serum vitamin D value was statistically significantly associated with an increase in FeNO value for children with asthma with COVID-19 disease (p = 0.027), as well as for the entire study group (p < 0.0005). Conclusions: Children with asthma who had acute respiratory infections, including COVID-19 disease, showed considerably reduced serum vitamin D levels and were linked to more significant airflow limitation, reduced asthma control and elevated airway inflammation, suggesting its potential role in influencing asthma severity and infection response. Full article
(This article belongs to the Section Clinical Pediatrics)
Show Figures

Figure 1

16 pages, 2408 KiB  
Article
Bacteriome Signature in SARS-CoV-2-Infected Patients Correlates with Increased Gut Permeability and Systemic Inflammatory Cytokines
by Larissa S. Souza, Alexandre S. Ferreira-Junior, Pedro C. Estella, Ricardo K. Noda, Lhorena F. Sousa, Miguel T. Y. Murata, Lucas A. L. Carvalho, João L. Brisotti, Daniel G. Pinheiro, Josias Rodrigues, Carlos M. C. B. Fortaleza and Gislane L. V. de Oliveira
Microorganisms 2025, 13(6), 1407; https://doi.org/10.3390/microorganisms13061407 - 16 Jun 2025
Viewed by 693
Abstract
The COVID-19 pandemic has highlighted the complex interplay between the gut microbiota and systemic immune responses, particularly through the gut–lung axis. Disruptions in gut microbial diversity and function—commonly referred to as dysbiosis—have been increasingly implicated in the pathogenesis of SARS-CoV-2 infection. In this [...] Read more.
The COVID-19 pandemic has highlighted the complex interplay between the gut microbiota and systemic immune responses, particularly through the gut–lung axis. Disruptions in gut microbial diversity and function—commonly referred to as dysbiosis—have been increasingly implicated in the pathogenesis of SARS-CoV-2 infection. In this study, we assessed the gut bacteriome and permeability in SARS-CoV-2-infected patients using 16S sequencing and ELISA assays, respectively. We also measured blood inflammatory cytokines and fecal secretory IgA to evaluate systemic and mucosal immune responses. Significant alterations in both alpha and beta diversity metrics were observed in patients with COVID-19 (n = 79) and those with post-COVID-19 condition (n = 141) compared to the controls (n = 97). Differential abundance and taxonomic analyses revealed distinct microbial profiles in the infected groups. Increased plasma levels of IL-2, IL-6, IL-17A, IFN-γ, and zonulin were detected in patient samples. Some genera were elevated during acute infection, which was positively correlated with C-reactive protein, while Enterobacteriaceae and Escherichia-Shigella were associated with increased zonulin levels, indicating compromised intestinal barrier function. These findings suggest that gut dysbiosis may contribute to bacterial translocation and systemic inflammation. Overall, our results highlight the importance of the gut–lung axis and suggest that modulating the gut microbiota could support immune regulation in SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue Correlations Between the Gastrointestinal Microbiome and Diseases)
Show Figures

Figure 1

Back to TopTop