1
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [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: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
2
|
Muştucu A, Güllülü RA, Cekic S, Kilic SS, Kırlı S. Evaluation of the effect of the COVID-19 pandemic on depression, anxiety and psychological resilience in patients with primary immunodeficiency. BMC Immunol 2025; 26:39. [PMID: 40389841 PMCID: PMC12087040 DOI: 10.1186/s12865-025-00721-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/08/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Primary immunodeficiencies (PIDs) are a group of diseases that develop as a result of primary or congenital malfunction of the immune system and progress with chronic and/or recurrent bacterial, fungal, protozoal and/or viral infections. In this study, we aimed to examine the effects of the COVID-19 pandemic on depression, anxiety levels and psychological resilience in patients with PID and to compare them with those in controls. METHODS Seventy patients, aged 18-65 years, who were being followed up with a diagnosis of PID and 69 people as healthy control group, participated in our study. The participants were evaluated cross-sectionally once; sociodemographic data form, Hamilton Depression Rating Scale (HAM-D), Hamilton Anxiety Rating Scale (HAM-A), Resilience Scale for Adults (RSA), and COVID-19 Evaluation form were administered to the participants. RESULTS HAM-A and HAM-D scores were significantly higher in PID patients compared to controls (HAM-D: 5.5 vs. 3.0, p < 0.001; HAM-A: 6.0 vs. 4.0, p = 0.008). RSA was significantly lower in the patient group (RSA total: 122.5 vs. 136.0, p < 0.001), and pandemic-related risk perception was higher (PRPS: 33.9 vs. 28.3, p < 0.001). Sleep, appetite, and attention-related disturbances were also more common in the patient group. Multivariate regression analyses revealed that PID diagnosis was an independent predictor of increased depression severity (HAM-D), lower psychological resilience (RSA), and greater pandemic-related risk perception. Female sex was independently associated with higher anxiety severity (HAM-A). A personal psychiatric history and greater number of comorbidities were also significant predictors of psychological vulnerability, particularly in relation to depression and anxiety. CONCLUSION Given the observed associations between PID and increased levels of depression, anxiety, and reduced psychological resilience during the pandemic, clinicians may consider heightened vigilance for psychological symptoms in this population during times of public health crisis.
Collapse
Affiliation(s)
- Anıl Muştucu
- Department of Psychiatry, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey.
| | - Rümeysa Ayşe Güllülü
- Department of Psychiatry, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Sukru Cekic
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Sara Sebnem Kilic
- Division of Pediatric Allergy and Clinical Immunology, Department of Pediatrics, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Selçuk Kırlı
- Department of Psychiatry, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
3
|
Almakrami M, Bazuqamah M, A. Alshehri M, M. S. Alqahtani A, F. Kadasah S, Harthi N, Ali Alyami R, Alqurashi A, A. Al Ruwaithi A. Identification of Significant Mutations in Spike Protein of SARS-CoV-2 Variants of Concern and the Discovery of Potent Inhibitors. Glob Health Epidemiol Genom 2025; 2025:5042190. [PMID: 40330793 PMCID: PMC12052452 DOI: 10.1155/ghe3/5042190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Background: SARS-CoV-2 is a positive-sense single-stranded RNA virus that has a propensity for infecting epithelial cells and the respiratory system. The two important proteins, structural and nonstructural proteins, make the architecture of this virus. Aim: This research aimed at studying significant mutations in spike protein of SARS-CoV-2 variants of concern (VoCs) and finding shared mutations among omicron and other four variants (alpha, beta, gamma, and delta). The purpose of this study was to draw structural comparisons between wild type and mutant proteins, followed by identifying potent inhibitors (ligand) that could be used against SARS-CoV-2 spike protein and its latest omicron VoC. Methodology: In this research, we had studied 16 major mutations as well as shared mutations (6) present in spike region of SARS-CoV-2. Subsequently, we determined the structure of the wild-type SARS-CoV-2 protein from the Protein Data Bank (PDB) with the ID 7R4I. Furthermore, the structure of the mutant protein of SARS-CoV-2 omicron variant was modeled in SWISS-MODEL. The ligand dataset for spike protein of SARS-CoV-2 was also collected from literature and different databases. Both wild type and mutant proteins were docked with ligand database in Molecular Operating Environment (MOE). The docking analysis was performed, and two best ligand molecules, AZ_2 and AZ_13, were finalized based on their energy values, interactions, and docking scores to be used against our wild and mutant proteins. Results: AZ_2 demonstrated a docking score of -6.1753 in MOE, with energy values of -4.3889 and -6.1753. It formed key hydrogen bond interactions. AZ_13 showed a docking score of -5.9, with energy values of -9.3 and -5.9, forming hydrogen donor and acceptor interactions with Asp950 (3.06 Å), Ile312 (3.13 Å), and Glu309 (3.27 Å). These interactions suggest strong binding affinity and potential efficacy. Thus, present research work emphasized on identification of significant mutations and finding a potent target-based drug against SARS-CoV-2 and its omicron variant. Outcomes: Based on this computational analysis performed, it is suggested that proposed compound can be used as remedy against SARS-CoV-2 and its omicron variant.
Collapse
Affiliation(s)
- Mohsen Almakrami
- Department of Pathology and Laboratory Medicine, King Khaled Hospital, Najran 66262, Saudi Arabia
| | - Mohammed Bazuqamah
- Department of Pathology and Laboratory Medicine, King Khaled Hospital, Najran 66262, Saudi Arabia
| | - Mohammed A. Alshehri
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdulaziz M. S. Alqahtani
- Department of Biology, Faculty of Science, University of Bisha, P.O. Box 551, Bisha 61922, Saudi Arabia
| | - Sultan F. Kadasah
- Department of Biology, Faculty of Science, University of Bisha, P.O. Box 551, Bisha 61922, Saudi Arabia
| | - Naif Harthi
- Emergency Medical Services, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Rami Ali Alyami
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Abdulmajeed Alqurashi
- Department of Biology, College of Science, Taibah University, Madinah 42353, Saudi Arabia
| | | |
Collapse
|
4
|
Souza HDS, Martins JSCC, Sousa TDC, Sardar S, Fintelman-Rodrigues N, Silva-Trujillo L, Souza TMLE, Siqueira MM, Fernandes JH, Matos ADR. Hypericin Suppresses SARS-CoV-2 Replication and Synergizes with Antivirals via Dual Targeting of RdRp and 3CLpro. Microorganisms 2025; 13:1004. [PMID: 40431177 PMCID: PMC12114490 DOI: 10.3390/microorganisms13051004] [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: 02/11/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
The continuous emergence of SARS-CoV-2 variants underscores the need for novel antiviral candidates. Hypericin (HY), a compound derived from Hypericum perforatum, exhibited potent in vitro activity against SARS-CoV-2 in Vero E6 cells, with low cytotoxicity (CC50 > 200 nM). HY showed no significant activity against Influenza A (H1N1) or dengue virus serotype 2, supporting its selective action. Antiviral effects were most evident when HY was administered post-infection, in a concentration-dependent manner, while cellular pretreatment or viral pre-incubation produced limited effects. Notably, HY also displayed virucidal activity, significantly reducing viral titers at 4 °C, 22 °C, and 37 °C. Combination treatments with remdesivir or nirmatrelvir enhanced antiviral efficacy by 50-70% relative to monotherapy, depending on compound concentration. Molecular simulations revealed stable interactions with conserved residues in RdRp and 3CLpro, suggesting a low risk of resistance. Together, these findings highlight the potential of HY as a selective antiviral and virucidal agent against SARS-CoV-2, particularly in combination regimens.
