1
|
Zhang P, Yang Q, Xu X, Feng H, Du B, Xu J, Liu B, Mu X, Wang J, Tong Z. Fluorescence excitation-emission matrix spectroscopy combined with machine learning for the classification of viruses for respiratory infections. Talanta 2025; 286:127462. [PMID: 39753074 DOI: 10.1016/j.talanta.2024.127462] [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: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 03/03/2025]
Abstract
Significant efforts were currently being made worldwide to develop a tool capable of distinguishing between various harmful viruses through simple analysis. In this study, we utilized fluorescence excitation-emission matrix (EEM) spectroscopy as a rapid and specific tool with high sensitivity, employing a straightforward methodological approach to identify spectral differences between samples of respiratory infection viruses. To achieve this goal, the fluorescence EEM spectral data from eight virus samples was divided into training and test sets, which were then analyzed using random forest and support vector machine classification models. We proposed a novel strategy for data fusion based on fast Fourier transform (FFT) and wavelet transform (WT) methods, which significantly enhanced classification accuracy from 45 % to 75 %. This approach improved the classification capability for similar spectral characteristics of viruses. Rhinovirus was further differentiated from rotavirus, while influenza A virus was distinguished from inactivated poliovirus vaccines and rhinovirus. This study demonstrated that the integration of fluorescence EEM spectroscopy with machine learning algorithms presented significant potential for the detection of unidentified harmful substances in the ambient environment.
Collapse
Affiliation(s)
- Pengjie Zhang
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Qianyu Yang
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Xinrui Xu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Huiping Feng
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Bin Du
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Jiwei Xu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Bing Liu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Xihui Mu
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China
| | - Jiang Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China.
| | - Zhaoyang Tong
- State Key Laboratory of NBC Protection for Civilian, Beijing, 102205, China.
| |
Collapse
|
2
|
Heeralall C, Ibrahim UH, Jenneker M, Singh S, Lazarus L, Mackraj I. Effect of COVID-19 Infection During Pregnancy on the Plasma/Extracellular Vesicles Pro-Inflammatory Cytokine Profile. Am J Reprod Immunol 2025; 93:e70071. [PMID: 40198239 PMCID: PMC11977858 DOI: 10.1111/aji.70071] [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: 09/02/2024] [Revised: 02/19/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
PURPOSE The Coronavirus disease (COVID-19) has impacted pregnant women significantly, with increased mortality and morbidity. The implications of this virus are linked to extracellular vesicles (EVs) and maternal inflammation due to the cytokine storm. Hence, this study aims to investigate the impact of COVID-19 on the pro-inflammatory cytokine profile in both plasma and EVs of South African pregnant women. METHODS Plasma samples were obtained from pregnant women in the third trimester, from which EVs were extracted using the Invitrogen Total Exosome Isolation Kit. These plasma-derived EVs were characterised using transmission electron microscopy and nanoparticle tracking analysis (NTA). RESULTS COVID-19 infection in pregnancy did not significantly affect the average particle size and concentration of isolated EVs. The levels of IFN gamma, IL-6, MIP-1 alpha and TNF alpha were analysed in the plasma and circulating EVs through a multiplex assay. Compared to the control group, a significant increase in IL-6, IFN-γ, TNF-α and MIP-1α levels were observed in both plasma and EVs content of COVID-19 pregnancies. CONCLUSION These findings suggest that COVID-19 infection impacts the pro-inflammatory cytokine profile in the plasma and EVs of South African pregnant women.
Collapse
Affiliation(s)
- C. Heeralall
- Discipline of Clinical AnatomySchool of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Usri H. Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - M. Jenneker
- Discipline of Obstetrics and Gynaecology, School of Clinical MedicineUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - S. Singh
- Optics & Imaging Centre, Doris Duke Medical Research InstituteCollege of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - L. Lazarus
- Discipline of Clinical AnatomySchool of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| | - Irene Mackraj
- Discipline of Human Physiology, School of Laboratory Medicine and Medical SciencesCollege of Health SciencesUniversity of KwaZulu‐NatalDurbanSouth Africa
| |
Collapse
|
3
|
Jensen LR, Posa M, Filipp S, Kelly M, Otero J. Administration of COVID-19 vaccination to adult caregivers in pediatric outpatient clinics: a vaccination cocooning project. Postgrad Med 2025:1-7. [PMID: 40133217 DOI: 10.1080/00325481.2025.2482513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Vaccination against COVID-19 is safe for patients greater than 6 months of age and reduces virus infectivity and severity. One strategy for protecting children unable to be vaccinated is cocooning, a concept that protects at-risk individuals by vaccinating adult caregivers (ACs). This study evaluates AC characteristics of those receiving COVID-19 vaccination in pediatric healthcare clinics. METHODS This study is a retrospective chart review that identified ACs greater than 21 years of age who received a COVID-19 vaccine at one of six UFHealth pediatric offices between March 2021-April 2022. Investigators collected demographic information of the AC and corresponding child. RESULTS The AC sample was predominantly female (79.8%), parents (75.3% mothers, 19.3% fathers), White (48.2%) or Black/African American (33.7%). Those who completed both primary doses had higher rates of children less than 5 years compared to those who did not (48.2%, 37.2%; p-value: 0.0225). ACs completing both primary doses were also younger compared to their counterparts (37.0, 39.9 years; p-value: 0.0017), with double the rate of government-funded insurance (42.9%, 20.9%; p-value: <0.0001), and significantly different racial/ethnic distribution (p-value 0.0021). CONCLUSIONS ACs who completed both primary doses were younger females and had higher rates of children under 5 years old. We believe these results reflect the convenience of offering AC vaccines at pediatric healthcare clinics because of the high frequency of well-child checks within the first 5 years of life. This cohort also had double the rate of government-funded insurance, attributable to the free administration of COVID-19 vaccines at UF-affiliated pediatric clinics.
Collapse
Affiliation(s)
| | - Molly Posa
- University of Florida College of Medicine, Gainesville, FL, USA
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Stephanie Filipp
- University of Florida College of Medicine, Gainesville, FL, USA
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Maria Kelly
- University of Florida College of Medicine, Gainesville, FL, USA
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jaclyn Otero
- University of Florida College of Medicine, Gainesville, FL, USA
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
4
|
Jarneborn A, Kopparapu PK, Jin T. The Dual-Edged Sword: Risks and Benefits of JAK Inhibitors in Infections. Pathogens 2025; 14:324. [PMID: 40333091 DOI: 10.3390/pathogens14040324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025] Open
Abstract
Janus kinase inhibitors (JAKis) represent a relatively new class of immunomodulatory drugs with potent effects on various cytokine signalling pathways. They have revolutionized the treatment landscape for autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, and ulcerative colitis. However, their ability to modulate immune responses presents a dual-edged nature, influencing both protective immunity and pathological inflammation. This review explores the complex role of JAKis in infectious settings, highlighting both beneficial and detrimental effects. On the one hand, experimental models suggest that JAK inhibition can impair host defence mechanisms, increasing susceptibility to certain bacterial and viral infections. For example, tofacitinib-treated mice exhibited more severe joint erosions in Staphylococcus aureus (S. aureus) septic arthritis and showed impaired viral clearance in herpes simplex encephalitis. Additionally, clinical data confirm an increased risk of herpes zoster in patients receiving JAKis, underscoring the need for rigorous monitoring. On the other hand, JAK inhibition has demonstrated protective effects in certain infectious and hyperinflammatory conditions. In sepsis models, including cecal ligation and puncture (CLP) and S. aureus bacteraemia, tofacitinib improved survival by attenuating excessive inflammation. Furthermore, JAKis, particularly baricitinib, have shown substantial efficacy in mitigating cytokine storms during severe COVID-19 infections, leading to improved clinical outcomes and reduced mortality. These observations suggest that JAKis have a role in modulating hyperinflammatory responses in select infectious contexts. In conclusion, JAKis present a complex interplay between immunosuppression and immunomodulation. While they increase the risk of certain infections, they also show potential in managing hyperinflammatory conditions such as cytokine storms. The key challenge is determining which patients and situations benefit most from JAKis while minimizing risks, requiring a careful and personalized treatment approach.
Collapse
Affiliation(s)
- Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, 413 46 Gothenburg, Sweden
| |
Collapse
|
5
|
dos S. P. Andrade AC, Lacasse E, Dubuc I, Gudimard L, Gravel A, Puhm F, Campolina-Silva G, Queiroz-Junior C, Allaeys I, Prunier J, Azeggouar Wallen O, Dumais É, Belleannée C, Droit A, Flamand N, Boilard É, Flamand L. Deficiency in platelet 12-lipoxygenase exacerbates inflammation and disease severity during SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2025; 122:e2420441122. [PMID: 40100623 PMCID: PMC11962506 DOI: 10.1073/pnas.2420441122] [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: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 03/20/2025] Open
Abstract
Platelets, known for maintaining blood balance, also participate in antimicrobial defense. Upon severeacute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, platelets become hyperactivated, releasing molecules such as cytokines, granule contents, and bioactive lipids. The key effector biolipids produced by platelets include 12-hydroxyeicosatetraenoic acid (12-HETE) and 12-hydroxyeicosatrienoic acid (12-HETrE), produced by 12-lipoxygenase (12-LOX), and prostaglandins and thromboxane, produced by cyclooxygenase-1. While prostaglandin E2 and thromboxane B2 were previously associated with lung inflammation in severe COVID-19, the role of platelet 12-LOX in SARS-CoV-2 infection remains unclear. Using mice deficient for platelets' 12-LOX, we report that SARS-CoV-2 infection resulted in higher lung inflammation characterized by histopathological tissue analysis, increased leukocyte infiltrates, and cytokine production relative to wild-type mice. In addition, distinct platelet and lung transcriptomic changes, including alterations in NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) inflammasome-related gene expression, were observed. Mass spectrometry lipidomic analysis in 12-LOX-deficient-infected mice revealed significant changes in bioactive lipid content, including reduced levels of 12-HETrE that inversely correlated with disease severity. Finally, platelet 12-LOX deficiency was associated with increased morbidity and lower survival rates relative to wild type (WT) mice. Overall, this study highlights the complex interplay between 12-LOX-related lipid metabolism and inflammatory responses during SARS-CoV-2 infection. The findings provide valuable insights into potential therapeutic targets aimed at mitigating severe outcomes, emphasizing the pivotal role of platelet enzymes in the host response to viral infections.
Collapse
Affiliation(s)
- Ana Claudia dos S. P. Andrade
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Emile Lacasse
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Isabelle Dubuc
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Leslie Gudimard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Annie Gravel
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Florian Puhm
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Gabriel Campolina-Silva
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Celso Queiroz-Junior
- Morphology Department, Universidade Federal de Minas Gerais, Belo Horizonte31270-901, Brazil
| | - Isabelle Allaeys
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Julien Prunier
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Oumaima Azeggouar Wallen
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Élizabeth Dumais
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Clémence Belleannée
- Division of Reproduction, mother and youth health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Arnaud Droit
- Division of Endocrinology and Nephrology, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
| | - Nicolas Flamand
- Centre de recherche de l’Institut Universitaire de cardiologie et pneumologie de Québec, Division of pneumology, Faculty of medicine, Université Laval, Québec City, QCG1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QCG1V 4G5, Canada
| | - Éric Boilard
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| | - Louis Flamand
- Division of Infectious and Immune Diseases, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, QCG1V 4G2, Canada
- Centre de Recherche ARThrite–Arthrite, Recherche, Traitements, Université Laval, Québec, QCG1V 4G2, Canada
- Department of microbiology, infectious disease and immunology, Faculty of Medicine, Université Laval, Québec City, QCG1V 0A6, Canada
| |
Collapse
|
6
|
Kameni MN, Tchoupe EB, Kamdem SD, Bhalla N, Assam Assam JP, Tepa AN, Neba FR, Nanda RK, Awuah AAA, Amuasi JH, Netongo PM. Mutations in ace2 gene modulate cytokine levels and alter immune responses in Mycobacterium tuberculosis and SARS-CoV-2 co-infection: a Cameroonian cohort. Front Immunol 2025; 16:1533213. [PMID: 40196114 PMCID: PMC11973369 DOI: 10.3389/fimmu.2025.1533213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction SARS-CoV-2 and Mycobacterium tuberculosis (Mtb) share similarities in their modes of transmission, pathophysiological symptoms, and clinical manifestations. An imbalance in the immune response characterised by elevated levels of some inflammatory cytokines caused by tuberculosis (TB) and COVID-19 may increase the risk of developing a severe disease-like condition. It has been reported that TB increases the expression levels of Ace2 (angiotensin converting enzyme 2) and Tmprss2 (transmembrane protease serine 2) proteins, which are essential for COVID-19 pathogenesis. Single nucleotide polymorphisms (SNPs) variants of ace2 and tmprss2 genes can impact virus and host-cell interactions and alter immune responses by modulating cytokine production. This may modify the susceptibility and/or severity in COVID-19-infected people. The role of SNPs in ace2 and tmprss2 in relation to Mtb and SARS-CoV-2 co-infection is relatively underexplored. Method In this study, genotype frequency of 10 SNPs of ace2 and 03 SNPs of tmprss2 genes in a Cameroonian cohort consisting of COVID-19-positive (n = 31), TB-positive (n = 43), TB-COVID-19 co-infected (n = 21), and a control group (n = 24) were studied. The immune response was estimated by quantitating inflammatory cytokine levels alongside self-reported and clinically diagnosed symptoms. The relationship between specific genetic mutations in these ace2 gene SNPs and their impact on cytokine expression levels in Mtb and SARS-CoV-2 co-infected patients was investigated. Results We identified wild-type, heterozygous, and double-mutant genotypes in seven SNPs (rs2285666, rs6632677, rs4646116, rs4646140, rs147311723, rs2074192 and rs4646142) in ace2 gene, which showed significant variations in distribution across the study groups. Our most significant findings include the association of double mutant alleles (AA) of rs4646140 and rs2074192 in the ace2 gene with decreased IL-6 and IL-2 expression levels respectively in TB-COVID-19 participants. Also, the double mutant alleles (AA) of rs4646116 were responsible for increased expression level of IL-2 in TB-COVID-19 patients. Additionally, elevated serum levels of AST, urea, and D-dimer, as well as increased plasma concentrations of IL-10, IFN-γ, and TNF-α, have been associated with co-infections involving Mtb and SARS-CoV-2. Conclusion These biomarkers may reflect the complex interplay between the two pathogens and their impact on host immune responses and disease progression. This study highlights the critical role of genetic and immunological factors in shaping altered immune responses during co-infections involving Mtb and SARS-CoV-2. By elucidating these factors, the findings provide a foundation for a deeper understanding of host-pathogen interactions and their implications for disease progression and outcomes. Furthermore, this research has the potential to drive advancements in diagnostic approaches enabling more accurate detection and monitoring of co-infections.