Collapse
Affiliation(s)
- Helena da Silva Souza
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Jéssica Santa Cruz Carvalho Martins
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Thiagos das Chagas Sousa
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Saiqa Sardar
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Natalia Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Lina Silva-Trujillo
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Thiago Moreno Lopes e Souza
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias, Oswaldo Cruz Institute, Rio de Janeiro 21040-361, Brazil; (N.F.-R.); (L.S.-T.); (T.M.L.e.S.)
- Center for Technological Development in Health, National Institute for Science and Technology on Innovation on Neglected Diseases Neglected Populations, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil
| | - Marilda Mendonça Siqueira
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| | - Jorge Hernandes Fernandes
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense (UENF), Campos dos Goytacazes 28013-602, Brazil
| | - Aline da Rocha Matos
- Laboratory of Respiratory Viruses, Exanthematics, Enteroviruses and Vital Emergencies, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro 21040-900, Brazil; (H.d.S.S.); (J.S.C.C.M.)
| |
Collapse
|
5
|
Huang Y, Li S, Ye W, Wang H, Su J, Gao L, Shi R, Mou X, Leng SX, Xiao C, Chen G. Viral Infections in Elderly Individuals: A Comprehensive Overview of SARS-CoV-2 and Influenza Susceptibility, Pathogenesis, and Clinical Treatment Strategies. Vaccines (Basel) 2025; 13:431. [PMID: 40333344 PMCID: PMC12031201 DOI: 10.3390/vaccines13040431] [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: 02/22/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
As age increases, the immune function of elderly individuals gradually decreases, increasing their susceptibility to infectious diseases. Therefore, further research on common viral infections in the elderly population, especially severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses, is crucial for scientific progress. This review delves into the genetic structure, infection mechanisms, and impact of coinfections with these two viruses and provides a detailed analysis of the reasons for the increased susceptibility of elderly individuals to dual viral infections. We evaluated the clinical manifestations in elderly individuals following coinfections, including complications in the respiratory, gastrointestinal, nervous, and cardiovascular systems. Ultimately, we have summarized the current strategies for the prevention, diagnosis, and treatment of SARS-CoV-2 and influenza coinfections in older adults. Through these studies, we aim to reduce the risk of dual infections in elderly individuals and provide a scientific basis for the prevention, diagnosis, and treatment of age-related viral diseases, thereby improving their health status.
Collapse
Affiliation(s)
- Yanhao Huang
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wenjie Ye
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Haoyun Wang
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou 510632, China;
| | - Lijuan Gao
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Ruohu Shi
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xinyi Mou
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Sean Xiao Leng
- Johns Hopkins Center on Aging and Immune Remodeling, Division of Geriatric Medicine and Gerontology, Departments of Medicine, Molecular Microbiology and Immunology, Johns Hopkins University School of Medicine and Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Chanchan Xiao
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| | - Guobing Chen
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), School of Medicine, Jinan University, Dongguan 523000, China;
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; (S.L.); (W.Y.); (H.W.); (L.G.); (R.S.); (X.M.)
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
6
|
Vaiasicca S, James DW, Melone G, Saeed O, Francis LW, Corradetti B. Amniotic fluid-derived mesenchymal stem cells as a therapeutic tool against cytokine storm: a comparison with umbilical cord counterparts. Stem Cell Res Ther 2025; 16:151. [PMID: 40156072 PMCID: PMC11951844 DOI: 10.1186/s13287-025-04262-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Several immunosuppressive therapies have been proposed as key treatment options for critically ill patients since the first appearance of severe acute respiratory syndrome coronavirus 2. Mesenchymal stem cells (MSCs) from different sources have been considered for their potential to attenuate the cytokine storm associated to COVID-19 and the consequent multi-organ failure, providing evidence for safe and efficacious treatments. Among them, administration of umbilical cord-derived MSCs (UC-MSCs) has demonstrated a significant increase in survival rates, largely due to their potent immunosuppressive properties. METHODS We applied next-generation sequencing (NGS) analysis to compare the transcriptomic profiles of MSCs isolated from two gestational sources: amniotic fluid (AF) obtained during prenatal diagnosis and their clinically relevant umbilical cord counterparts, for which datasets were publicly available. A full meta-analysis was performed to identify suitable GEO and NGS datasets for comparison between AF- and UC-MSC samples. RESULTS Transcriptome analysis revelaed significant differences between groups, despite both cell lines being strongly involved in the tissue development, crucial to achieve the complex task of wound healing. Significantly enriched hallmark genes suggest AF-MSC superior immunomodulatory features against signaling pathways actively involved in the cytokine storm (i.e., IL-2/STAT, TNF-a/NFkB, IL-2/STAT5, PI3K/AKT/mTOR). CONCLUSIONS The data presented here suggest that AF-MSCs hold significant promise for treating not only COVID-19-associated cytokine storms but also a variety of other inflammatory syndromes (i.e., those induced by bacterial infections, autoimmune disorders, and therapeutic interventions). Realizing the full potential of AF-MSCs as a comprehensive therapeutic approach in inflammatory disease management will require more extensive clinical trials and in-depth mechanistic studies.
Collapse
Affiliation(s)
- Salvatore Vaiasicca
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
- Department of Life and Environmental Life, Polytechnic University of Marche, Ancona, Italy
| | - David W James
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Gianmarco Melone
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Omar Saeed
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Lewis W Francis
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK
| | - Bruna Corradetti
- Centre of NanoHealth, Swansea University Medical School, Swansea, UK.
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Section Oncology/Hematology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
Wen Y, Zhao J, Zhang Z. Heterogeneity and longitudinal transcriptomic characteristics of Tregs in COVID-19 patients. Front Immunol 2025; 16:1548173. [PMID: 40114921 PMCID: PMC11922936 DOI: 10.3389/fimmu.2025.1548173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance by suppressing immune responses against pathogens. The fluctuation of Treg proportions in COVID-19 remains a topic of debate, and the mechanisms triggering Treg activation in COVID-19 are still unclear. Understanding these issues is essential for better managing immune responses in COVID-19 patients. Methods We collected a cohort of COVID-19 patients with varying disease severity and stage to explore the transcriptomic and functional traits of Tregs in these individuals. Using transcriptomic analysis, we evaluated the proportion and functionality of different Treg subsets, specifically HLA_DR+ Tregs, across different stages of COVID-19 patients. Results Our analysis revealed that the proportion of CCR7 + Tregs decreased as the disease advanced, while the cell proportion of HLA_DR+ regs escalated with the severity of the disease. Moreover, the transcription actor CARHSP1 exhibited apositive correlation with the proportion of HLA_DR+ Tregs. Notably, the heightened suppressive function of HLA_DR+ Tregs in severe COVID-19 patients, with interactions between PF4 and CXCR3, contributed to the homeostasis of HLA_DR+ Tregs in severe COVID-19 patients. Furthermore, we observed that Tregs in COVID-19 patients exhibited weakened TCR clonotype expansion, and the suppression of HLA_DR+ Tregs with expanded TCR clonotypes in severe COVID-19 cases did not show a significant increase compared to asymptomatic and mild COVID-19 groups. The findings indicate that Tregs may be activated through the bystander effect, as evidenced by the analysis of TCR clonotype characteristics. Discussion Our research delineates the diversity of dynamic alterations in Tregs and sheds light on potential mechanisms underlying Treg activation, providing a theoretical foundation and offering treatment strategies for managing COVID-19 patients.
Collapse
Affiliation(s)
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Liu J, Cong B, Cao X. Lung DC-T immunity hub in immune surveillance: new concepts and future directions. Cancer Commun (Lond) 2025; 45:209-214. [PMID: 39687995 PMCID: PMC11947608 DOI: 10.1002/cac2.12643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Juan Liu
- National Key Laboratory of Immunity and InflammationInstitute of Immunology, Naval Medical UniversityShanghaiP. R. China
| | - Boyi Cong
- Institute of Immunology, College of Life Sciences, Nankai UniversityTianjinP. R. China
- Department of ImmunologyCenter for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingP. R. China
| | - Xuetao Cao
- Institute of Immunology, College of Life Sciences, Nankai UniversityTianjinP. R. China
- Department of ImmunologyCenter for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical SciencesBeijingP. R. China
| |
Collapse
|
9
|
Chae DH, Park HS, Kim KM, Yu A, Park JH, Oh MK, Choi SW, Ryu J, Dunbar CE, Yoo HM, Yu KR. SARS-CoV-2 pseudovirus dysregulates hematopoiesis and induces inflammaging of hematopoietic stem and progenitor cells. Exp Mol Med 2025; 57:616-627. [PMID: 40025168 PMCID: PMC11958793 DOI: 10.1038/s12276-025-01416-1] [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: 03/25/2024] [Revised: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 03/04/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primarily affects the respiratory system but may induce hematological alterations such as anemia, lymphopenia and thrombocytopenia. Previous studies have reported that SARS-CoV-2 efficiently infects hematopoietic stem and progenitor cells (HSPCs); however, the subsequent effects on hematopoiesis and immune reconstitution have not yet been described. Here we evaluated the pathological effects of infection of umbilical-cord-blood-derived HSPCs with the SARS-CoV-2 Omicron variant pseudovirus (PsV). Transcriptomic analysis of Omicron PsV-infected HSPCs revealed the upregulation of genes involved in inflammation, aging and the NLRP3 inflammasome, suggesting a potential trigger of inflammaging. Omicron PsV-infected HSPCs presented decreased numbers of multipotential progenitors (granulocyte‒erythrocyte‒macrophage‒megakaryocyte colony-forming units) ex vivo and repopulated primitive hematopoietic stem cells (Ki-67-hCD34+ cells) in an HSPC transplantation NOD-scid IL2rγnull mouse model (Omicron mouse). Furthermore, Omicron PsV infection induced myeloid-biased differentiation of HSPCs. Treatment with nanographene oxide, an antiviral agent, partially mitigated the myeloid bias and inflammaging phenotype both in vitro and in vivo. These findings provide insights into the abnormal hematopoietic and immune effects of SARS-CoV-2 infection and highlight potential therapeutic interventions.
Collapse
Affiliation(s)
- Dong-Hoon Chae
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Sung Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kyoung-Myeon Kim
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Aaron Yu
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Han Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Mi-Kyung Oh
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Institutes of Convergence Technology, INBCT, Seoul, Republic of Korea
| | - Jaechul Ryu
- Institutes of Convergence Technology, INBCT, Seoul, Republic of Korea
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea.
- Department of Precision Measurement, University of Science and Technology, Daejeon, Republic of Korea.
| | - Kyung-Rok Yu
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Fratta Pasini AM, Stranieri C, Di Leo EG, Bertolone L, Aparo A, Busti F, Castagna A, Vianello A, Chesini F, Friso S, Girelli D, Cominacini L. Identification of Early Biomarkers of Mortality in COVID-19 Hospitalized Patients: A LASSO-Based Cox and Logistic Approach. Viruses 2025; 17:359. [PMID: 40143288 PMCID: PMC11946718 DOI: 10.3390/v17030359] [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: 12/05/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to identify possible early biomarkers of mortality among clinical and biochemical parameters, iron metabolism parameters, and cytokines detected within 24 h from admission in hospitalized COVID-19 patients. We enrolled 80 hospitalized patients (40 survivors and 40 non-survivors) with COVID-19 pneumonia and acute respiratory failure. The median time from the onset of COVID-19 symptoms to hospital admission was lower in non-survivors than survivors (p < 0.05). Respiratory failure, expressed as the ratio of arterial oxygen partial pressure to the fraction of inspired oxygen (P/F), was more severe in non-survivors than survivors (p < 0.0001). Comorbidities were similar in both groups. Among biochemical parameters and cytokines, eGFR and interleukin (IL)-1β were found to be significantly lower (p < 0.05), while LDH, IL-10, and IL-8 were significantly higher in non-survivors than in survivors (p < 0.0005, p < 0.05 and p < 0.005, respectively). Among other parameters, LDH values distribution showed the most significant difference between study groups (p < 0.0001). LASSO feature selection combined with Cox proportional hazards and logistic regression models was applied to identify features distinguishing between survivors and non-survivors. Both approaches highlighted LDH as the strongest predictor, with IL-22 and creatinine emerging in the Cox model, while IL-10, eGFR, and creatinine were influential in the logistic model (AUC = 0.744 for Cox, 0.723 for logistic regression). In a similar manner, we applied linear regression for predicting LDH levels, identifying the P/F ratio as the top predictor, followed by IL-10 and eGFR (NRMSE = 0.128). Collectively, these findings underscore LDH's critical role in mortality prediction, with P/F and IL-10 as key determinants of LDH increases in this Italian COVID-19 cohort.