Collapse
Affiliation(s)
- Mary Ngongang Kameni
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Microbiology, University of Yaounde I, Yaounde, Cameroon
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Eric Berenger Tchoupe
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Clinical Biochemistry, Faculty of Medicine and Biomedical Science, University of Yaounde I, Yaounde, Cameroon
| | - Severin Donald Kamdem
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Nikhil Bhalla
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Arnaud Njuiget Tepa
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Fuh Roger Neba
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Ranjan Kumar Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - John Humphrey Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Palmer Masumbe Netongo
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Biology Program, School of Science, Navajo Technical University, Crownpoint, NM, United States
- Department of Biochemistry, University of Yaounde I, Yaounde, Cameroon
| |
Collapse
|
7
|
Hailu G, Legesse M, Mulu A, Medhin G, Tsegaye MM, Alemayehu DH, Ayele A, Gebreegziabxier A, Tayachew A, Aguine A, Dejene H, Tessema SK, Onywera H, Stanislas AE, Abate E, Marcello A, Bitew M. SARS-CoV-2 Genetic Variants Identified in Selected Regions of Ethiopia Through Whole Genome Sequencing: Insights from the Fifth Wave of COVID-19. Genes (Basel) 2025; 16:351. [PMID: 40149502 PMCID: PMC11942139 DOI: 10.3390/genes16030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic highlighted SARS-CoV-2 variants with increased transmissibility and immune evasion. In Ethiopia, where cases surged, the understanding of the virus's dynamics was limited. This study analyzed SARS-CoV-2 variants during the fifth wave, crucial for guiding vaccines, therapeutics, diagnostics, and understanding disease severity. METHOD From June to August 2022, 150 SARS-CoV-2-positive samples were randomly selected from the Ethiopian Public Health Institute repository. Sixty-three high-quality genome sequences were analyzed. RESULTS Of the 63 sequences, 70% were from males and 30% from females, with a median age of 34. Omicron dominated (97%, 61/63), primarily clade 22A (64%, 40/63), followed by 22B (18%, 11/63) and 21K (14%, 9/63). Delta accounted for 3.2% (2/63). Omicron was identified in all (25) vaccinated study participants. Ethiopian sequences showed limited evolutionary divergence and lower genetic diversity compared to global sequences. CONCLUSION Omicron was the predominant variant during Ethiopia's fifth wave, indicating recent community transmission. Despite minor genetic diversity differences, ongoing surveillance remains critical for tracking variants and informing public health interventions.
Collapse
Affiliation(s)
- Getnet Hailu
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Mengistu Legesse
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Andargachew Mulu
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Mesfin Mengesha Tsegaye
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Dawit Hailu Alemayehu
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | - Abaysew Ayele
- Armaur Hansson Research Institute, Addis Ababa P.O. Box 1005, Ethiopia; (A.M.); (M.M.T.); (D.H.A.); (A.A.)
| | | | - Adamu Tayachew
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Adimkewu Aguine
- Ethiopian Public Health Institute, Addis Ababa P.O. Box 1242, Ethiopia; (A.G.); (A.A.)
| | - Haileyesus Dejene
- College of Veterinary and Animal Science, University of Gondar, Gondar P.O. Box 196, Ethiopia;
| | - Sofonias K. Tessema
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa P.O. Box 3243, Ethiopia; (S.K.T.); (H.O.)
| | - Harris Onywera
- Africa Centres for Disease Control and Prevention (Africa CDC), Addis Ababa P.O. Box 3243, Ethiopia; (S.K.T.); (H.O.)
| | | | - Ebba Abate
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa P.O. Box 1176, Ethiopia; (G.H.); (M.L.); (G.M.); (A.T.); (E.A.)
| | - Alessandro Marcello
- Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149 Trieste, Italy;
| | - Molalegne Bitew
- Bio and Emerging Technology Institute, Addis Ababa P.O. Box 5954, Ethiopia
| |
Collapse
|
8
|
Pourjoula F, Mirlohi SH, Ghanbari N. Comparative analysis of croup severity and treatment in pediatric patient: a study of COVID-19 positive vs. negative cases during peak Omicron. BMC Pediatr 2025; 25:194. [PMID: 40089722 PMCID: PMC11909977 DOI: 10.1186/s12887-025-05536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The Omicron variant of SARS-CoV-2 has been associated with unique clinical presentations in children, including croup-like symptoms such as barking cough, hoarseness, and respiratory distress. This study aimed to compare the clinical, laboratory, and treatment characteristics of hospitalized pediatric patients with croup who tested positive or negative for COVID-19 during the Omicron wave. METHODS A retrospective, descriptive-analytical study was conducted on 111 pediatric patients hospitalized with croup at Bahrami Children's Hospital and the Children's Medical Center in Iran from January 21 to March 20, 2022. Patients were categorized into two groups: PCR-positive (Omicron group, n = 30) and PCR-negative (non-Omicron group, n = 81). Data on demographics, clinical severity, laboratory indices, treatments, and outcomes were extracted and analyzed using SPSS version 20. RESULTS The mean age of the Omicron group was significantly younger (16.93 ± 24.80 months) compared to the non-Omicron group (32.58 ± 37.26 months; p = 0.049). Symptom severity was higher in the Omicron group, with moderate to severe symptoms observed in 73.4% of patients, compared to 32.1% in the non-Omicron group (p = 0.001). The Omicron group had longer hospital stays (2.59 ± 3.93 vs. 2.11 ± 2.75 days; p = 0.016) and required more nebulized epinephrine (2.47 ± 1.27 vs. 1.77 ± 1.003 days; p = 0.003) and repeat corticosteroid doses (83.3% vs. 38.3%; p = 0.0001). Laboratory findings showed no significant differences between the groups (all p > 0.05). CONCLUSION Children with croup during the Omicron surge exhibited increased symptom severity, required more intensive treatment, and experienced longer hospital stays compared to those without COVID-19. These findings emphasize the need for heightened clinical awareness and tailored management strategies for Omicron-related croup in pediatric populations.
Collapse
Affiliation(s)
- Fatemeh Pourjoula
- Tehran University of Medical Sciences, school of medicine, Tehran, Iran
| | - Seyed Hossein Mirlohi
- Pediatric Respiratory Disease and Sleep Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Ghanbari
- Emergency devision, school of medicine, Bahrami hospital, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
9
|
Zhang X, Han X, Li C, Cui J, Yuan X, Meng J, Han Z, Han X, Chen W, Xiong J, Xie W, Xie L. Clinical Outcomes of Hospitalized Immunocompromised Patients With COVID-19 and the Impact of Hyperinflammation: A Retrospective Cohort Study. J Inflamm Res 2025; 18:3385-3397. [PMID: 40070925 PMCID: PMC11895693 DOI: 10.2147/jir.s482940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025] Open
Abstract
Purpose Immunocompromised patients are at increased risk for severe outcomes from COVID-19 due to their altered immune responses, yet their inflammatory profiles and the interplay between immunosuppression remain poorly understood. We aimed to illustrate the inflammation profile and clinical outcomes of hospitalized immunocompromised patients with COVID-19. Methods We conducted a retrospective study using a multicenter database and included adult hospitalized patients with Corona virus disease 2019 (COVID-19) in China's late 2022 COVID-19 wave. Crude and adjusted 28- and 60-day mortality was compared between the two groups. Inflammatory phenotypes were evaluated by serum interleukin-6 (IL-6) and C-reactive protein (CRP) level. The interplay between overt inflammation and immunosuppression was analyzed. Results Among the 4078 included patients, 348 (8.5%) were immunocompromised. Immunocompromised patients had lower crude mortality but higher adjusted mortality at 28-day (hazard ratio [HR] = 1.55; 95% CI 1.08 to 2.23) and 60-day (HR = 1.47; 95% CI 1.05 to 2.06). Besides, immunocompromised patients had a higher risk of developing hyperinflammation (odd ratio [OR] =1.92; 95% CI 1.47 to 2.50, p <0.001). Moreover, hyperinflammation mediated a major part of the deleterious survival effect of immunosuppression on COVID-19. Conclusion Immunodeficiency not only increases short-term mortality risk but also predisposes patients to hyperinflammation. The complex interplay between immunosuppression, hyperinflammation, and COVID-19 outcomes warrants more detailed profiling of inflammation and immunity in this population.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xiaobo Han
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- Chinese PLA Medical School, Beijing, People’s Republic of China
| | - Chenglong Li
- National Institute of Health Data Science, Peking University, Beijing, People’s Republic of China
- Institute of Medical Technology, Health Science Center, Peking University, Beijing, People’s Republic of China
| | - Junchang Cui
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- Chinese PLA Medical School, Beijing, People’s Republic of China
| | - Xin Yuan
- Department of Pulmonary and Critical Care Medicine, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Jiguang Meng
- Department of Pulmonary and Critical Care Medicine, The Fourth Medical Center of PLA General Hospital, Beijing, People’s Republic of China
- Naval Clinical College, Anhui Medical University, Hefei, People’s Republic of China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Xinjie Han
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Wei Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, People’s Republic of China
| | - Junchen Xiong
- Department of Pulmonary and Critical Care Medicine, The Fourth Medical Center of PLA General Hospital, Beijing, People’s Republic of China
- Naval Clinical College, Anhui Medical University, Hefei, People’s Republic of China
| | - Wuxiang Xie
- Peking University Clinical Research Institute, Peking University First Hospital, Beijing, People’s Republic of China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, People’s Republic of China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, The Eighth Medical Center, Chinese PLA General Hospital, Beijing, People’s Republic of China
- Chinese PLA Medical School, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Bandeira TDJPG, Oliveira ALSD, Martins LFP, Matos RM, Santos SRNGD, Lopes MC, Sobreira RTP, Rocha HAL. Molecular detection of respiratory viruses: an observational study on respiratory co-infections in children and adults. Braz J Microbiol 2025; 56:537-543. [PMID: 39690324 PMCID: PMC11885765 DOI: 10.1007/s42770-024-01581-x] [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: 06/20/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024] Open
Abstract
Respiratory tract infection (RTI) is the most important cause of visiting both public and private medical care services. During the decade preceding Covid-19, in developing countries, there was a gap in the epidemiology of respiratory viral infections, probably due to the difficulty of detecting viral agents in clinical microbiology laboratories routine, at that time. In this context, the aim of this study was to describe the occurrence of virus, demographic factors associated and the likelihood of coinfections detection by multiplex PCR methods. The target also was to determine the involvement of respiratory viruses other than SARS-CoV-2 as agents of upper respiratory tract infections (URTIs) using rapid respiratory panels by multiplex real time PCR with flow chip methodology. Samples of SARSs patients, 530 in total, were tested between 2022 and 2023, obtained from nasopharyngeal swabs, nasopharyngeal exudates or nasopharyngeal aspirates. Of 530 tests, 30% was human rhinovirus, the most prevalent, followed by respiratory syncytial virus B (21.5%) and human adenovirus (17.4%). Among the total of positive samples, 83.8% belonged to patients aged between 0 and 10 years, 62.6% of whom were aged between 1 and 10 years. Multiple virus detection was found in 50.4% of the samples tested, 93.1% of which were from patients under the age of 18. Viral co-infection was evident in the finding and was higher (88.7%) in patients under the age of 18 yeas.
Collapse
|
11
|
Fu X, Lin R, Du W, Tavares A, Liang Y. Explainable hybrid transformer for multi-classification of lung disease using chest X-rays. Sci Rep 2025; 15:6650. [PMID: 39994381 PMCID: PMC11850790 DOI: 10.1038/s41598-025-90607-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Lung disease is an infection that causes chronic inflammation of the human lung cells, which is one of the major causes of death around the world. Thoracic X-ray medical image is a well-known cheap screening approach used for lung disease detection. Deep learning networks, which are used to identify disease features in X-rays medical images, diagnosing a variety of lung diseases, are playing an increasingly important role in assisting clinical diagnosis. This paper proposes an explainable transformer with a hybrid network structure (LungMaxViT) combining CNN initial stage block with SE block to improve feature recognition for predicting Chest X-ray images for multiple lung disease classification. We contrast four classical pre-training models (ResNet50, MobileNetV2, ViT and MaxViT) through transfer learning based on two public datasets. The LungMaxVit, based on maxvit pre-trained with ImageNet 1K datasets, is a hybrid transformer with fine-tuning hyperparameters on the both X-ray datasets. The LungMaxVit outperforms all the four mentioned models, achieving a classification accuracy of 96.8%, AUC scores of 98.3%, and F1 scores of 96.7% on the COVID-19 dataset, while AUC scores of 93.2% and F1 scores of 70.7% on the Chest X-ray 14 dataset. The LungMaxVit distinguishes by its superior performance in terms of Accuracy, AUC and F1-score compared with other hybrids Networks. Several enhancement techniques, such as CLAHE, flipping and denoising, are employed to improve the classification performance of our study. The Grad-CAM visual technique is leveraged to represent the heat map of disease detection, explaining the consistency among clinical doctors and neural network models in the treatment of lung disease from Chest X-ray. The LungMaxVit shows the robust results and generalization in detecting multiple lung lesions and COVID-19 on Chest X-ray images.
Collapse
Affiliation(s)
- Xiaoyang Fu
- School of Computer Science, Zhuhai College of Science and Technology, Zhuhai, 519040, China
| | - Rongbin Lin
- School of Computer Science, Zhuhai College of Science and Technology, Zhuhai, 519040, China
| | - Wei Du
- School of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Adriano Tavares
- Department of Industrial Electronics, University of Minho, 4800-058, Guimarães, Portugal
| | - Yanchun Liang
- School of Computer Science, Zhuhai College of Science and Technology, Zhuhai, 519040, China.
- School of Computer Science and Technology, Jilin University, Changchun, 130012, China.
| |
Collapse
|
12
|
Zhu J, Wu J, Lu M, Jiao Q, Liu X, Liu L, Li M, Zhang B, Yan J, Yu Y, Pan L. Acute lung injury induced by recombinant SARS-CoV-2 spike protein subunit S1 in mice. Respir Res 2025; 26:59. [PMID: 39972348 PMCID: PMC11837662 DOI: 10.1186/s12931-025-03143-7] [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: 10/16/2024] [Accepted: 02/08/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The intricacies of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing acute lung injury (ALI) and modulating inflammatory factor dynamics in vivo remain poorly elucidated. The present study endeavors to explore the impact of the recombinant SARS-CoV-2 spike protein S1 subunit (S1SP) on ALI and inflammatory factor profiles in mice, aiming to uncover potential therapeutic targets and intervention strategies for the prevention and management of Coronavirus Disease 2019 (COVID-19). METHODS To mimic COVID-19 infection, K18-hACE2 transgenic mice were intratracheally instilled with S1SP, while C57BL/6 mice were administered LPS to form a positive control group. This setup facilitated the examination of lung injury severity, inflammatory factor levels, and alterations in signaling pathways in mice mimicking COVID-19 infection. Histopathological assessment through HE staining, along with analysis of lung wet/dry ratio and ultrasound imaging, revealed severe lung injury. RESULTS After molding, K18-hACE2 mice exhibited a pronounced reduction in body weight and showed more significant lung injury (P < 0.05). Notably, there was a significant elevation in vascular permeability, total protein, and total white blood cells in bronchoalveolar lavage fluid (BALF) (P < 0.05), indicative of tissue damage. Additionally, the tight junction of lung tissue was compromised (P < 0.05), accompanied by intense oxidative stress marked by decreased SOD activity and elevated MDA content (P < 0.05). Cytokine levels, including IL-6, IL-1β, TNF-α, and MIG, were significantly upregulated in both BALF and serum of S1SP + K18 mice (P < 0.05). Furthermore, S1SP prominently augmented the expression of p-p65/P65 and attenuated IκBα expression in the NF-κB signaling pathway of humanized mice (P < 0.05), corroborating a heightened inflammatory response at the tissue level (P < 0.05). CONCLUSION The administration of S1SP to K18-hACE2 mice resulted in severe lung injury, enhanced vascular permeability, and compromised epithelial barrier function in vivo. This was accompanied by disruption of lung tight junctions, the manifestation of severe oxidative stress and a cytokine storm, as well as the activation of the NF-κB signaling pathway, highlighting key pathological processes underlying COVID-19-induced lung injury.
Collapse
Affiliation(s)
- Jiwei Zhu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Jinglin Wu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Manlu Lu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Qianqian Jiao
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Xiaojing Liu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Mingzhen Li
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Bin Zhang
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China
| | - Junhong Yan
- Department of Ultrasound Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China.
| | - Yan Yu
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China.
| | - Lei Pan
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, 661 Yellow River Road, Binzhou, 256603, China.
| |
Collapse
|
13
|
He L, Wang Y, Yuan F, Morrissey S, Geller AE, Hu X, Xu R, Ma X, Zhang HG, McLeish K, Huang J, Zhang X, Yan J. Metabolomics Profiling Reveals Critical Roles of Indoxyl Sulfate in the Regulation of Innate Monocytes in COVID-19. Cells 2025; 14:256. [PMID: 39996729 PMCID: PMC11853107 DOI: 10.3390/cells14040256] [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: 01/10/2025] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is intricately related to the reprogramming of host metabolism. However, existing studies have mainly focused on peripheral blood samples and barely identified specific metabolites that are critically involved in the pathology of coronavirus disease 2019 (COVID-19). In the current small-scale study, we performed metabolic profiling in plasma (n = 61) and paired bronchoalveolar lavage fluid (BALF) samples (n = 20) using parallel two-dimensional liquid chromatography-mass spectrometry (2DLC-MS). In addition, we studied how an identified metabolite regulates the immunopathogenesis of COVID-19. The results unveiled distinct metabolome changes between healthy donors, and moderate and severe patients in both plasma and BALF, indicating that locations and disease severity play critical roles in COVID-19 metabolic alteration. Notably, a vital metabolite, indoxyl sulfate, was found to be elevated in both the plasma and BALF of severe COVID-19 patients. Indoxyl sulfate selectively induced TNF-α production, reduced co-stimulatory signals, and enhanced apoptosis in human monocytes. Moreover, its levels negatively correlated with the strength of co-stimulatory signals and antigen presentation capability in monocytes of COVID-19 patients. Collectively, our findings suggest that the levels of indoxyl sulfate could potentially serve as a functional biomarker to monitor COVID-19 disease progression and guide more individualized treatment for COVID-19 patients.
Collapse
Affiliation(s)
- Liqing He
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA; (L.H.); (R.X.); (X.M.); (X.Z.)
| | - Yunke Wang
- Immuno-Oncology Program, Brown Cancer Center, Division of Immunotherapy, MD Department of Surgery, University of Louisville, Louisville, KY 40292, USA (X.H.)
| | - Fang Yuan
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA; (L.H.); (R.X.); (X.M.); (X.Z.)
| | - Samantha Morrissey
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40292, USA
| | - Anne E. Geller
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40292, USA
| | - Xiaoling Hu
- Immuno-Oncology Program, Brown Cancer Center, Division of Immunotherapy, MD Department of Surgery, University of Louisville, Louisville, KY 40292, USA (X.H.)
| | - Raobo Xu
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA; (L.H.); (R.X.); (X.M.); (X.Z.)
| | - Xipeng Ma
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA; (L.H.); (R.X.); (X.M.); (X.Z.)
| | - Huang-ge Zhang
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40292, USA
| | - Kenneth McLeish
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville, Louisville, KY 40292, USA;
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville Hospital, Louisville, KY 40292, USA
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, KY 40292, USA; (L.H.); (R.X.); (X.M.); (X.Z.)
| | - Jun Yan
- Immuno-Oncology Program, Brown Cancer Center, Division of Immunotherapy, MD Department of Surgery, University of Louisville, Louisville, KY 40292, USA (X.H.)
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
14
|
Warner BM, Vendramelli R, Boese AS, Audet J, Tailor N, Meilleur C, Glowach N, Willman M, Truong T, Moffat E, Tierney K, Kosak B, Dhanidina I, Engstrom J, Korczak B, McGowan I, Embury-Hyatt C, Kobasa D. Treatment with the CCR5 antagonist OB-002 reduces lung pathology, but does not prevent disease in a Syrian hamster model of SARS-CoV-2 infection. PLoS One 2025; 20:e0316952. [PMID: 39908288 PMCID: PMC11798459 DOI: 10.1371/journal.pone.0316952] [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: 08/11/2023] [Accepted: 12/18/2024] [Indexed: 02/07/2025] Open
Abstract
Since the emergence of SARS-CoV-2 and the COVID-19 pandemic, a wide range of treatment options have been evaluated in preclinical studies and clinical trials, with several being approved for use in humans. Immunomodulatory drugs have shown success in dampening the deleterious inflammatory response seen in severe COVID-19 patients, but there remains an urgent need for development of additional therapeutic options for COVID-19 treatment. A potential drug target is the CCR5-CCL5 axis, and blocking this pathway may protect against severe disease. Here we evaluated whether OB-002, an analog of human CCL5 and a potent antagonist of CCR5, provides therapeutic benefit in SARS-CoV-2 infected Syrian hamsters. Daily treatment with OB-002 altered immune gene transcription in the lungs, and reduced pathology following infection, but did not prevent weight loss or viral replication in the lungs of infected animals, even in combination with the antiviral drug remdesivir. Our data suggest that targeting the CCR5-CCL5 pathway in SARS-CoV-2 infection in hamsters is insufficient to significantly impact disease development in this model.
Collapse
Affiliation(s)
- Bryce M. Warner
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Robert Vendramelli
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Amrit S. Boese
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Jonathan Audet
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Nikesh Tailor
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Courtney Meilleur
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Nathan Glowach
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Marnie Willman
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Estella Moffat
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | - Kevin Tierney
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Diseases, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
15
|
Eltayeb A, Redwan EM. T-cell immunobiology and cytokine storm of COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:1-30. [PMID: 40246342 DOI: 10.1016/bs.pmbts.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The 2019 coronavirus illness (COVID 2019) first manifests as a newly identified pneumonia and may quickly escalate to acute respiratory distress syndrome, which has caused a global pandemic. Except for individualized supportive care, no curative therapy has been steadfastly advised for COVID-19 up until this point. T cells and virus-specific T lymphocytes are required to guard against viral infection, particularly COVID-19. Delayed immunological reconstitution (IR) and cytokine storm (CS) continue to be significant barriers to COVID-19 cure. While severe COVID-19 patients who survived the disease had considerable lymphopenia and increased neutrophils, especially in the elderly, their T cell numbers gradually recovered. Exhausted T lymphocytes and elevated levels of pro-inflammatory cytokines, including IL6, IL10, IL2, and IL17, are observed in peripheral blood and the lungs. It implies that while convalescent plasma, IL-6 blocking, mesenchymal stem cells, and corticosteroids might decrease CS, Thymosin α1 and adaptive COVID-19-specific T cells could enhance IR. There is an urgent need for more clinical research in this area throughout the world to open the door to COVID-19 treatment in the future.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
16
|
Wang L, Tian W, Wang S, Liu Y, Wang H, Xiao J, Yu Z, Xie L, Chen Y. Serum proteomics identifies biomarkers for predicting non-survivors in elderly COVID-19 patients. J Proteomics 2025; 311:105356. [PMID: 39547396 DOI: 10.1016/j.jprot.2024.105356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
In December 2022, China ceased the zero-COVID-19 policy, resulting in an increase in hospitalizations and deaths due to COVID-19, particularly among the elderly population. Predicting non-survivors aims to identify high-risk patients and enable targeted interventions to improve survival rates. Additionally, understanding factors affecting prognosis provides essential insights for further research and optimization of treatment strategies. We applied 4D-DIA mass spectrometry for serum proteome analysis and provided a comprehensive characterization of disease features in elderly patients within the Chinese population. Our study elucidated that immune disorders, lung damage, and cardiovascular disorders are predominant causes of death in these patients. Compared to clinical indices, proteomic analysis is more sensitive in tracing these disorders. We also provided a prediction panel for survival outcomes of elderly patients using levels of CXCL10, CXCL16 and IL1RA, which were validated by ELISA. These biomarkers will help improve predictive efficacy for survival outcomes in elderly patients.
Collapse
Affiliation(s)
- Lin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Wenmin Tian
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Sen Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yuhong Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Hongli Wang
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China
| | - Junjie Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Zhongkuo Yu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing 100091, China.
| | - Yang Chen
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing 100191, China; Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
17
|
Ueki H, Wang IH, Kiso M, Horie K, Iida S, Mine S, Ujie M, Hsu HW, Wu CHH, Imai M, Suzuki T, Kamitani W, Kawakami E, Kawaoka Y. Neutrophil adhesion to vessel walls impairs pulmonary circulation in COVID-19 pathology. Nat Commun 2025; 16:455. [PMID: 39805823 PMCID: PMC11730596 DOI: 10.1038/s41467-024-55272-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
Microthrombus formation is associated with COVID-19 severity; however, the detailed mechanism remains unclear. In this study, we investigated mouse models with severe pneumonia caused by SARS-CoV-2 infection by using our in vivo two-photon imaging system. In the lungs of SARS-CoV-2-infected mice, increased expression of adhesion molecules in intravascular neutrophils prolonged adhesion time to the vessel wall, resulting in platelet aggregation and impaired lung perfusion. Re-analysis of scRNA-seq data from peripheral blood mononuclear cells from COVID-19 cases revealed increased expression levels of CD44 and SELL in neutrophils in severe COVID-19 cases compared to a healthy group, consistent with our observations in the mouse model. These findings suggest that pulmonary perfusion defects caused by neutrophil adhesion to pulmonary vessels contribute to COVID-19 severity.
Collapse
Affiliation(s)
- Hiroshi Ueki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
| | - I-Hsuan Wang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Maki Kiso
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenta Horie
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Sohtaro Mine
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michiko Ujie
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hung-Wei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chen-Hui Henry Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Wataru Kamitani
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Eiryo Kawakami
- Department of Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Kanagawa, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center, Tokyo, Japan.
| |
Collapse
|
18
|
Peh HY, Chen J. Pro-resolving lipid mediators and therapeutic innovations in resolution of inflammation. Pharmacol Ther 2025; 265:108753. [PMID: 39566561 DOI: 10.1016/j.pharmthera.2024.108753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/07/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
This review summarizes findings presented at the 19th World Congress of Basic & Clinical Pharmacology 2023 (Glasgow, Scotland, July 3rd to 7th, 2023) from 8 speakers in the field of resolution of inflammation, resolution pharmacology and resolution biology. It is now accepted that the acute inflammatory response is protective to defend the host against infection or tissue injury. Acute inflammation is self-limited and programmed to be limited in space and time: this is achieved through endogenous resolution processes that ensure return to homeostasis. Resolution is brought about by agonist mediators that include specialized pro-resolving lipid mediators (SPMs) and pro-resolving proteins and peptides such as annexin A1 and angiotensin-(1-7), all acting to initiate anti-inflammatory and pro-resolving processes. If the inflammatory reaction remains unchecked through dysfunctional resolution mechanism, it can become chronic and contribute to a plethora of human diseases, including respiratory, cardiovascular, metabolic, allergic diseases, and arthritis. Herein, we discuss how non-resolving inflammation plays a role in the pathogenesis of these diseases. In addition to SPMs, we highlight the discovery, biosynthesis, biofunctions, and latest research updates on innovative therapeutics (including annexin-A1 peptide-mimetic RTP-026, small molecule FPR2 agonist BM-986235/LAR-1219, biased agonist for FPR1/FPR2 Cmpd17b, lipoxin mimetics AT-01-KG and AT-02-CT, melanocortin receptor agonist AP1189, gold nanoparticles, angiotensin-(1-7), and CD300a) that can promote resolution of inflammation directly or through modulation of SPMs production. Drug development strategies based on the biology of the resolution of inflammation can offer novel therapeutic means and/or add-on therapies for the treatment of chronic diseases.
Collapse
Affiliation(s)
- Hong Yong Peh
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Pharmacology, Singapore; Immunology Programme and Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom; Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
19
|
Bao Y, Ma Q, Chen L, Feng K, Guo W, Huang T, Cai YD. Recognizing SARS-CoV-2 infection of nasopharyngeal tissue at the single-cell level by machine learning method. Mol Immunol 2025; 177:44-61. [PMID: 39700903 DOI: 10.1016/j.molimm.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
SARS-CoV-2 has posed serious global health challenges not only because of the high degree of virus transmissibility but also due to its severe effects on the respiratory system, such as inducing changes in multiple organs through the ACE2 receptor. This virus makes changes to gene expression at the single-cell level and thus to cellular functions and immune responses in a variety of cell types. Previous studies have not been able to resolve these mechanisms fully, and so our study tries to bridge knowledge gaps about the cellular responses under conditions of infection. We performed single-cell RNA-sequencing of nasopharyngeal swabs from COVID-19 patients and healthy controls. We assembled a dataset of 32,588 cells for 58 subjects for analysis. The data were sorted into eight cell types: ciliated, basal, deuterosomal, goblet, myeloid, secretory, squamous, and T cells. Using machine learning, including nine feature ranking algorithms and two classification algorithms, we classified the infection status of single cells and analyzed gene expression to pinpoint critical markers of SARS-CoV-2 infection. Our findings show distinct gene expression profiles between infected and uninfected cells across diverse cell types, with key indicators such as FKBP4, IFITM1, SLC35E1, CD200R1, MT-ATP6, KRT13, RBM15, and FTH1 illuminating unique immune responses and potential pathways for viral spread and immune evasion. The machine learning methods effectively differentiated between infected and non-infected cells, shedding light on the cellular heterogeneity of SARS-CoV-2 infection. The findings will improve our knowledge of the cellular dynamics of SARS-CoV-2.