Collapse
Affiliation(s)
- Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Edoardo Giuseppe Di Leo
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Lorenzo Bertolone
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Antonino Aparo
- Interdepartmental Laboratory of Medical Research, Research Center LURM, University of Verona, 37134 Verona, Italy;
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Fabio Chesini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| |
Collapse
|
11
|
Gao L, Xu Z, Hu J, Zhang Q, Fu S, Wang W, Xie C, Zhang Y, Wang Y, Gong F. Impact of COVID-19 infection on Kawasaki disease and immune status in children. Sci Rep 2025; 15:6417. [PMID: 39984588 PMCID: PMC11845713 DOI: 10.1038/s41598-025-91042-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/18/2025] [Indexed: 02/23/2025] Open
Abstract
Patients infected with SARS-CoV-2 may experience acute and long-term immune disorders. Immunological factors are thought to play an important role in Kawasaki disease. To analyze the impact of COVID-19 infection on Kawasaki disease, this study retrospectively analyzed 161 children with Kawasaki disease onset during the COVID-19 pandemic. The proportion of IVIG-Resistant individuals and the rate of corticosteroid use in the 1-7 weeks from COVID-19 infection to Kawasaki disease onset were higher than that of the noninfected group, even after excluding suspected cases of multiple system inflammatory syndrome. Compared to the noninfected group, the level of CD4 was lower, and the levels of CD3-CD16+CD56+, complement C4, TNF-α, and IFN-γ were higher in the 1-7 weeks after COVID-19 infection. In conclusion, the risk of IVIG resistance was significantly increased in children with Kawasaki disease onset 1-7 weeks after COVID-19 infection, which may be related to the long-term impact of COVID-19 on immunity.
Collapse
Affiliation(s)
- Lichao Gao
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Zhufei Xu
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jian Hu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Qing Zhang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Songling Fu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Wei Wang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Chunhong Xie
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Yiying Zhang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China
| | - Yujia Wang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China.
| | - Fangqi Gong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
12
|
Imtiaz K, Farooqui N, Ahmed K, Zhamalbekova A, Anwar MF, Nasir A, Ansar Z, Gul K, Hussain A, Sarría-Santamera A, Abidi SH. Analysis of differential expression of matrix metalloproteinases and defensins in the nasopharyngeal milieu of mild and severe COVID-19 cases. PLoS One 2025; 20:e0304311. [PMID: 39965032 PMCID: PMC11835293 DOI: 10.1371/journal.pone.0304311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION A subset of COVID-19 disease patients suffers a severe form of the illness; however, underlying early pathophysiological mechanisms associated with the severe form of COVID-19 disease remain to be fully understood. Several studies showed the association of COVID-19 disease severity with the changes in the expression profile of various matrix metalloproteinases (MMPs) and defensins (DA). However, the link between the changes in the expression of MMPs and DA in the nasopharyngeal milieu during early phases of infection and disease severity remains poorly understood. Therefore, we performed differential gene expression analysis of MMPs and DA in the nasopharyngeal swab samples collected from normal (COVID-19 negative), mild, and severe COVID-19 cases and examined the association between MMP and DA expression and disease severity. MATERIAL AND METHOD A total of 118 previously collected nasopharyngeal samples from mild and severe COVID-19 patients (as per the WHO criteria) and 10 healthy individuals (COVID-19 negative, controls) were used in this study. A real-time qPCR assay was used to determine the viral loads and assess the mRNA expression of MMPs and DA. One-way ANOVA was applied to perform multiple comparisons (estimate differences) in MMPs and defensin gene expression in the normal vs mild vs severe groups. In addition, a multivariable logistic regression analysis was carried out with all the variables from the data set using 'severity' as the outcome variable. RESULTS Our results showed that as compared to controls, DA1, DA3, and DA4 expression was significantly (p < 0.05) upregulated in the mild group, whereas the expression of DA6 was significantly downregulated in both mild and severe groups (p-value < 0.05). Similarly, compared to controls, the expression of MMP1 and MMP7 was significantly downregulated in both mild and severe groups, whereas MMP2 expression was upregulated in the mild group (p-value < 0.05). Additionally, the regression analysis showed that the expression of MMP1, MMP2, and MMP9 was significantly associated with the severity of the disease. CONCLUSION The early detection of changes in the expression of MMPs and defensins may act as a useful biomarker/predictor for possible severe COVID-19 disease, which may be useful in the clinical management of patients to reduce COVID-19-associated morbidity and mortality.
Collapse
Affiliation(s)
- Khekashan Imtiaz
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Nida Farooqui
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Khalid Ahmed
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Muhammad Faraz Anwar
- Department of Biochemistry, Bahria University Medical and Dental College, Karachi, Pakistan
| | - Asghar Nasir
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Zeeshan Ansar
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Khitab Gul
- Department of Biosciences, Muhammad Ali Jinnah University, Karachi, Pakistan
| | - Azhar Hussain
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Syed Hani Abidi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| |
Collapse
|
13
|
Quinn AE, Zhao L, Bell SD, Huq MH, Fang Y. Exploring Asthma as a Protective Factor in COVID-19 Outcomes. Int J Mol Sci 2025; 26:1678. [PMID: 40004141 PMCID: PMC11855143 DOI: 10.3390/ijms26041678] [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: 01/19/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Asthma has long been associated with increased susceptibility to viral respiratory infections, leading to significant exacerbations and poorer clinical outcomes. Contrarily and interestingly, emerging data and research surrounding the COVID-19 pandemic have shown that patients with asthma infected with SARS-CoV-2 experienced decreased severity of disease, lower hospitalization rates, as well as decreased morbidity and mortality. Research has shown that eosinophils could enhance immune defense against viral infections, while inhaled corticosteroids can assist in controlling systematic inflammation. Moreover, reduced ACE-2 expression in individuals with asthma may restrict viral entry, and the Th2 immune response may offset the Th1 response typically observed in severe COVID-19 patients. These factors may help explain the favorable outcomes seen in asthmatic patients during the COVID-19 pandemic. This review highlights potential protective mechanisms seen in asthmatic patients, including eosinophilia, the use of inhaled corticosteroids, reduced ACE-2 expression, and a dominate Th2 immune response. Such a study will be helpful to better manage patients with asthma who have contracted COVID-19.