Collapse
Affiliation(s)
- YuSheng Bao
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - QingLan Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - KaiYan Feng
- Department of Computer Science, Guangdong AIB Polytechnic College, Guangzhou 510507, China.
| | - Wei Guo
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
20
|
Tyagi S, Tyagi N, Singh A, Gautam A, Singh A, Jindal S, Singh RP, Chaturvedi R, Kushwaha HR. Linking COVID-19 and cancer: Underlying mechanism. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167563. [PMID: 39510388 DOI: 10.1016/j.bbadis.2024.167563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/13/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), lead to a global health crisis with a spectrum of clinical manifestations. A potentially vulnerable category for SARS-CoV-2 infection was identified in patients with other medical conditions. Intriguingly, parallels exist between COVID-19 and cancer at the pathophysiological level, suggesting a possible connection between them. This review discusses all possible associations between COVID-19 and cancer. Expression of receptors like angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) increases COVID-19 susceptibility. SARS-CoV-2 infection might increase cancer susceptibility and accelerate cancer progression through mechanisms involving cytokine storm, tissue hypoxia, impaired T-cell responses, autophagy, neutrophil activation, and oxidative stress. These mechanisms collectively contribute to immune suppression, hindered apoptosis, and altered cellular signaling in the tumor microenvironment, creating conditions favorable for tumor growth, metastasis, and recurrence. Approved vaccines and their impact on cancer patients along-with new clinical trials are also described.
Collapse
Affiliation(s)
- Sourabh Tyagi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nipanshi Tyagi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Anu Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Akanksha Gautam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Awantika Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shelja Jindal
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rana P Singh
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Rupesh Chaturvedi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Hemant Ritturaj Kushwaha
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
21
|
Güven O, Karakurt G, Naser A, Selçuk H, Keleş DV, Gedik E, Avsever M, Köse FF. The Impact of COVID-19 Infection on the Development of Stroke, Pulmonary Embolism, and Myocardial Infarction: A Retrospective Study. Cureus 2025; 17:e77665. [PMID: 39968441 PMCID: PMC11835032 DOI: 10.7759/cureus.77665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION This study compares the period during which thromboembolic disease develops after contact with the virus before, during, and after the pandemic. METHODS In this study, the medical records of patients with a preliminary diagnosis of myocardial infarction (MI), pulmonary embolism (PE), and ischemic stroke who presented to the Emergency Department before, during, and after the pandemic (when vaccination rates increased) were retrospectively examined. Data on whether these patients had COVID-19 or were vaccinated, the time interval between infection/vaccination and the onset of these conditions, and the prognosis were analyzed. RESULTS In the MI group, patients developed embolism the longest after infection and the shortest after vaccination. Among MI patients, the rate of those who received the BioNTech vaccine during the normalization period was higher than that of those who received Sinovac (p = 0.005). In stroke patients, during the pandemic, the time to post-vaccine embolism was shorter (p < 0.001). Additionally, infection and vaccination increased the mortality rate in stroke and PE patients (p < 0.001). CONCLUSION This study demonstrates that thromboembolic events can occur at varying rates and durations after exposure to the virus. While the causes of thrombosis are multifactorial, contact with the virus may act as a triggering factor, even if COVID-19 does not have a direct effect.
Collapse
Affiliation(s)
- Oya Güven
- Department of Emergency Medicine, Kırklareli University Faculty of Medicine, Kırklareli, TUR
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Gökhan Karakurt
- Department of Pulmonology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Abdulrahman Naser
- Department of Cardiology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Hakan Selçuk
- Department of Emergency Medicine, Babaeskı State Hospital, Kırklareli, TUR
| | - Dilek V Keleş
- Department of Nursing, Kırklareli University Faculty of Health Sciences, Kırklareli, TUR
| | - Emre Gedik
- Department of Neurology, Kırklareli Training and Research Hospital, Kırklareli, TUR
| | - Mert Avsever
- Department of Emergency Medicine, St. John's Hospital, Edinburgh, GBR
| | - Fatih Furkan Köse
- Department of Emergency Medicine, Kırklareli Training and Research Hospital, Kırklareli, TUR
| |
Collapse
|
22
|
Kerget B, Topal BN, Çınar İ, Alper F, Sağlam L. Comparison of methotrexate and methylprednisolone as addition to antifibrotic therapy in progressive pulmonary fibrosis due to COVID-19. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024054. [PMID: 39655593 PMCID: PMC11708951 DOI: 10.36141/svdld.v41i4.15614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/16/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Post-covid pulmonary fibrosis (PCPF) is an essential cause of hypoxic respiratory failure, especially in patients with severe COVID-19 infection. In our study, we aimed to compare the effectiveness of methylprednisolone and methotrexate treatments in patients diagnosed with PCPF and in whom progression was observed despite nintedanib treatment. Methods: Forty-eight patients diagnosed with PCPF between April 2022 and February 2023 were followed up in our study. Progressive pulmonary fibrosis was observed in 18 of these patients despite nintedanib treatment. Nintedanib + methylprednisolone treatment was started in Group 1 patients, and nintedanib + methotrexate treatment was started in Group 2 patients, and after three months, a respiratory function test (PFT), 6-minute walk test (6MWT), saturation, pulse, and side effect levels were compared. RESULTS In comparing the groups at the end of the 3rd month, the change in PFT parameters was higher in Group 2 patients than in Group 1 patients. However, there was no statistically significant difference. However, the increase in fingertip saturation, 6MWT levels, and decrease in pulse levels were statistically significantly different in Group 2 patients compared to Group 1 patients (p=0.001 for all). It was observed that complaints of muscle and joint pain, weight gain, and atrophy in peripheral extremities in Group 1 patients were statistically significantly higher than in Group 2 patients (p=0.001, 0.002, 0.001, respectively). CONCLUSION Methotrexate can be used as an alternative to methylprednisolone in PCPF due to its low side effect profile and its effectiveness in PFT, 6MWT, and saturation levels.
Collapse
Affiliation(s)
- Buğra Kerget
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Burcu Nur Topal
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - İsmail Çınar
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Fatih Alper
- Department of Radiology, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Leyla Sağlam
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| |
Collapse
|
23
|
Gonzalez-Orozco M, Tseng HC, Hage A, Xia H, Behera P, Afreen K, Peñaflor-Tellez Y, Giraldo MI, Huante M, Puebla-Clark L, van Tol S, Odle A, Crown M, Teruel N, Shelite TR, Moreno-Contreras J, Terasaki K, Makino S, Menachery V, Endsley M, Endsley JJ, Najmanovich RJ, Bashton M, Stephens R, Shi PY, Xie X, Freiberg AN, Rajsbaum R. TRIM7 ubiquitinates SARS-CoV-2 membrane protein to limit apoptosis and viral replication. Nat Commun 2024; 15:10438. [PMID: 39616206 PMCID: PMC11608229 DOI: 10.1038/s41467-024-54762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024] Open
Abstract
SARS-CoV-2 is a highly transmissible virus that causes COVID-19 disease. Mechanisms of viral pathogenesis include excessive inflammation and viral-induced cell death, resulting in tissue damage. Here we show that the host E3-ubiquitin ligase TRIM7 acts as an inhibitor of apoptosis and SARS-CoV-2 replication via ubiquitination of the viral membrane (M) protein. Trim7-/- mice exhibit increased pathology and virus titers associated with epithelial apoptosis and dysregulated immune responses. Mechanistically, TRIM7 ubiquitinates M on K14, which protects cells from cell death. Longitudinal SARS-CoV-2 sequence analysis from infected patients reveal that mutations on M-K14 appeared in circulating variants during the pandemic. The relevance of these mutations was tested in a mouse model. A recombinant M-K14/K15R virus shows reduced viral replication, consistent with the role of K15 in virus assembly, and increased levels of apoptosis associated with the loss of ubiquitination on K14. TRIM7 antiviral activity requires caspase-6 inhibition, linking apoptosis with viral replication and pathology.
Collapse
Affiliation(s)
- Maria Gonzalez-Orozco
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hsiang-Chi Tseng
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Adam Hage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Padmanava Behera
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Kazi Afreen
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Yoatzin Peñaflor-Tellez
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Maria I Giraldo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Matthew Huante
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lucinda Puebla-Clark
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Sarah van Tol
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Abby Odle
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Matthew Crown
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Natalia Teruel
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Thomas R Shelite
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Joaquin Moreno-Contreras
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Kaori Terasaki
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Shinji Makino
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Vineet Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mark Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Janice J Endsley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rafael J Najmanovich
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Matthew Bashton
- Hub for Biotechnology in the Built Environment, Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, UK
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Ricardo Rajsbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Virus-Host-Innate-Immunity, RBHS Institute for Infectious and Inflammatory Diseases, and Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
24
|
Oh DS, Kim E, Normand R, Lu G, Shook LL, Lyall A, Jasset O, Demidkin S, Gilbert E, Kim J, Akinwunmi B, Tantivit J, Tirard A, Arnold BY, Slowikowski K, Goldberg MB, Filbin MR, Hacohen N, Nguyen LH, Chan AT, Yu XG, Li JZ, Yonker L, Fasano A, Perlis RH, Pasternak O, Gray KJ, Choi GB, Drew DA, Sen P, Villani AC, Edlow AG, Huh JR. SARS-CoV-2 infection elucidates features of pregnancy-specific immunity. Cell Rep 2024; 43:114933. [PMID: 39504241 PMCID: PMC11724703 DOI: 10.1016/j.celrep.2024.114933] [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: 12/14/2023] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnancy is a risk factor for increased severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory infections, but the mechanisms underlying this risk are poorly understood. To gain insight into the role of pregnancy in modulating immune responses at baseline and upon SARS-CoV-2 infection, we collected peripheral blood mononuclear cells and plasma from 226 women, including 152 pregnant individuals and 74 non-pregnant women. We find that SARS-CoV-2 infection is associated with altered T cell responses in pregnant women, including a clonal expansion of CD4-expressing CD8+ T cells, diminished interferon responses, and profound suppression of monocyte function. We also identify shifts in cytokine and chemokine levels in the sera of pregnant individuals, including a robust increase of interleukin-27, known to drive T cell exhaustion. Our findings reveal nuanced pregnancy-associated immune responses, which may contribute to the increased susceptibility of pregnant individuals to viral respiratory infection.
Collapse
Affiliation(s)
- Dong Sun Oh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eunha Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; BK21 Graduate Program, Department of Biomedical Sciences and Department of Neuroscience, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Rachelly Normand
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia L Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda Lyall
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Olyvia Jasset
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stepan Demidkin
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emily Gilbert
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joon Kim
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Babatunde Akinwunmi
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Marcia B Goldberg
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Michael R Filbin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Long H Nguyen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lael Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roy H Perlis
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn J Gray
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David A Drew
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA; Transplant, Oncology, and Immunocompromised Host Group, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Andrea G Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Amoddeo A. In silico assessment of CAR macrophages activity against SARS-CoV-2 infection. Heliyon 2024; 10:e39689. [PMID: 39524874 PMCID: PMC11550025 DOI: 10.1016/j.heliyon.2024.e39689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Macrophage engineering with chimeric antigen receptor is a promising technique first applied to the treatment of tumours and recently suggested as a possible immunotherapeutic route against the COVID-19 disease. Four immunotherapies based on engineered macrophages have been tested in vitro revealing promising, with one of them acting without increasing the cytokines level. We present a mathematical model aimed at the evaluation of both the SARS-CoV-2 virions dynamics and the cytokines production induced, while such newly developed constructs interact with the immune system once administered. The importance of the study lies both in monitoring the dynamics of the infection and in evaluating the cytokine production, since clinical studies show that in critical COVID-19 patients an abnormal cytokines production occurs, a concern to be accounted for in designing appropriate therapeutic strategies. The mathematical model was built in the context of the continuum approach of the mass conservation, while the numerical simulations have been performed introducing parameters deduced from the experiments, using the finite element method. The model simulations allow to analyse and to compare the immune mechanisms underlying the virus dynamics, deepening the investigation for two selected immunotherapies, suggesting that a synergistic work of involved cytokines with phagocytic activity of macrophages occurs. The best SARS-CoV-2 clearance relies not only on the phagocytic capacity of the engineered macrophages, but also on the production of T-lymphocytes, pro- and anti-inflammatory cytokines which in the two cases examined in depth can decrease by 99.7 %, 99.6 % and 69 % respectively, passing from the most effective immunotherapy to the least effective one. This study is the first mathematical model that analyses the dynamics of macrophages engineered to fight the COVID-19, and paves the way for their possible exploitation against such a challenging disease, going beyond existing models involving other immune cells.
Collapse
Affiliation(s)
- Antonino Amoddeo
- Department of Civil, Energy, Environment and Materials Engineering, Università’Mediterranea’ di Reggio Calabria, Via R. Zehender 1, Feo di Vito, I-89122, Reggio Calabria, Italy
| |
Collapse
|
26
|
Xu Y, Huang Y, Yuan Z, Liu W, Wang L, Liu L. Systematic evaluation of therapeutic effectiveness of Azvudine in treating COVID-19 hospitalized patients: a retrospective cohort study. Front Cell Infect Microbiol 2024; 14:1453234. [PMID: 39575306 PMCID: PMC11578945 DOI: 10.3389/fcimb.2024.1453234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Background Azvudine, a repurposed oral small molecule antiviral drug, has potential effects in combating the SARS-CoV-2 virus. However, studies on its clinical efficacy in patients with COVID-19 are still limited and controversial, and further research and validation are necessary. Methods A retrospective cohort study was conducted on COVID-19 patients who were hospitalized in the General Hospital of Central Theater Command from 1 December 2022 to 31 January 2023. We included 132 patients treated with Azvudine and 132 controls after screening and propensity score matching. The primary outcomes including all-cause mortality and a composite outcome of disease progression such as non-invasive respiratory support, invasive respiratory support, admission to intensive care unit (ICU), and death were compared. Results Azvudine recipients had a much lower incidence rate of composite disease progression outcome than controls (13.9075/1000 person-days versus 25.7731/1000 person-days, P<0.05). Azvudine recipients also possessed a lower all-cause mortality rate than controls (2.6797/1000 person-days versus 8.5910/1000 person-days, P<0.01). Azvudine treatment significantly reduced the risk of composite disease progression (HR: 0.37, 95% CI: 0.16-0.84, P=0.017) and all-cause death (HR: 0.25, 95% CI: 0.08-0.81, P=0.021) after adjusting potential confounding factors such as age, sex, severity of COVID-19, complications, concomitant therapy, time from symptoms to treatment, and important laboratory indicators. The subgroup analyses of composite disease progression outcome and all-cause death indicated robustness of Azvudine's in treating COVID-19 patients in general. Conclusion Our study demonstrates that Azvudine has a significant positive impact on the clinical recovery of hospitalized patients with COVID-19. These findings provide important support for the use of Azvudine as a therapeutic option for COVID-19, given the current divergent views on its therapeutic efficacy and its importance in public health and medical care.
Collapse
Affiliation(s)
- Yingkai Xu
- Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
- Department of Transfusion Medicine; General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Yuan Huang
- Department of Gynaecology and Obstetrics; General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Zihan Yuan
- Medical College, Wuhan University of Science and Technology, Wuhan, Hubei, China
- Department of Transfusion Medicine; General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Wanbing Liu
- Department of Transfusion Medicine; General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Li Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Kaifeng, Henan, China
| | - Lei Liu
- Department of Transfusion Medicine; General Hospital of Central Theater Command, Wuhan, Hubei, China
| |
Collapse
|
27
|
Barbon S, Armellin F, Passerini V, De Angeli S, Primerano S, Del Pup L, Durante E, Macchi V, De Caro R, Parnigotto PP, Veronesi A, Porzionato A. Innate immune response in COVID-19: single-cell multi-omics profile of NK lymphocytes in a clinical case series. Cell Commun Signal 2024; 22:496. [PMID: 39407208 PMCID: PMC11476714 DOI: 10.1186/s12964-024-01867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND COVID-19 pandemic caused by the Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) represents the biggest global health emergency in recent decades. The host immune response to SARS-CoV-2 seems to play a key role in disease pathogenesis and clinical manifestations, with Natural Killer (NK) lymphocytes being among the targets of virus-induced regulation. METHODS This study performed a single-cell multi-omics analysis of transcripts and proteins of NK lymphocytes in COVID-19 patients, for the characterization of the innate immunological response to infection. NK cells were isolated from peripheral blood samples collected from adult subjects divided into 3 study groups: (1) non-infected subjects (Naïve group, n = 3), (2) post COVID-19 convalescent subjects (Healed group, n = 3) and (3) patients that were vaccinated against SARS-CoV-2 (Vaccine group, n = 3). Cells were then analysed by the BD Rhapsody System for the single-cell multi-omics investigation of transcriptome and membrane proteins. RESULTS The bioinformatic analysis identified 5 cell clusters which differentially expressed gene/protein markers, defining NK cell subsets as "Active NK cells" and "Mature NK cells". Calculating the relative proportion of each cluster within patient groups, more than 40% of the Naïve group cell population was found to belong to Mature NKs, whereas more than 75% of the Vaccine group cell population belonged to the cluster of Active NKs. Regarding the Healed group, it seemed to show intermediate phenotype between Active and Mature NK cells. Differential expression of specific genes, proteins and signaling pathways was detected comparing the profile of the 3 experimental groups, revealing a more activated NK cell phenotype in vaccinated patients versus recovered individuals. CONCLUSIONS The present study detected differential expression of NK cell markers in relation to SARS-CoV-2 infection and vaccine administration, suggesting the possibility to identify key molecular targets for clinical-diagnostic use of the individual response to viral infection and/or re-infection.
Collapse
Affiliation(s)
- Silvia Barbon
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy.
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy.
| | - Fabrizio Armellin
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Verena Passerini
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Sergio De Angeli
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Simona Primerano
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Laura Del Pup
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Elisabetta Durante
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy
| | - Veronica Macchi
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
| | - Raffaele De Caro
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| | - Arianna Veronesi
- Complex Operative Unit of Transfusion Medicine - Marca Trevigiana Local Unit of Health and Social Services 2, Treviso Hospital, Piazzale dell'Ospedale 1, 31100, Treviso, Italy.
| | - Andrea Porzionato
- Section of Human Anatomy, Department of Neuroscience, University of Padova, Via Gabelli 65, 35121, Padova, Italy
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling - T.E.S. Onlus, Padova, Italy
| |
Collapse
|
28
|
Liu L, Zhang L, Hao X, Wang Y, Zhang X, Ge L, Wang P, Tian B, Zhang M. Coronavirus envelope protein activates TMED10-mediated unconventional secretion of inflammatory factors. Nat Commun 2024; 15:8708. [PMID: 39379362 PMCID: PMC11461611 DOI: 10.1038/s41467-024-52818-0] [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: 03/30/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
The precise cellular mechanisms underlying heightened proinflammatory cytokine production during coronavirus infection remain incompletely understood. Here we identify the envelope (E) protein in severe coronaviruses (SARS-CoV-2, SARS, or MERS) as a potent inducer of interleukin-1 release, intensifying lung inflammation through the activation of TMED10-mediated unconventional protein secretion (UcPS). In contrast, the E protein of mild coronaviruses (229E, HKU1, or OC43) demonstrates a less pronounced effect. The E protein of severe coronaviruses contains an SS/DS motif, which is not present in milder strains and facilitates interaction with TMED10. This interaction enhances TMED10-oligomerization, facilitating UcPS cargo translocation into the ER-Golgi intermediate compartment (ERGIC)-a pivotal step in interleukin-1 UcPS. Progesterone analogues were identified as compounds inhibiting E-enhanced release of proinflammatory factors and lung inflammation in a Mouse Hepatitis Virus (MHV) infection model. These findings elucidate a molecular mechanism driving coronavirus-induced hyperinflammation, proposing the E-TMED10 interaction as a potential therapeutic target to counteract the adverse effects of coronavirus-induced inflammation.
Collapse
Affiliation(s)
- Lei Liu
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Lijingyao Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinyan Hao
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yang Wang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaochun Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peihui Wang
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Meili Lake Translational Research Park, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Boxue Tian
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Min Zhang
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, 100084, China.
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
29
|
Shi Q, Chen Z, Yang J, Liu X, Su Y, Wang M, Xi J, Yang F, Li F. Review of Codonopsis Radix biological activities: A plant of traditional Chinese tonic. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118334. [PMID: 38740108 DOI: 10.1016/j.jep.2024.118334] [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: 11/12/2023] [Revised: 04/06/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Codonopsis Radix, commonly known as Dangshen in Chinese, is frequently used to treat deficiencies of spleen and lung Qi, gastrointestinal discomfort, fatigue, asthmatic breathing, sallow complexion, lack of strength, shortness of breath, deficiencies of both Qi and blood, as well as impairments to both Qi and body fluids in suboptimal health status. AIM OF THE REVIEW This review systematically expounds on the modern pharmacological studies related to the use of Codonopsis Radix in invigorating Qi and nourishing the body in recent years. The aim is to provide theoretical research and reference for the in-depth and systematic exploration and development of the applications of Codonopsis Radix in the fields of food and medicine. MATERIALS AND METHODS This study employs "Codonopsis Radix," "Codonopsis," and "Dangshen" as keywords to gather pertinent information on Codonopsis Radix medicine through electronic searches of classical literature and databases such as PubMed, Elsevier, Google Scholar, Wiley, EMBASE, Cochrane Library, Web of Science, CNKI, Wanfang, VIP, and Baidu Scholar. RESULTS From previous studies, activities such as immune system modulation, gastrointestinal motility regulation, cardiac function revitalization, lung function improvement, blood circulation enhancement, aging process deceleration, learning and memory augmentation, fatigue resistance enhancement, and liver and kidney damage protection of Codonopsis Radix have been reported. Recognized as an important medicine and food homologous traditional Chinese herbal remedy for supplementing deficiencies, its mode of action is multi-elemental, multi-systemic, multi-organ, multi-mechanistic, and multi-targeted. Furthermore, the benefits of its tonic surpass its therapeutic value, establishing it as an extraordinary preventive and therapeutic medicine. CONCLUSIONS With its long history of traditional applications and the revelations of contemporary pharmacological research, Codonopsis Radix exhibits great potential as both a therapeutic agent and a dietary supplement for further research in medicine, nutrition, and healthcare.
Collapse
Affiliation(s)
- Qi Shi
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Zhengjun Chen
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jie Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuxia Liu
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yuanjin Su
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Miao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Jiayu Xi
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Fude Yang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| | - Fang Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| |
Collapse
|
30
|
Secerlı J, Çetinkaya S, Leblebici İS, Özdemir LA, Yücel Ç, Karaismailoğlu E, Kara U, Özcan A, Öcal N, Arslan Y, Şenkal S, Erdem O, Bacanlı MG. Effects of Immunotoxicity biomarkers, essential elements and vitamin D levels on the severity levels of COVID-19 disease in Turkey. Toxicol Res (Camb) 2024; 13:tfae177. [PMID: 39439527 PMCID: PMC11491277 DOI: 10.1093/toxres/tfae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Many mechanisms are thought to play a role in the pathogenesis of the COVID-19 pandemic, which started in 2019 and affected the whole world. It has been claimed that a deficiency in the immune system can significantly affect the severity of COVID-19 disease. It is important that the levels of essential elements and vitamin D are at certain levels for the healthy functioning of the immune system. Therefore, in this study, it was aimed to evaluate immunotoxicity biomarkers (tumor necrosis factor-alpha (TNF-α), interleukin (IL)-10, interferon (IFN)-γ, monocyte chemotactic protein-1 (MCP-1)), vitamin D, and essential element levels in COVID-19 patients in Turkey. According to the results of the study, it was found that the magnesium (Mg), zinc (Zn), and selenium (Se) levels decreased as the severity of the disease worsened, while the iron (Fe), and copper (Cu) levels were similar to the mild group and the control group, and the levels decreased as the disease worsened. It has also been found that vitamin D levels decrease as the severity of the disease worsens. Compared to the control group, TNF-α, MCP-1, and IFN-γ levels were found to decrease as the severity of the disease worsened. Also, it was observed that there was a significant relationship between essential metal levels and disease progression in most of the patient groups.
Collapse
Affiliation(s)
- Jülide Secerlı
- Department of Pharmaceutical Toxicology, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| | - Serdar Çetinkaya
- Department of Pharmaceutical Toxicology, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| | - İlknur Sıla Leblebici
- Department of Pharmaceutical Toxicology, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| | - Latif Alperen Özdemir
- Department of Chest Diseases, University of Health Sciences Turkey, Gülhane Research and Training Hospital, 06018 Ankara, Türkiye
| | - Çiğdem Yücel
- Department of Pharmaceutical Biochemistry, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| | - Eda Karaismailoğlu
- Department of Biostatistics, University of Health Sciences Turkey, Gülhane Faculty of Medicine, 06018 Ankara, Türkiye
| | - Umut Kara
- General Intensive Care Unit, Special Acıbadem Ataşehir Hospital, 34758 İstanbul, Türkiye
| | - Aydan Özcan
- Department of Sports Medicine, University of Health Sciences Turkey, Gülhane Faculty of Medicine, 06018 Ankara, Türkiye
| | - Nesrin Öcal
- Department of Chest Diseases, University of Health Sciences Turkey, Gülhane Research and Training Hospital, 06018 Ankara, Türkiye
| | - Yakup Arslan
- Department of Chest Diseases, University of Health Sciences Turkey, Gülhane Research and Training Hospital, 06018 Ankara, Türkiye
| | - Serkan Şenkal
- General Intensive Care Unit, Special Acıbadem Ataşehir Hospital, 34758 İstanbul, Türkiye
| | - Onur Erdem
- Department of Pharmaceutical Toxicology, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| | - Merve Güdül Bacanlı
- Department of Pharmaceutical Toxicology, University of Health Sciences Turkey, Gülhane Faculty of Pharmacy, 06018 Ankara, Türkiye
| |
Collapse
|
31
|
Guo M, Shang S, Li M, Cai G, Li P, Chen X, Li Q. Understanding autoimmune response after SARS-CoV-2 infection and the pathogenesis/mechanisms of long COVID. MEDICAL REVIEW (2021) 2024; 4:367-383. [PMID: 39444797 PMCID: PMC11495526 DOI: 10.1515/mr-2024-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/04/2024] [Indexed: 10/25/2024]
Abstract
COVID-19 posed a major challenge to the healthcare system and resources worldwide. The popularization of vaccines and the adoption of numerous prevention and control measures enabled the gradual end of the COVID-19 pandemic. However, successive occurrence of autoimmune diseases in patients with COVID-19 cannot be overlooked. Long COVID has been the major focus of research due to the long duration of different symptoms and the variety of systems involved. Autoimmunity may play a crucial role in the pathogenesis of long COVID. Here, we reviewed several autoimmune disorders occurring after COVID-19 infection and the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Ming Guo
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Shunlai Shang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Mengfei Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
- Haihe Laboratory of CellEcosystem, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| |
Collapse
|
32
|
Ma X, Huang T, Li X, Zhou X, Pan H, Du A, Zeng Y, Yuan K, Wang Z. Exploration of the link between COVID-19 and gastric cancer from the perspective of bioinformatics and systems biology. Front Med (Lausanne) 2024; 11:1428973. [PMID: 39371335 PMCID: PMC11449776 DOI: 10.3389/fmed.2024.1428973] [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: 05/07/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19), an infectious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has caused a global pandemic. Gastric cancer (GC) poses a great threat to people's health, which is a high-risk factor for COVID-19. Previous studies have found some associations between GC and COVID-19, whereas the underlying molecular mechanisms are not well understood. Methods We employed bioinformatics and systems biology to explore these links between GC and COVID-19. Gene expression profiles of COVID-19 (GSE196822) and GC (GSE179252) were obtained from the Gene Expression Omnibus (GEO) database. After identifying the shared differentially expressed genes (DEGs) for GC and COVID-19, functional annotation, protein-protein interaction (PPI) network, hub genes, transcriptional regulatory networks and candidate drugs were analyzed. Results We identified 209 shared DEGs between COVID-19 and GC. Functional analyses highlighted immune-related pathways as key players in both diseases. Ten hub genes (CDK1, KIF20A, TPX2, UBE2C, HJURP, CENPA, PLK1, MKI67, IFI6, IFIT2) were identified. The transcription factor/gene and miRNA/gene interaction networks identified 38 transcription factors (TFs) and 234 miRNAs. More importantly, we identified ten potential therapeutic agents, including ciclopirox, resveratrol, etoposide, methotrexate, trifluridine, enterolactone, troglitazone, calcitriol, dasatinib and deferoxamine, some of which have been reported to improve and treat GC and COVID-19. Conclusion This research offer valuable insights into the molecular interplay between COVID-19 and GC, potentially guiding future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Wang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Tao Z, Pu Q, Shen Y, Zhang S, Wang C, Hu Z, Jin Y, Zhu X, Weng Y. Clinical characteristics and prognostic factors of pulmonary and extrapulmonary cryptococcosis. BMC Infect Dis 2024; 24:1018. [PMID: 39304813 DOI: 10.1186/s12879-024-09895-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Cryptococcosis is progressively acknowledged among people, irrespective of the human with or without immunodeficiency virus (HIV). This change in epidemiology has been recorded in recent years, prompting closer examination and a broader understanding of the disease manifestations and risk factors. METHODS The data of cryptococcal infections in China during 11 years were retrospectively analyzed. According to the position of infection, the patients were categorized into the pulmonary infection group and extrapulmonary infection group. The composition of the two groups was compared, and the potential risk factors of disseminated infection were analyzed. Logistic regression was used to analyze the prognostic risk factors of the disease. RESULTS A total of 165 patients were enrolled. 113 (68.5%) were male, and the age was 47.49 (18-82) years. 101 cases (61.2%) had a normal immune function and 64 cases (38.8%) had impaired immune function. 45 patients had extrapulmonary infection, involving the central nervous system, bone and joint, skin and bloodstream, and 120 patients had simple pulmonary infection. The mortality of the extrapulmonary infection group (48.9%) was significantly higher than that of the pulmonary infection group (0.8%). According to univariate logistic regression analysis, immune status (hazard ratio [HR], 4.476; 95% confidence interval [CI], 1.725-11.618; P = 0.002), infection position ([HR], 113.826; [CI], 14.607-886.967; P < 0.001), white blood cell count, ([HR],1.209;[CI], 1.054-1.386; P = 0.007), hemoglobin ([HR], 0.970; [CI], 0.955-0.986; P < 0.001), platelet count ([HR], 0.993; [CI], 0.987-0.999; P = 0.026), neutrophil percentage ([HR], 1.115; [CI], 1.065-1.168; P < 0.001), lymphocyte percentage ([HR], 0.875; [CI], 0.827-0.927; P < 0.001), neutrophil-to-lymphocyte Ratio (NLR) ([HR], 1.144; [CI], 1.072-1.221; P < 0.001), monocyte percentage ([HR], 0.752; [CI], 0.618-0.915; P = 0.004) were related to the prognosis. Multivariate logistic regression analysis showed that the infection position was remained related to the prognosis with statistical significance ([HR], 0.018; [CI], 0.001-0.384; P = 0.001). CONCLUSION Extrapulmonary infection of Cryptococcosis is an important risk factor for prognosis. High levels of neutrophils and NLR, and low levels of lymphocytes and monocytes may lead to disseminated infection of Cryptococcosis. Further studies are needed to reduce the occurrence rate of extrapulmonary infection and mortality.