Collapse
Affiliation(s)
- Anthony E. Quinn
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Muhammad H. Huq
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (A.E.Q.); (S.D.B.); (M.H.H.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
14
|
Lee C, Khan R, Mantsounga CS, Sharma S, Pierce J, Amelotte E, Butler CA, Farinha A, Parry C, Caballero O, Morrison JA, Uppuluri S, Whyte JJ, Kennedy JL, Zhang X, Choudhary G, Olson RM, Morrison AR. IL-1β-driven NF-κB transcription of ACE2 as a Mechanism of Macrophage Infection by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.24.630260. [PMID: 39763770 PMCID: PMC11703209 DOI: 10.1101/2024.12.24.630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with the enveloped RNA betacoronavirus, SARS-CoV-2, led to a global pandemic involving over 7 million deaths. Macrophage inflammatory responses impact COVID-19 severity; however, it is unclear whether macrophages are infected by SARS-CoV-2. We sought to identify mechanisms regulating macrophage expression of ACE2, the primary receptor for SARS-CoV-2, and to determine if macrophages are susceptible to productive infection. We developed a humanized ACE2 (hACE2) mouse whereby hACE2 cDNA was cloned into the mouse ACE2 locus under control of the native promoter. We validated the susceptibility of hACE2 mice to SARS-CoV-2 infection relative to wild-type mice and an established K18-hACE2 model of acute fulminating disease. Intranasal exposure to SARS-CoV-2 led to pulmonary consolidations with cellular infiltrate, edema, and hemorrhage, consistent with pneumonia, yet unlike the K18-hACE2 model, hACE2 mice survived and maintained stable weight. Infected hACE2 mice also exhibited a unique plasma chemokine, cytokine, and growth factor inflammatory signature relative to K18-hACE2 mice. Infected hACE2 mice demonstrated evidence of viral replication in infiltrating lung macrophages, and infection of macrophages in vitro revealed a transcriptional profile indicative of altered RNA and ribosomal processing machinery as well as activated cellular antiviral defense. Macrophage IL-1β-driven NF-κB transcription of ACE2 was an important mechanism of dynamic ACE2 upregulation, promoting macrophage susceptibility to infection. Experimental models of COVID-19 that make use of native hACE2 expression will allow for mechanistic insight into factors that can either promote host resilience or increase susceptibility to worsening severity of infection.
Collapse
Affiliation(s)
- Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Andrew Farinha
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Olivya Caballero
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeremi A. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeffrey J. Whyte
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Cardiovascular Research Center, Lifespan Cardiovascular Research Institute, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Lead contact and corresponding author
| |
Collapse
|
15
|
Trimbake D, Singh D, K. YG, Babar P, S. VD, Tripathy AS. Durability of Functional SARS-CoV-2-Specific Immunological Memory and T Cell Response up to 8-9 Months Postrecovery From COVID-19. J Immunol Res 2025; 2025:9743866. [PMID: 39963186 PMCID: PMC11832264 DOI: 10.1155/jimr/9743866] [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: 06/03/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025] Open
Abstract
Research on long-term follow-up in individuals who have recovered from coronavirus disease-19 (COVID-19) would yield insights regarding their immunity status and identify those who need booster vaccinations. This study evaluated the longevity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific cellular and humoral memory responses, as well as T cell effector functionalities, at 1-2 months (n = 40), 8-9 months (n = 40), and 12 months/1 year (n = 27) following recovery from SARS-CoV-2 infection. CTL response by enzyme-linked immunospot (ELISPOT); levels of cytokine by Bio-Plex, natural killer (NK), CD4+ helper, and CD8+ cytotoxic T cell functionalities using flow cytometry; anti-SARS-CoV-2 IgG by ELISA; and levels of neutralizing antibodies (NAbs) by surrogate virus NAb assay were assessed. The levels of SARS-CoV-2-specific IgG and NAb at 1-2 and 8-9 months postrecovery were hand in hand and appeared declining. SARS-CoV-2-specific B, memory B and plasma cells, and T cells sustained up to 8-9 months. Increased expression of CD107a/IFN-γ by NK cells and cytotoxic T cells at 8-9 months could be indicative of SARS-CoV-2-specific effector functions. Recovered individuals with positive and negative IgG antibody status displayed T cell response up to 1 year and 8-9 months, respectively, emphasizing the durabilty of effector immunity up to 8-9 months regardless of IgG antibody status. Overall, the recovered individuals exhibited robust immunological memory, sustained T cell response with effector functionality against SARS-CoV-2 that persists for at least 8-9 months.
Collapse
Affiliation(s)
- Diptee Trimbake
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Dharmendra Singh
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Yogesh Gurav K.
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Prasad Babar
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| | - Varsha Dange S.
- Department of Medicine, Pimpri Chinchwad Municipal Corporation, Pimpri, Pune 411018, Maharashtra, India
| | - Anuradha S. Tripathy
- Department of Dengue and Chikungunya, Indian Council of Medical Research-National Institute of Virology, 20-A, Dr Ambedkar Road, Pune 411001, India
| |
Collapse
|
16
|
Herrero Y, Velazquez C, Pascuali N, Hauk V, de Zúñiga I, Martínez G, Lavolpe M, Neuspiller F, Veiga MF, Scotti L, Abramovich D, Parborell F. Ovarian function after COVID-19: long-term effects and vaccine safety in ART patients. J Assist Reprod Genet 2025; 42:563-576. [PMID: 39883303 PMCID: PMC11871259 DOI: 10.1007/s10815-025-03403-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: 11/01/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
PURPOSE This study aimed to evaluate the long-term impact of mild COVID-19 infection and COVID-19 vaccination on ovarian function in patients undergoing assisted reproductive technology (ART). Specifically, we assessed ovarian outcomes between 9 and 18 months post-infection and investigated the effects of COVID-19 vaccines (inactivated virus and adenovirus) on reproductive parameters. METHODS The study included two objectives: (a) examining ovarian function in post-COVID-19 patients (9-18 months post-infection) compared to a control group and (b) comparing reproductive outcomes in vaccinated versus unvaccinated patients. According to the study objectives, ART patients were divided into the following groups: a control group (n = 30), a post-COVID-19 group (n = 55), an unvaccinated group (n = 70), and a vaccinated group (n = 55). Findings revealed a reduction in the number of retrieved and mature oocytes in patients over 36 years in the post-COVID-19 group. Lower IL-1β levels were found in follicular fluid (FF) of post-COVID-19 patients, while VEGF levels were reestablished between 9 and 18 months post-infection. Although cell migration was reduced in endothelial cells incubated with post-COVID-19 FF, angiogenic factor levels and DNA integrity remained stable. No significant differences in retrieved or mature oocytes were observed between vaccinated and unvaccinated patients. CONCLUSIONS VEGF levels and DNA integrity in FF from post-COVID-19 patients were normalized between 9 and 18 months post-infection. Additionally, COVID-19 vaccination did not negatively impact ovarian response in ART patients, supporting vaccine safety in reproductive contexts.
Collapse
Affiliation(s)
- Yamila Herrero
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Candela Velazquez
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Natalia Pascuali
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
- Department of Pathology, College of Medicine, University of Illinois at Chicago (UIC), Chicago, IL, USA
| | - Vanesa Hauk
- Immunopharmacology Laboratory, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | - Leopoldina Scotti
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
- Centro de Investigaciones y Transferencia del Noroeste de La Provincia de Buenos Aires (CITNOBA-UNSADA-CONICET), San Antonio de Areco, Argentina
| | - Dalhia Abramovich
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina.
| |
Collapse
|
17
|
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
|
18
|
Malaspina P, Jodice C, Ciminelli BM, Biancolella M, Colona VL, Latini A, Leonardis F, Rogliani P, Novelli A, Novelli G, Novelletto A. Genetic diversity of the immunoglobulin heavy chain locus in cohorts of patients affected with SARS-CoV-2. Hum Genomics 2025; 19:7. [PMID: 39885568 PMCID: PMC11780896 DOI: 10.1186/s40246-025-00719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/17/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND The Immunoglobulin Heavy Chain (IGH) genomic region is responsible for the production of circulating antibodies and warrants careful investigation for its association with COVID-19 characteristics. Multiple allelic variants within and across different IGH gene segments form a limited set of haplotypes. Previous studies have shown associations between some of these haplotypes and clinical outcomes of COVID-19. We typed 445 individuals of European ancestry, stratified for gender, age, and clinical status for 4 SNPs, two of which result in amino acid substitutions in IGHA2 and IGHG4, respectively. We analyzed associations at the single-locus level and for 4-loci haplotypes, inferred by phasing, after stratifying the overall cohort by gender, age, and disease severity. RESULTS Only weak evidence of significant differences between subgroups was obtained at the level of a single SNP. However, when the haplotypic data were analyzed for the young and old subgroups separately, uneven partitioning was observed regarding the occurrence of severe cases and Resistors. We then examined the cross-tabulation of disease severity in males and females, based on the presence of each haplotype in the genotype. Two haplotypes were underrepresented in young severe cases compared to old severe ones. The same two haplotypes were overrepresented among young Resistors. These findings provide stronger support for, the weak associations observed at the single locus level. CONCLUSIONS Two haplotypes seem to act as protective factors specifically in young individuals, counteracting the general increase in vulnerability with age. This observation aligns with stronger genetic effects seen in young patients for other susceptibility genes. Our findings complement previous research identifying specific genetic variants that influence COVID-19 susceptibility and severity, emphasizing the complex interplay between host genetics and viral infection outcomes. Our results are consistent with a potential causative role of IGH regulatory regions (e.g. HS1.2), which are flanked by the SNP set here analyzed.
Collapse
Affiliation(s)
- Patrizia Malaspina
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy.
| | - Carla Jodice
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Bianca Maria Ciminelli
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Michela Biancolella
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| | - Vito Luigi Colona
- Research Unit of Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Latini
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
| | | | | | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, Rome, Italy
- Tor Vergata University Hospital, Rome, Italy
| | - Andrea Novelletto
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133, Rome, Italy
| |
Collapse
|
19
|
Li L, Zhang X, Yan H, Dai M, Gao H, Wang Y, Jiang P, Dai E. Different immunological characteristics of asymptomatic and symptomatic COVID-19 patients without vaccination in the acute and convalescence stages. PeerJ 2025; 13:e18451. [PMID: 39897496 PMCID: PMC11786710 DOI: 10.7717/peerj.18451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/14/2024] [Indexed: 02/04/2025] Open
Abstract
The immune status of Coronavirus disease 2019 (COVID-19) patients in different stages of infection remains difficult to determine. In this study, we performed high-throughput single-cell mass cytometry on peripheral blood samples from 10 COVID-19 patients and four healthy donors to analyze their immune status at acute and convalescence phases. During the acute stage, the proportion of neutrophils increased significantly while natural killer (NK) cells decreased. In contrast, during the convalescence phase, the proportion of plasma cells decreased from the acute stage of disease onset and was lower than normal. The proportions of B, mast and plasma cell subsets decreased significantly with the process of disease recovery. Further analysis of the subsets of major immune cell types in COVID-19 patients with different clinical presentations in different stages showed that in the acute stages of disease progression, the T helper cell 1 (Th1), IgD+ B and neutrophil subsets increased in COVID-19 patients, especially in symptomatic patients, while the central memory CD4+T cells (CD4 TCM), mucosa-associated invariant T (MAIT) and NK cell subsets decreased significantly, especially in symptomatic patients. Then CD4 TCM and MAIT returned to normal levels at the recovery phase. Dynamic assessment displayed that the immune imbalance at the onset of COVID-19 could be corrected during recovery. Our study provides additional information on the immune status of COVID-19 patients with different clinical manifestations in different stages. These findings may provide new insights into COVID-19 immunotherapy and immune intervention.
Collapse
Affiliation(s)
- Li Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Intensive Care Unit, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Xin Zhang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Tuberculosis, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Huimin Yan
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Clinical Research Center, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Muwei Dai
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University and Hebei Cancer Hospital, Shijiazhuang, Hebei, China
| | - Huixia Gao
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Yuling Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Ping Jiang
- Department of Cardiovascular Medicine, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Erhei Dai
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
- Department of Laboratory Medicine, The Fifth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| |
Collapse
|
20
|
Li J, Mao N, Wang Y, Deng S, Chen K. Novel insights into the ROCK-JAK-STAT signaling pathway in upper respiratory tract infections and neurodegenerative diseases. Mol Ther 2025; 33:32-50. [PMID: 39511889 PMCID: PMC11764622 DOI: 10.1016/j.ymthe.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Acute upper respiratory tract infections are a major public health issue, with uncontrolled inflammation triggered by upper respiratory viruses being a significant cause of patient deterioration or death. This study focuses on the Janus kinase-signal transducer and activator of transcription Rho-associated coiled-coil containing protein kinase (JAK-STAT-ROCK) signaling pathway, providing an in-depth analysis of the interplay between uncontrolled inflammation after upper respiratory tract infections and the development of neurodegenerative diseases. It offers a conceptual framework for understanding the lung-brain-related immune responses and potential interactions. The relationship between the ROCK-JAK-STAT signaling pathway and inflammatory immunity is a complex and multi-layered research area and exploring potential common targets could open new avenues for the prevention and treatment of related inflammation.
Collapse
Affiliation(s)
- Jiaxuan Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China
| | - Naihui Mao
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, P.R. China.