Collapse
Affiliation(s)
- Ziwei Tao
- Department of Infectious Disease, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China
| | - Qinqin Pu
- Department of Infectious Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yongli Shen
- Department of Infectious Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Sicheng Zhang
- Department of Acute and Critical Care Medicine, Hefei First People's Hospital, Hefei, 230001, China
| | - Chuanyou Wang
- Department of Infectious Disease, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China
| | - Zhe Hu
- Department of Infectious Disease, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China
| | - Yi Jin
- Department of Gastroenterology, Jiangsu Provincial Rongjun Hospital, Wuxi, 214000, China
| | - Xiaowu Zhu
- Department of Infectious Disease, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230000, China.
| | - Yali Weng
- Department of Infectious Disease, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
34
|
McLarnon T, McDaid D, Lynch SM, Cooper E, McLaughlin J, McGilligan VE, Watterson S, Shukla P, Zhang SD, Bucholc M, English A, Peace A, O’Kane M, Kelly M, Bhavsar M, Murray EK, Gibson DS, Walsh CP, Bjourson AJ, Rai TS. Potential Plasma Proteins (LGALS9, LAMP3, PRSS8 and AGRN) as Predictors of Hospitalisation Risk in COVID-19 Patients. Biomolecules 2024; 14:1163. [PMID: 39334929 PMCID: PMC11429560 DOI: 10.3390/biom14091163] [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: 08/07/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has posed unprecedented challenges to healthcare systems worldwide. Here, we have identified proteomic and genetic signatures for improved prognosis which is vital for COVID-19 research. Methods: We investigated the proteomic and genomic profile of COVID-19-positive patients (n = 400 for proteomics, n = 483 for genomics), focusing on differential regulation between hospitalised and non-hospitalised COVID-19 patients. Signatures had their predictive capabilities tested using independent machine learning models such as Support Vector Machine (SVM), Random Forest (RF) and Logistic Regression (LR). Results: This study has identified 224 differentially expressed proteins involved in various inflammatory and immunological pathways in hospitalised COVID-19 patients compared to non-hospitalised COVID-19 patients. LGALS9 (p-value < 0.001), LAMP3 (p-value < 0.001), PRSS8 (p-value < 0.001) and AGRN (p-value < 0.001) were identified as the most statistically significant proteins. Several hundred rsIDs were queried across the top 10 significant signatures, identifying three significant SNPs on the FSTL3 gene showing a correlation with hospitalisation status. Conclusions: Our study has not only identified key signatures of COVID-19 patients with worsened health but has also demonstrated their predictive capabilities as potential biomarkers, which suggests a staple role in the worsened health effects caused by COVID-19.
Collapse
Affiliation(s)
- Thomas McLarnon
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Darren McDaid
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Seodhna M. Lynch
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Eamonn Cooper
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Joseph McLaughlin
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Victoria E. McGilligan
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Steven Watterson
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Priyank Shukla
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Shu-Dong Zhang
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Magda Bucholc
- School of Computing, Engineering & Intelligent Systems, Ulster University, Derry BT48 7JL, UK
| | - Andrew English
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
- School of Health and Life Sciences, Teesside University, Campus Heart, Middlesbrough TS1 3BX, UK
| | - Aaron Peace
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
- Altnagelvin Area Hospital, Western Health and Social Care Trust, Derry BT47 6SB, UK
| | - Maurice O’Kane
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
- Clinical Chemistry Laboratory, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - Martin Kelly
- Altnagelvin Area Hospital, Western Health and Social Care Trust, Derry BT47 6SB, UK
| | - Manav Bhavsar
- Altnagelvin Area Hospital, Western Health and Social Care Trust, Derry BT47 6SB, UK
| | - Elaine K. Murray
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - David S. Gibson
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Colum P. Walsh
- Biomedical Sciences Research Institute, University of Ulster, Coleraine BT52 1SA, UK
| | - Anthony J. Bjourson
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| | - Taranjit Singh Rai
- Personalised Medicine Centre, C-TRIC Building, Altnagelvin Area Hospital, School of Medicine, Ulster University, Glenshane Road, Derry-Londonderry BT47 6SB, UK
| |
Collapse
|
35
|
Lieber CM, Kang HJ, Sobolik EB, Sticher ZM, Ngo VL, Gewirtz AT, Kolykhalov AA, Natchus MG, Greninger AL, Suthar MS, Plemper RK. Efficacy of late-onset antiviral treatment in immunocompromised hosts with persistent SARS-CoV-2 infection. J Virol 2024; 98:e0090524. [PMID: 39207133 PMCID: PMC11406939 DOI: 10.1128/jvi.00905-24] [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: 05/22/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Immunocompromised people are at high risk of prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and progression to severe coronavirus disease 2019 (COVID-19). However, the efficacy of late-onset direct-acting antiviral (DAA) therapy with therapeutics in clinical use and experimental drugs to mitigate persistent viral replication is unclear. In this study, we employed an immunocompromised mouse model, which supports prolonged replication of SARS-CoV-2 to explore late-onset treatment options. Tandem immuno-depletion of CD4+ and CD8+ T cells in C57BL/6 mice followed by infection with SARS-CoV-2 variant of concern (VOC) beta B.1.351 resulted in prolonged infection with virus replication for 5 weeks after inoculation. Early-onset treatment with nirmatrelvir/ritonavir (paxlovid) or molnupiravir was only moderately efficacious, whereas the experimental therapeutic 4'-fluorouridine (4'-FlU, EIDD-2749) significantly reduced virus load in the upper and lower respiratory compartments 4 days postinfection (dpi). All antivirals significantly lowered virus burden in a 7-day treatment regimen initiated 14 dpi, but paxlovid-treated animals experienced rebound virus replication in the upper respiratory tract 7 days after treatment end. Viral RNA was detectable 28 dpi in paxlovid-treated animals, albeit not in the molnupiravir or 4'-FlU groups, when treatment was initiated 14 dpi and continued for 14 days. Low-level virus replication continued 35 dpi in animals receiving vehicle but had ceased in all treatment groups. These data indicate that late-onset DAA therapy significantly shortens the duration of persistent virus replication in an immunocompromised host, which may have implications for clinical use of antiviral therapeutics to alleviate the risk of progression to severe disease in highly vulnerable patients. IMPORTANCE Four years after the onset of the global coronavirus disease 2019 (COVID-19) pandemic, the immunocompromised are at greatest risk of developing life-threatening severe disease. However, specific treatment plans for this most vulnerable patient group have not yet been developed. Employing a CD4+ and CD8+ T cell-depleted immunocompromised mouse model of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we explored therapeutic options of persistent infections with standard-of-care paxlovid, molnupiravir, and the experimental therapeutic 4'-fluorouridine (4'-FlU). Late-onset treatment initiated 14 days after infection was efficacious, but only 4'-FlU was rapidly sterilizing. No treatment-experienced viral variants with reduced susceptibility to the drugs emerged, albeit virus replication rebounded in animals of the paxlovid group after treatment end. This study supports the use of direct-acting antivirals (DAAs) for late-onset management of persistent SARS-CoV-2 infection in immunocompromised hosts. However, treatment courses likely require to be extended for maximal therapeutic benefit, calling for appropriately powered clinical trials to meet the specific needs of this patient group.
Collapse
Affiliation(s)
- Carolin M. Lieber
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia, USA
| | - Hae-Ji Kang
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia, USA
| | - Elizabeth B. Sobolik
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | - Zachary M. Sticher
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | - Vu L. Ngo
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia, USA
| | - Andrew T. Gewirtz
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia, USA
| | | | - Michael G. Natchus
- Emory Institute for Drug Development, Emory University, Atlanta, Georgia, USA
| | - Alexander L. Greninger
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | - Mehul S. Suthar
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Richard K. Plemper
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Rozanovic M, Várady-Szabó K, Domokos K, Kiss T, Loibl C, Márovics G, Rendeki S, Csontos C. Non-Conventional Prognostic Markers in Life-Threatening COVID-19 Cases-When Less Is More. J Clin Med 2024; 13:5369. [PMID: 39336857 PMCID: PMC11432304 DOI: 10.3390/jcm13185369] [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: 08/10/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: In this study, we aimed to compare the predictive power of non-conventional (neutrophil/lymphocyte ratio-NLR; platelet/lymphocyte ratio-PLR) and conventional markers (C-reactive protein-CRP; procalcitonin-PCT; interleukin-6-IL-6) in terms of disease progression and mortality in severe SARS-CoV-2 patients. Methods: In this prospective observatory study, blood samples were collected daily, focusing on the established inflammatory markers. Critically ill COVID-19 patients who required ICU admission were included. Patient treatment followed established COVID-19 protocols, and the data analysis was performed using SPSS with non-normal distribution methods. The study cohort primarily included patients infected with the delta variant. Results: A mortality rate of 76.6% was observed among 167 patients during the study period. Significant differences in conventional and non-conventional markers between survivor and non-survivor groups were observed. The PCT levels were significantly elevated (p < 0.005) in the deceased group. Among the non-conventional markers, the NLR was consistently higher in non-survivors and emerged as a significant predictor of mortality, whereas the PLR was not elevated among the non-survivors. ROC analyses indicated that PCT and the NLR were the markers with the highest predictive power for mortality. The multivariate logistic regression analysis identified NLR, PCT, CRP, and IL-6 as significant predictors of mortality across different days. The NLR showed a consistent, though not always statistically significant, association with increased mortality risk, particularly on Days 2 and 5. Conclusions: The NLR's accessibility and simplicity of determination make it a valuable and practical tool for monitoring inflammatory processes in viral infections. Our findings suggest that incorporating NLR analysis into routine clinical practice could enhance the early identification of high-risk patients, thereby improving patient management and outcomes.
Collapse
Affiliation(s)
- Martin Rozanovic
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Kata Várady-Szabó
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Kamilla Domokos
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Tamás Kiss
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Csaba Loibl
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Gergely Márovics
- Department of Public Health Medicine, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Szilárd Rendeki
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| | - Csaba Csontos
- Department of Anaesthesiology and Intensive Care, University of Pécs Medical School, 7624 Pécs, Hungary; (M.R.); (K.D.); (T.K.); (C.C.)
| |
Collapse
|
37
|
Wang W, Mao Q, Tian Y, Zhang Y, Xiang Z, Ren L. FMD-UNet: fine-grained feature squeeze and multiscale cascade dilated semantic aggregation dual-decoder UNet for COVID-19 lung infection segmentation from CT images. Biomed Phys Eng Express 2024; 10:055031. [PMID: 39142295 DOI: 10.1088/2057-1976/ad6f12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024]
Abstract
With the advancement of computer-aided diagnosis, the automatic segmentation of COVID-19 infection areas holds great promise for assisting in the timely diagnosis and recovery of patients in clinical practice. Currently, methods relying on U-Net face challenges in effectively utilizing fine-grained semantic information from input images and bridging the semantic gap between the encoder and decoder. To address these issues, we propose an FMD-UNet dual-decoder U-Net network for COVID-19 infection segmentation, which integrates a Fine-grained Feature Squeezing (FGFS) decoder and a Multi-scale Dilated Semantic Aggregation (MDSA) decoder. The FGFS decoder produces fine feature maps through the compression of fine-grained features and a weighted attention mechanism, guiding the model to capture detailed semantic information. The MDSA decoder consists of three hierarchical MDSA modules designed for different stages of input information. These modules progressively fuse different scales of dilated convolutions to process the shallow and deep semantic information from the encoder, and use the extracted feature information to bridge the semantic gaps at various stages, this design captures extensive contextual information while decoding and predicting segmentation, thereby suppressing the increase in model parameters. To better validate the robustness and generalizability of the FMD-UNet, we conducted comprehensive performance evaluations and ablation experiments on three public datasets, and achieved leading Dice Similarity Coefficient (DSC) scores of 84.76, 78.56 and 61.99% in COVID-19 infection segmentation, respectively. Compared to previous methods, the FMD-UNet has fewer parameters and shorter inference time, which also demonstrates its competitiveness.
Collapse
Affiliation(s)
- Wenfeng Wang
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| | - Qi Mao
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| | - Yi Tian
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| | - Yan Zhang
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| | - Zhenwu Xiang
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| | - Lijia Ren
- School of Electronic and Electrical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, People's Republic of China
| |
Collapse
|
38
|
Sukumaran SY, Herrscher C, Rasol NE, Othman MA, Liew SY, Ismail NH, Séron K, Litaudon M, Awang K, El Kalamouni C, Apel C, Zahari A. Targeted Isolation of Antiviral Labdane Diterpenes from the Bark of Neo-uvaria foetida (Annonaceae) using LC-MS/MS-Based Molecular Networking. JOURNAL OF NATURAL PRODUCTS 2024; 87:1941-1951. [PMID: 39028935 DOI: 10.1021/acs.jnatprod.4c00342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
In the search of new inhibitors for human coronavirus (HCoV), we screened extracts of endemic Annonaceae plants on an assay using a cellular model of Huh-7 cells infected with the human alphacoronavirus HCoV-229E. The EtOAc bark extract of the rare Southeast Asian plant Neo-uvaria foetida exhibited inhibition of HCoV-229E and SARS-CoV-2 viruses with IC50 values of 3.8 and 7.8 μg/mL, respectively. Using LC-MS/MS and molecular networking analysis guided isolation, we discovered two new labdane-type diterpenoids, 8-epi-acuminolide (1) and foetidalabdane A (4), and three known labdane diterpenoids, acuminolide (2), 17-O-acetylacuminolide (3), and spiroacuminolide (5). A new norlabdane diterpene, 16-foetinorlabdoic acid (6), was also isolated and identified. Excluding compounds 5 and 6, all other metabolites were active against the virus HCoV-229E. Terpenoids 1 and 4 presented antiviral activity against SARS-CoV-2 with IC50 values of 63.3 and 93.5 μM, respectively, indicating lower potency. Additionally, virological assays demonstrated that compounds 1, 2, and 3 exert antiviral effects against Zika virus by specifically interfering with the late stage of its infectious cycle with IC50 values of 76.0, 31.9, and 14.9 μM, respectively.
Collapse
Affiliation(s)
- S Yaallini Sukumaran
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Charline Herrscher
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, Sainte Clotilde 94791, France
| | - Nurulfazlina Edayah Rasol
- Atta-ur-Rahman Institute for Natural Product Discovery, Level 9 FF3, Universiti Teknologi MARA Puncak Alam Campus, Bandar Puncak Alam 42300, Malaysia
| | - Muhamad Aqmal Othman
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Sook Yee Liew
- Chemistry Division, Centre for Foundation Studies in Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, Level 9 FF3, Universiti Teknologi MARA Puncak Alam Campus, Bandar Puncak Alam 42300, Malaysia
| | - Karin Séron
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017-Center for Infection and Immunity of Lille (CIIL), Lille F-59000, France
| | - Marc Litaudon
- Institut de Chimie des Substances Naturelles, CNRS, UPR 2301, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Khalijah Awang
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Chaker El Kalamouni
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, Sainte Clotilde 94791, France
| | - Cécile Apel
- Institut de Chimie des Substances Naturelles, CNRS, UPR 2301, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Azeana Zahari
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
39
|
Tayer AH, Jahromi HK, Kamravan M, Farhangdoost F, Ahmadi T, Kolaei M. Evaluation of circulating microvesicles and their procoagulant activity in patients with COVID-19. BMC Res Notes 2024; 17:233. [PMID: 39175048 PMCID: PMC11342662 DOI: 10.1186/s13104-024-06875-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024] Open
Abstract
OBJECTIVE Several pathological conditions trigger the formation of microvesicles (MVs), including infectious diseases such as COVID-19. The shedding of MVs increases the levels of inflammatory factors (e.g., interleukin-6; IL-6) and ultimately leads to an inflammatory cascade response, while also increasing the procoagulant response. The current study aimed to evaluate the level of circulating MVs and their procoagulant activity as well as the serum level of IL-6 in patients with COVID-19 and healthy controls. In this case-control study, 65 patients with COVID-19 and 30 healthy individuals were sampled after obtaining written informed consent. MVs counting was measured using conjugated CD61, CD45, CD235a, and Annexin-V antibodies. Additionally, the procoagulant activity of MVs and the IL-6 level were estimated using enzyme-linked immunosorbent assay (ELISA). RESULTS The majority of MVs were platelet-derived MVs (PMVs). Patients with COVID-19 had significantly higher levels of MVs, procoagulant MVs, and IL-6 compared to healthy controls (p < 0.001). MVs were significantly correlated with procoagulant MVs, D-Dimer levels, fibrinogen, and IL-6, but not with platelet, lymphocyte, and neutrophil counts. CONCLUSION Elevated levels of procoagulant MVs and their association with inflammatory and coagulation markers in patients with COVID-19 are suggested as a novel circulatory biomarker to evaluate and predict the procoagulant activity and severity of COVID-19.
Collapse
Affiliation(s)
- Akbar Hashemi Tayer
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Hossein Kargar Jahromi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Maryam Kamravan
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Farzad Farhangdoost
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Tara Ahmadi
- Student Research Committee, Jahrom University of Medical Sciences, Jahrom, Iran
| | | |
Collapse
|
40
|
Yan C, Zhou W, Zhang Y, Zhou X, Qiao Q, Miao L, Xu Z. Super-Resolution Imaging Reveals the Mechanism of Endosomal Acidification Inhibitors Against SARS-CoV-2 Infection. Chembiochem 2024; 25:e202400404. [PMID: 38877380 DOI: 10.1002/cbic.202400404] [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: 05/02/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/16/2024]
Abstract
In this study, super-resolution structured illumination microscope (SIM) was used to analyze molecular mechanism of endocytic acidification inhibitors in the SARS-CoV-2 pandemic, such as Chloroquine (CQ), Hydroxychloroquine (HCQ) and Bafilomycin A1 (BafA1). We fluorescently labeled the SARS-CoV-2 RBD and its receptor ACE2 protein with small molecule dyes. Utilizing SIM imaging, the real-time impact of inhibitors (BafA1, CQ, HCQ, Dynasore) on the RBD-ACE2 endocytotic process was dynamically tracked in living cells. Initially, the protein activity of RBD and ACE2 was ensured after being labeled. And then our findings revealed that these inhibitors could inhibit the internalization and degradation of RBD-ACE2 to varying degrees. Among them, 100 nM BafA1 exhibited the most satisfactory endocytotic inhibition (~63.9 %) and protein degradation inhibition (~97.7 %). And it could inhibit the fusion between endocytic vesicles in the living cells. Additionally, Dynasore, a widely recognized dynein inhibitor, also demonstrated cell acidification inhibition effects. Together, these inhibitors collectively hinder SARS-CoV-2 infection by inhibiting both the viral internalization and RNA release. The comprehensive evaluation of pharmacological mechanisms through super-resolution fluorescence imaging has laid a crucial theoretical foundation for the development of potential drugs to treat COVID-19.
Collapse
Affiliation(s)
- Chunyu Yan
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Wei Zhou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Yan Zhang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Xuelian Zhou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Qinglong Qiao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Lu Miao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Zhaochao Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
- School of Chemistry, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| |
Collapse
|
41
|
Alqahtani SAM. Mucosal immunity in COVID-19: a comprehensive review. Front Immunol 2024; 15:1433452. [PMID: 39206184 PMCID: PMC11349522 DOI: 10.3389/fimmu.2024.1433452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Mucosal immunity plays a crucial role in defending against coronaviruses, particularly at respiratory sites, serving as the first line of defense against viral invasion and replication. Coronaviruses have developed various immune evasion strategies at the mucosal immune system, hindering the recognition of infected cells and evading antibody responses. Understanding the immune mechanisms and responses is crucial for developing effective vaccines and therapeutics against coronaviruses. The role of mucosal immunity in COVID-19 is significant, influencing both local and systemic immune responses to the virus. Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Mucosally delivered vaccines and those under clinical trials are being compared and contrasted to understand their effectiveness in inducing mucosal immunity against coronaviruses. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community-acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality. This comprehensive review article outlines the current evidence about the role of mucosal immune responses in the clearance of SARS-CoV-2 infection, as well as potential mucosal mechanisms of protection against (re-)infection. It also proposes that there is a significant role for mucosal immunity and for secretory as well as circulating IgA antibodies in COVID-19, and that it is important to elucidate this in order to comprehend especially the asymptomatic and mild states of the infection, which appear to account for the majority of cases. Moreover, it is possible that mucosal immunity can be exploited for beneficial diagnostic, therapeutic, or prophylactic purposes. The findings from recent studies on mucosal immunity in COVID-19 can be used to develop effective vaccines and treatments that can effectively target both mucosal and systemic immune responses.
Collapse
|
42
|
Agafina A, Aguiar VC, Rossovskaya M, Fartoukh MS, Hajjar LA, Thiéry G, Timsit JF, Gordeev I, Protsenko D, Carbone J, Pellegrini R, Stadnik CMB, Avdeev S, Ferrer M, Heinz CC, Häder T, Langohr P, Bobenhausen I, Schüttrumpf J, Staus A, Ruehle M, Weissmüller S, Wartenburg-Demand A, Torres A. Efficacy and safety of trimodulin in patients with severe COVID-19: results from a randomised, placebo-controlled, double-blind, multicentre, phase II trial (ESsCOVID). Eur J Med Res 2024; 29:418. [PMID: 39138518 PMCID: PMC11321023 DOI: 10.1186/s40001-024-02008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Trimodulin (human polyvalent immunoglobulin [Ig] M ~ 23%, IgA ~ 21%, IgG ~ 56% preparation) has previously been associated with a lower mortality rate in a subpopulation of patients with severe community-acquired pneumonia on invasive mechanical ventilation (IMV) and with clear signs of inflammation. The hypothesis for the ESsCOVID trial was that trimodulin may prevent inflammation-driven progression of severe coronavirus disease 2019 (COVID-19) to critical disease or even death. METHODS Adults with severe COVID-19 were randomised to receive intravenous infusions of trimodulin or placebo for 5 consecutive days in addition to standard of care. The primary efficacy endpoint was a composite of clinical deterioration (Days 6-29) and 28-day all-cause mortality (Days 1-29). RESULTS One-hundred-and-sixty-six patients received trimodulin (n = 84) or placebo (n = 82). Thirty-three patients died, nine during the treatment phase. Overall, 84.9% and 76.5% of patients completed treatment and follow-up, respectively. The primary efficacy endpoint was reported in 33.3% of patients on trimodulin and 34.1% of patients on placebo (P = 0.912). No differences were observed in the proportion of patients recovered on Day 29, days of invasive mechanical ventilation, or intensive care unit-free days. Rates of treatment-emergent adverse events were comparable. A post hoc analysis was conducted in patients with early systemic inflammation by excluding those with high CRP (> 150 mg/L) and/or D-dimer (≥ 3 mg/L) and/or low platelet counts (< 130 × 109/L) at baseline. Forty-seven patients in the trimodulin group and 49 in the placebo group met these criteria. A difference of 15.5 percentage points in clinical deterioration and mortality was observed in favour of trimodulin (95% confidence interval: -4.46, 34.78; P = 0.096). CONCLUSION Although there was no difference in the primary outcome in the overall population, observations in a subgroup of patients with early systemic inflammation suggest that trimodulin may have potential in this setting that warrants further investigation. ESSCOVID WAS REGISTERED PROSPECTIVELY AT CLINICALTRIALS.GOV ON OCTOBER 6, 2020.: NCT04576728.
Collapse
Affiliation(s)
| | | | | | - Muriel Sarah Fartoukh
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Intensive Réanimation, Hôpital Tenon, and DMU APPROCHES, Sorbonne Université, Paris, France
| | - Ludhmila Abrahao Hajjar
- Instituto Do Coração InCor, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Jean-François Timsit
- Medical and Infectious Diseases ICU (M12) APHP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | | | - Javier Carbone
- Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | | | | | - Sergey Avdeev
- First Moscow State Medical University, Moscow, Russia
| | - Miquel Ferrer
- Hospital Clinic of Barcelona, IDIBAPS, CibeRes (CB06/06/0028) University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | - Antoni Torres
- Respiratory and Intensive Care Unit, Hospital Clinic of Barcelona, IDIBAPS, CibeRes (CB06/06/0028), University of Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Fanelli M, Petrone V, Chirico R, Radu CM, Minutolo A, Matteucci C. Flow cytometry for extracellular vesicle characterization in COVID-19 and post-acute sequelae of SARS-CoV-2 infection. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:417-437. [PMID: 39697632 PMCID: PMC11648478 DOI: 10.20517/evcna.2024.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 12/20/2024]
Abstract
Infection with SARS-CoV-2, the virus responsible for COVID-19 diseases, can impact different tissues and induce significant cellular alterations. The production of extracellular vesicles (EVs), which are physiologically involved in cell communication, is also altered during COVID-19, along with the dysfunction of cytoplasmic organelles. Since circulating EVs reflect the state of their cells of origin, they represent valuable tools for monitoring pathological conditions. Despite challenges in detecting EVs due to their size and specific cellular compartment origin using different methodologies, flow cytometry has proven to be an effective method for assessing the role of EVs in COVID-19. This review summarizes the involvement of plasmatic EVs in COVID-19 patients and individuals with Long COVID (LC) affected by post-acute sequelae of SARS-CoV-2 infection (PASC), highlighting their dual role in exerting both pro- and antiviral effects. We also emphasize how flow cytometry, with its multiparametric approach, can be employed to characterize circulating EVs, particularly in infectious diseases such as COVID-19, and suggest their potential role in chronic impairments during post-infection.