| |
Collapse
|
21
|
Zheng J, Dhakal H, Qing E, Shrestha R, Geller AE, Morrissey SM, Saxena D, Hu X, Li H, Li H, Wilhelmsen K, Wendt LH, Klumpp K, Hume PS, Janssen WJ, Brody R, Palmer KE, Uriarte SM, Ten Eyck P, Meyerholz DK, Merchant ML, McLeish K, Gallagher T, Huang J, Yan J, Perlman S. CXCL12 ameliorates neutrophilia and disease severity in SARS-CoV-2 infection. J Clin Invest 2025; 135:e188222. [PMID: 39773555 PMCID: PMC11827850 DOI: 10.1172/jci188222] [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: 10/18/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025] Open
Abstract
Neutrophils, particularly low-density neutrophils (LDNs), are believed to contribute to acute COVID-19 severity. Here, we showed that neutrophilia can be detected acutely and even months after SARS-CoV-2 infection in patients and mice, while neutrophil depletion reduced disease severity in mice. A key factor in neutrophilia and severe disease in infected mice was traced to the chemokine CXCL12 secreted by bone marrow cells and unexpectedly, endothelial cells. CXCL12 levels were negatively correlated with LDN numbers in longitudinal analyses of patient blood samples. CXCL12 blockade in SARS-CoV-2-infected mice increased blood/lung neutrophil numbers, thereby accelerating disease progression without changing lung virus titers. The exaggerated mortality caused by CXCL12 blockade could be reversed by neutrophil depletion. In addition, blocking interactions between SARS-CoV-2 and angiotensin-converting enzyme 2 (ACE2) reduced CXCL12 levels, suggesting a signal transduction from virus-mediated ACE2 ligation to increased CXCL12 secretion. Collectively, these results demonstrate a previously unappreciated role of CXCL12 in diminishing neutrophilia, including low-density neutrophilia, and its deleterious effects in SARS-CoV-2 infections. The results also support the involvement of SARS-CoV-2-endothelial cell interactions in viral pathogenesis.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Microbiology and Immunology and
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hima Dhakal
- Department of Microbiology and Immunology and
| | - Enya Qing
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Rejeena Shrestha
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Anne E. Geller
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Samantha M. Morrissey
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Divyasha Saxena
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Xiaoling Hu
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Haiyan Li
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | | | - Linder H. Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | - Patrick S. Hume
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenneth E. Palmer
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology and
| | - Silvia M. Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Patrick Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Kenneth McLeish
- Department of Medicine, Division of Nephrology and Hypertension and
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
22
|
Snyder TM, Gittelman RM, Klinger M, May DH, Osborne EJ, Taniguchi R, Jabran Zahid H, Kaplan IM, Dines JN, Noakes MT, Pandya R, Chen X, Elasady S, Svejnoha E, Ebert P, Pesesky MW, De Almeida P, O’Donnell H, DeGottardi Q, Keitany G, Lu J, Vong A, Elyanow R, Fields P, Al-Asadi H, Greissl J, Baldo L, Semprini S, Cerchione C, Nicolini F, Mazza M, Delmonte OM, Dobbs K, Laguna-Goya R, Carreño-Tarragona G, Barrio S, Imberti L, Sottini A, Quiros-Roldan E, Rossi C, Biondi A, Bettini LR, D’Angio M, Bonfanti P, Tompkins MF, Alba C, Dalgard C, Sambri V, Martinelli G, Goldman JD, Heath JR, Su HC, Notarangelo LD, Paz-Artal E, Martinez-Lopez J, Howie B, Carlson JM, Robins HS. Magnitude and dynamics of the T-cell response to SARS-CoV-2 infection at both individual and population levels. Front Immunol 2025; 15:1488860. [PMID: 39840037 PMCID: PMC11747429 DOI: 10.3389/fimmu.2024.1488860] [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: 08/30/2024] [Accepted: 12/05/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction T cells are involved in the early identification and clearance of viral infections and also support the development of antibodies by B cells. This central role for T cells makes them a desirable target for assessing the immune response to SARS-CoV-2 infection. Methods Here, we combined two high-throughput immune profiling methods to create a quantitative picture of the T-cell response to SARS-CoV-2. First, at the individual level, we deeply characterized 3 acutely infected and 58 recovered COVID-19 subjects by experimentally mapping their CD8 T-cell response through antigen stimulation to 545 Human Leukocyte Antigen (HLA) class I presented viral peptides. Then, at the population level, we performed T-cell repertoire sequencing on 1,815 samples (from 1,521 COVID-19 subjects) as well as 3,500 controls to identify shared "public" T-cell receptors (TCRs) associated with SARS-CoV-2 infection from both CD8 and CD4 T cells. Results Collectively, our data reveal that CD8 T-cell responses are often driven by a few immunodominant, HLA-restricted epitopes. As expected, the T-cell response to SARS-CoV-2 peaks about one to two weeks after infection and is detectable for at least several months after recovery. As an application of these data, we trained a classifier to diagnose SARS-CoV-2 infection based solely on TCR sequencing from blood samples, and observed, at 99.8% specificity, high early sensitivity soon after diagnosis (Day 3-7 = 85.1% [95% CI = 79.9-89.7]; Day 8-14 = 94.8% [90.7-98.4]) as well as lasting sensitivity after recovery (Day 29+/convalescent = 95.4% [92.1-98.3]). Discussion The approaches described in this work provide detailed insights into the adaptive immune response to SARS-CoV-2 infection, and they have potential applications in clinical diagnostics, vaccine development, and monitoring.
Collapse
Affiliation(s)
| | | | - Mark Klinger
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Damon H. May
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | | | - Ian M. Kaplan
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Ravi Pandya
- Microsoft Research, Redmond, WA, United States
| | - Xiaoyu Chen
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Peter Ebert
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | | | | | | | - Jennifer Lu
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Allen Vong
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | - Paul Fields
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | | - Lance Baldo
- Adaptive Biotechnologies, Seattle, WA, United States
| | - Simona Semprini
- Unit of Microbiology - The Great Romagna Hub Laboratory, Pievesestina ITALY and DIMES, University of Bologna, Bologna, Italy
| | - Claudio Cerchione
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Immunotherapy, Cell Therapy and Biobank (ITCB), IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Ottavia M. Delmonte
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kerry Dobbs
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rocio Laguna-Goya
- Department of Immunology, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | | | - Santiago Barrio
- Hematology Department, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Luisa Imberti
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Alessandra Sottini
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Eugenia Quiros-Roldan
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Camillo Rossi
- Laboratorio CREA, Department of Infectious and Tropical Diseases, and Medical Officer, ASST Spedali Civili di Brescia and University of Brescia, Brescia, Italy
| | - Andrea Biondi
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Laura Rachele Bettini
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Mariella D’Angio
- Department of Pediatrics and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale San Gerardo, Monza, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, University of Milano-Bicocca-Ospedale San Gerardo, Monza, Italy
| | - Miranda F. Tompkins
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Camille Alba
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Vittorio Sambri
- Unit of Microbiology - The Great Romagna Hub Laboratory, Pievesestina ITALY and DIMES, University of Bologna, Bologna, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Jason D. Goldman
- Swedish Medical Center, Seattle, WA, United States
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - James R. Heath
- Institute for Systems Biology, Seattle, WA, United States
| | - Helen C. Su
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Luigi D. Notarangelo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Estela Paz-Artal
- Department of Immunology, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Joaquin Martinez-Lopez
- Hematology Department, Hospital 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Bryan Howie
- Adaptive Biotechnologies, Seattle, WA, United States
| | | | | |
Collapse
|
23
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [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: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
24
|
Castillo-Galán S, Parra V, Cuenca J. Unraveling the pathogenesis of viral-induced pulmonary arterial hypertension: Possible new therapeutic avenues with mesenchymal stromal cells and their derivatives. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167519. [PMID: 39332781 DOI: 10.1016/j.bbadis.2024.167519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Pulmonary hypertension (PH) is a severe condition characterized by elevated pressure in the pulmonary artery, where metabolic and mitochondrial dysfunction may contribute to its progression. Within the PH spectrum, pulmonary arterial hypertension (PAH) stands out with its primary pulmonary vasculopathy. PAH's prevalence varies from 0.4 to 1.4 per 100,000 individuals and is associated with diverse conditions, including viral infections such as HIV. Notably, recent observations highlight an increased occurrence of PAH among COVID-19 patients, even in the absence of pre-existing cardiopulmonary disorders. While current treatments offer partial relief, there's a pressing need for innovative therapeutic strategies, among which mesenchymal stromal cells (MSCs) and their derivatives hold promise. This review critically evaluates recent investigations into viral-induced PAH, encompassing pathogens like human immunodeficiency virus, herpesvirus, Cytomegalovirus, Hepatitis B and C viruses, SARS-CoV-2, and Human endogenous retrovirus K (HERKV), with a specific emphasis on mitochondrial dysfunction. Furthermore, we explore the underlying rationale driving novel therapeutic modalities, including MSCs, extracellular vesicles, and mitochondrial interventions, within the framework of PAH management.