Collapse
Affiliation(s)
- Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Rossella Chirico
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
| | - Claudia Maria Radu
- Department of Medicine - DIMED, Thrombotic and Hemorrhagic Diseases Unit, University of Padua, Padua 35128 Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
- Authors contributed equally
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome 00133, Italy
- Authors contributed equally
| |
Collapse
|
44
|
Maqbool M, Hussain MS, Shaikh NK, Sultana A, Bisht AS, Agrawal M. Noncoding RNAs in the COVID-19 Saga: An Untold Story. Viral Immunol 2024; 37:269-286. [PMID: 38968365 DOI: 10.1089/vim.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024] Open
Affiliation(s)
- Mudasir Maqbool
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M. Padalia Pharmacy College, Ahmedabad, India
| | - Ayesha Sultana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya University, Mangalore, India
| | - Ajay Singh Bisht
- Shri Guru Ram Rai University School of Pharmaceutical Sciences, Dehradun, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K. R. Mangalam University, Gurugram, India
| |
Collapse
|
45
|
He M, Wang Z, Xiang D, Sun D, Chan YK, Ren H, Lin Z, Yin G, Deng Y, Yang W. A H₂S-Evolving Alternately-Catalytic Enzyme Bio-Heterojunction with Antibacterial and Macrophage-Reprogramming Activity for All-Stage Infectious Wound Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405659. [PMID: 38943427 DOI: 10.1002/adma.202405659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/24/2024] [Indexed: 07/01/2024]
Abstract
The disorder of the macrophage phenotype and the hostile by-product of lactate evoked by pathogenic infection in hypoxic deep wound inevitably lead to the stagnant skin regeneration. In this study, hydrogen sulfide (H2S)-evolving alternately catalytic bio-heterojunction enzyme (AC-BioHJzyme) consisting of CuFe2S3 and lactate oxidase (LOD) named as CuFe2S3@LOD is developed. AC-BioHJzyme exhibits circular enzyme-mimetic antibacterial (EMA) activity and macrophage re-rousing capability, which can be activated by near-infrared-II (NIR-II) light. In this system, LOD exhausts lactate derived from bacterial anaerobic respiration and generated hydrogen peroxide (H2O2), which provides an abundant stock for the peroxidase-mimetic activity to convert the produced H2O2 into germicidal •OH. The GPx-mimetic activity endows AC-BioHJzyme with a glutathione consumption property to block the antioxidant systems in bacterial metabolism, while the O2 provided by the CAT-mimetic activity can generate 1O2 under the NIR-II irradiation. Synchronously, the H2S gas liberated from CuFe2S3@LOD under the infectious micromilieu allows the reduction of Fe(III)/Cu(II) to Fe(II)/Cu(І), resulting in sustained circular EMA activity. In vitro and in vivo assays indicate that the CuFe2S3@LOD AC-BioHJzyme significantly facilitates the infectious cutaneous regeneration by killing bacteria, facilitating epithelialization/collagen deposition, promoting angiogenesis, and reprogramming macrophages. This study provides a countermeasure for deep infectious wound healing via circular enzyme-mimetic antibiosis and macrophage re-rousing.
Collapse
Affiliation(s)
- Miaomiao He
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Zuyao Wang
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Danni Xiang
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Dan Sun
- Department Advanced Composite Research Group (ACRG), School of Mechanical and Aerospace Engineering, Queen's University Belfast, Belfast, BT9 5AH, UK
| | - Yau Kai Chan
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, Hong Kong SAR, 999077, China
| | - Huilin Ren
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhijie Lin
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Guangfu Yin
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yi Deng
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, Hong Kong SAR, 999077, China
| | - Weizhong Yang
- College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
46
|
van der Straten K, Guerra D, Kerster G, Claireaux M, Grobben M, Schriek AI, Boyd A, van Rijswijk J, Tejjani K, Eggink D, Beaumont T, de Taeye SW, de Bree GJ, Sanders RW, van Gils MJ. Primary SARS-CoV-2 variant of concern infections elicit broad antibody Fc-mediated effector functions and memory B cell responses. PLoS Pathog 2024; 20:e1012453. [PMID: 39146376 PMCID: PMC11349224 DOI: 10.1371/journal.ppat.1012453] [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: 04/22/2024] [Revised: 08/27/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024] Open
Abstract
Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by human sera is a strong correlate of protection against symptomatic and severe Coronavirus Disease 2019 (COVID-19). The emergence of antigenically distinct SARS-CoV-2 variants of concern (VOCs) and the relatively rapid waning of serum antibody titers, however, raises questions about the sustainability of serum protection. In addition to serum neutralization, other antibody functionalities and the memory B cell (MBC) response are suggested to help maintaining this protection. In this study, we investigate the breadth of spike (S) protein-specific serum antibodies that mediate effector functions by interacting with Fc-gamma receptor IIa (FcγRIIa) and FcγRIIIa, and of the receptor binding domain (RBD)-specific MBCs, following a primary SARS-CoV-2 infection with the D614G, Alpha, Beta, Gamma, Delta, Omicron BA.1 or BA.2 variant. Irrespectively of the variant causing the infection, the breadth of S protein-specific serum antibodies that interact with FcγRIIa and FcγRIIIa and the RBD-specific MBC responses exceeded the breadth of serum neutralization, although the Alpha-induced B cell response seemed more strain-specific. Between VOC groups, both quantitative and qualitative differences in the immune responses were observed, suggesting differences in immunogenicity. Overall, this study contributes to the understanding of protective humoral and B cell responses in the light of emerging antigenically distinct VOCs, and highlights the need to study the immune system beyond serum neutralization to gain a better understanding of the protection against emerging variants.
Collapse
Affiliation(s)
- Karlijn van der Straten
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Denise Guerra
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Gius Kerster
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Mathieu Claireaux
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Marloes Grobben
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Angela I. Schriek
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Anders Boyd
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Stichting HIV monitoring, Amsterdam, the Netherlands
| | - Jacqueline van Rijswijk
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Khadija Tejjani
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Dirk Eggink
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Tim Beaumont
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Steven W. de Taeye
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Godelieve J. de Bree
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Amsterdam UMC, location Academic Medical Center, Department of Internal Medicine, Amsterdam, The Netherlands
| | - Rogier W. Sanders
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Marit J. van Gils
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
47
|
da Silva CS, Martinelli KG, Viana MWM, Soares DDS, Corrêa YGS, da Silva LL, de Paula VS, Rodrigues LLS, Villar LM. Liver and Inflammatory Biomarkers Are Related to High Mortality in Hospitalized Patients with COVID-19 in Brazilian Amazon Region. Life (Basel) 2024; 14:869. [PMID: 39063622 PMCID: PMC11277996 DOI: 10.3390/life14070869] [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: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 07/28/2024] Open
Abstract
COVID-19 is a multisystem disease with many clinical manifestations, including liver damage and inflammation. The objective of this study is to analyze inflammation biomarkers in relation to the clinical outcome and respiratory symptoms of COVID-19. This is a retrospective cohort of patients with COVID-19 admitted to the Hospital Regional do Baixo Amazonas from 2020 to 2022. Data were collected from electronic medical records from admission to the 30th day of hospitalization and soon after hospital discharge. A total of 397 patients were included in the study. In the longitudinal follow-up of liver markers, a significant difference was found for AST on day 14, with a higher median in the death group. Among the hematological markers, lymphopenia was observed throughout the follow-up, with the death group having the most altered values. When comparing the evolution of biomarkers in the Non-Invasive Ventilation (NIV) and Invasive Mechanical Ventilation (IMV) groups, AST showed a significant difference only on day 14 and GGT on day 1, being greater in the IMV group, and indirect bilirubin on day 7 being more altered in the NIV group. In conclusion, death during hospitalization or a more severe form of COVID-19 was related to significant changes in liver and inflammatory biomarkers.
Collapse
Affiliation(s)
- Carla Sousa da Silva
- Programa de Pós-Graduação em Ciências da Saúde, Instituto de Saúde Coletiva, Universidade Federal do Oeste do Pará (UFOPA), Santarém 66075-110, Brazil; (C.S.d.S.); (L.L.S.R.)
- Laboratório de Hepatites Virais, Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil;
| | | | | | - Deliane dos Santos Soares
- Residência Multiprofissional em Estratégia Saúde da Família para as Populações do Baixo Amazonas, Instituto de Saúde Coletiva, Universidade Federal do Oeste do Pará (UFOPA), Santarém 66075-110, Brazil;
| | - Yasmin Garcia Silva Corrêa
- Instituto de Biodiversidade e Florestas, Universidade Federal do Oeste do Pará (UFOPA), Santarém 66075-110, Brazil;
| | - Lucas Lima da Silva
- Laboratório de Hepatites Virais, Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil;
| | - Vanessa Salete de Paula
- Laboratório de Virologia e Parasitologia Molecular, Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil;
| | - Luana Lorena Silva Rodrigues
- Programa de Pós-Graduação em Ciências da Saúde, Instituto de Saúde Coletiva, Universidade Federal do Oeste do Pará (UFOPA), Santarém 66075-110, Brazil; (C.S.d.S.); (L.L.S.R.)
- Laboratório de Aids e Imunologia Molecular, Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil
| | - Livia Melo Villar
- Laboratório de Hepatites Virais, Instituto Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro 21040-360, Brazil;
| |
Collapse
|
48
|
Suarez-Castillejo C, Calvo N, Preda L, Córdova Díaz R, Toledo-Pons N, Martínez J, Pons J, Vives-Borràs M, Pericàs P, Ramón L, Iglesias A, Cànaves-Gómez L, Valera Felices JL, Morell-García D, Núñez B, Sauleda J, Sala-Llinàs E, Alonso-Fernández A. Cardiopulmonary Complications after Pulmonary Embolism in COVID-19. Int J Mol Sci 2024; 25:7270. [PMID: 39000378 PMCID: PMC11242326 DOI: 10.3390/ijms25137270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/14/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
Although pulmonary embolism (PE) is a frequent complication in COVID-19, its consequences remain unknown. We performed pulmonary function tests, echocardiography and computed tomography pulmonary angiography and identified blood biomarkers in a cohort of consecutive hospitalized COVID-19 patients with pneumonia to describe and compare medium-term outcomes according to the presence of PE, as well as to explore their potential predictors. A total of 141 patients (56 with PE) were followed up during a median of 6 months. Post-COVID-19 radiological lung abnormalities (PCRLA) and impaired diffusing capacity for carbon monoxide (DLCOc) were found in 55.2% and 67.6% cases, respectively. A total of 7.3% had PE, and 6.7% presented an intermediate-high probability of pulmonary hypertension. No significant difference was found between PE and non-PE patients. Univariate analysis showed that age > 65, some clinical severity factors, surfactant protein-D, baseline C-reactive protein, and both peak red cell distribution width and Interleukin (IL)-10 were associated with DLCOc < 80%. A score for PCRLA prediction including age > 65, minimum lymphocyte count, and IL-1β concentration on admission was constructed with excellent overall performance. In conclusion, reduced DLCOc and PCRLA were common in COVID-19 patients after hospital discharge, but PE did not increase the risk. A PCRLA predictive score was developed, which needs further validation.
Collapse
Affiliation(s)
- Carla Suarez-Castillejo
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Néstor Calvo
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luminita Preda
- Servicio de Radiodiagnóstico, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Rocío Córdova Díaz
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Nuria Toledo-Pons
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Joaquín Martínez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Pons
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Miquel Vives-Borràs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
| | - Pere Pericàs
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Cardiología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Luisa Ramón
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Amanda Iglesias
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Cànaves-Gómez
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jose Luis Valera Felices
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Morell-García
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Servicio de Análisis Clínicos, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
| | - Belén Núñez
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
| | - Jaume Sauleda
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ernest Sala-Llinàs
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alberto Alonso-Fernández
- Servicio de Neumología, Hospital Universitario Son Espases, 07120 Palma de Mallorca, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma de Mallorca, Spain
- Facultad de Medicina, Universidad de las Islas Baleares, 07122 Palma, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
49
|
Du Z, Zhou Q, Shen Y, Lu R, Jiang Y, Zhu H. Analysis of antimicrobial drug usage in psychiatric specialized hospitals during the pandemic in China. Expert Rev Anti Infect Ther 2024; 22:597-602. [PMID: 38696326 DOI: 10.1080/14787210.2024.2351022] [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: 07/19/2023] [Accepted: 03/13/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND This study aims to compare antimicrobial drug usage in our hospital to Jiangsu Province and China from 2020 to 2022. RESEARCH DESIGN AND METHODS A detailed analysis was performed using data from the National Antimicrobial Drug Clinical Application Monitoring Network. Several parameters were studied: the rate of antimicrobial drug use, number and types of drugs used, the rate of combined use, rate of microbiological examinations, drug use intensity, and cumulative Defined Daily Doses (DDDs). RESULTS From 2020 to 2022, our hospital's antimicrobial drug usage rate was consistently lower than Jiangsu Province and China. The average number of drug types and the combined drug use rate were higher in 2020 but fell below those in Jiangsu Province and China in 2021 and 2022. Our microbiological examination rate consistently surpassed that of Jiangsu Province and China. Furthermore, our Antimicrobial Usage Density and cumulative DDDs were notably lower. While AUD remained stable, DDDs showed a decreasing trend. The most dominant drug in DDDs was cefditoren, a third-generation cephalosporin. CONCLUSIONS During the pandemic years, our hospital not only met the requirements for antimicrobial drug usage, microbiological examination, AUD, and cumulative DDDs but also demonstrated a consistent year-by-year decrease in drug usage and DDDs.
Collapse
Affiliation(s)
- Zhiqiang Du
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Qin Zhou
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Yuan Shen
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Rongrong Lu
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Ying Jiang
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Haohao Zhu
- Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
50
|
Chen C, Hu H, Li Z, Qi M, Qiu Y, Hu Z, Feng F, Tang W, Diao H, Sun W, Tang Z. Dietary tryptophan improves growth and intestinal health by promoting the secretion of intestinal β-defensins against enterotoxigenic Escherichia coli F4 in weaned piglets. J Nutr Biochem 2024; 129:109637. [PMID: 38574828 DOI: 10.1016/j.jnutbio.2024.109637] [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: 11/23/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Adequate dietary L-tryptophan (Trp) governs intestinal homeostasis in piglets. However, the defensive role of Trp in the diet against enterotoxigenic Escherichia coli F4 (K88) in pigs is still poorly understood. Here, sixty (6.15 ± 1.52 kg, 24-day-old, Duroc × Landrace × Yorkshire) weaned piglets were used for an E. coli F4 attack test in a 2 × 2 factorial design. The growth (ADG, ADFI, GH), immune factors (IL-10, IgA, IgG, IgM), Trp metabolite 5-HT, intestinal morphology (jejunal and colonic VH), mRNA expression of β-defensins (jejunal BD-127, BD-119, ileal BD-1, BD-127), and abundance of beneficial microorganisms in the colon (Prevotella 9, Lactobacillus, Phascolarctobacterium, Faecalibacterium) were higher in the piglets in the HT (High Trp) and HTK (High Trp, K88) groups than in the LT (Low Trp) and LTK (Low Trp, K88) groups (P<.05), while FCR, diarrhea rate, diarrhea index, serum Trp, Kyn, IDO, D-LA, ET, and abundance of harmful microorganisms in the colon (Spirochaetes, Fusobacteria, Prevotella, Christensenellaceae R7) were lower in the HT and HTK groups than in the LT and LTK groups (P<.05). High Trp reduced the expression of virulence genes (K88 and LT) after E. coli F4 attack (P<.05). The IL-6, TNF-α was lower in the HTK group than in the LT, LTK group (P<.05). In short, a diet containing 0.35% Trp protected piglets from enterotoxigenic E. coli F4 (K88) via Trp metabolism promoting BD expression in the intestinal mucosa, which improved growth and intestinal health.
Collapse
Affiliation(s)
- Chen Chen
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Hong Hu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhangcheng Li
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Min Qi
- Yunnan Animal Husbandry Station, Kunming 650225, China
| | - Yibin Qiu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhijin Hu
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Fu Feng
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China; Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtche Group Co., Ltd., Chengdu 610066, China
| | - Weizhong Sun
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Zhiru Tang
- Animal Nutrition and Bio-feed, Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|