Collapse
Affiliation(s)
- Sebastián Castillo-Galán
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| | - Valentina Parra
- Laboratory of Differentiation and Cell Metabolism (D&M), Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; SYSTEMIX Center for Systems Biology, O'Higgins University, Rancagua, Chile
| | - Jimena Cuenca
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CIIB), Faculty of Medicine, Universidad de los Andes, Chile; IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile; Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile; Cells for Cells, Santiago, Chile.
| |
Collapse
|
25
|
Krasnenkova SF, Zayratyants OV, Midiber KY, Mikhaleva LM. [Liver pathology in COVID-19]. Arkh Patol 2025; 87:53-59. [PMID: 39943730 DOI: 10.17116/patol20258701153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
The literature review presents an analysis of the pathogenesis and pathological anatomy of liver damage in COVID-19. Liver damage with the steatosis, vascular disorders, mild portal and lobular inflammatory infiltration, cholestasis and clinically - liver failure is observed in majority of the patients with COVID-19. Chronic liver diseases with infection SARS-CoV-2 tend to decompensate, which significantly worsens the prognosis of the disease. Pathogenesis of liver damage in COVID19 is unclear. There was no convincing evidence for the hypothesis of cytotoxicity for hepatocytes or cholangiocytes by SARS-CoV-2. Similar liver morphological changes described by different authors suggest their nonspecific nature and multifactorial pathogenesis related to hypoxia, cytokin storm, systemic inflammatory response syndrome, sepsis and shock, Covid-associated angio- and coagulopathy, as well as drug-induced hepatotoxicity. Further research is needed to clarify the pathogenesis and pathological anatomy of the liver pathology in COVID-19.
Collapse
Affiliation(s)
- S F Krasnenkova
- Russian University of Medicine, Moscow, Russia
- Research Institute of Organization of Medicine and Medicine Management, Moscow, Russia
| | - O V Zayratyants
- Russian University of Medicine, Moscow, Russia
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
| | - K Yu Midiber
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
- Peoples' Friendship University of Russia named after Patrice Lumumba, Moscow, Russia
| | - L M Mikhaleva
- Avtsyn Research Institute of Human Morphology of Petrovsky National Research Centre of Surgery, Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| |
Collapse
|
26
|
Montgomery S, Vingeliene S, Li H, Backman H, Udumyan R, Jendeberg J, Rasmussen G, Sundqvist M, Fall K, Hiyoshi A, Nyberg F. SARS-CoV-2 infection and risk of subsequent demyelinating diseases: national register-based cohort study. Brain Commun 2024; 6:fcae406. [PMID: 39659973 PMCID: PMC11629974 DOI: 10.1093/braincomms/fcae406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/31/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Demyelinating diseases including multiple sclerosis are associated with prior infectious exposures, so we assessed whether SARS-CoV-2 infection is associated with subsequent diagnoses of non-multiple sclerosis demyelinating diseases and multiple sclerosis. All residents of Sweden aged 3-100 years were followed between 1 January 2020 and 30 November 2022, excluding those with demyelinating disease prior to 2020, comprising 9 959 818 individuals divided into uninfected and those who were infected were categorized into those with and without hospital admission for the infection as a marker of infection severity. Cox regression assessed the risk of two separate outcomes: hospital diagnosed non-multiple sclerosis demyelinating diseases of the CNS and multiple sclerosis. The exposures were modelled as time-varying covariates (uninfected, infection without hospital admission and infected with hospital admission). Hospital admission for COVID-19 was associated with raised risk of subsequent non-multiple sclerosis demyelinating disease, but only 12 individuals had this outcome among the exposed, and of those, 7 has an unspecified demyelinating disease diagnosis. Rates per 100 000 person-years (and 95% confidence intervals) were 3.8 (3.6-4.1) among those without a COVID-19 diagnosis and 9.0 (5.1-15.9) among those admitted to hospital for COVID-19, with an adjusted hazard ratio and (and 95% confidence interval) of 2.35 (1.32-4.18, P = 0.004). Equivalent associations with multiple sclerosis (28 individuals had this outcome among the exposed) were rates of 9.5 (9.1-9.9) and 21.0 (14.5-30.5) and an adjusted hazard ratio of 2.48 (1.70-3.61, P < 0.001). Only a small number of non-multiple sclerosis demyelinating disease diagnoses were associated with hospital admission for COVID-19, and while the number with multiple sclerosis was somewhat higher, longer duration of follow-up will assist in identifying whether the associations are causal or due to shared susceptibility or surveillance bias, as these diseases can have long asymptomatic and prodromal phases.
Collapse
Affiliation(s)
- Scott Montgomery
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 703 62 Örebro, Sweden
- Division of Clinical Epidemiology, Department of Medicine, Solna, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Epidemiology and Public Health, University College London, London WC1E 7HB, UK
| | - Snieguole Vingeliene
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 703 62 Örebro, Sweden
| | - Huiqi Li
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Helena Backman
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden
| | - Ruzan Udumyan
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 703 62 Örebro, Sweden
| | - Johan Jendeberg
- Department of Radiology, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden
| | - Gunlög Rasmussen
- Department of Infectious Diseases, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden
| | - Martin Sundqvist
- Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, 701 85 Örebro, Sweden
| | - Katja Fall
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 703 62 Örebro, Sweden
- The Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ayako Hiyoshi
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 703 62 Örebro, Sweden
| | - Fredrik Nyberg
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
27
|
Qiu M, Song X, Zhang Q, Zou S, Pang L, Nian X. Immunophenotyping characteristics and clinical outcome of COVID-19 patients treated with azvudine during the Omicron surge. Front Immunol 2024; 15:1465238. [PMID: 39654887 PMCID: PMC11625784 DOI: 10.3389/fimmu.2024.1465238] [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: 07/16/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Background Little is known about immunophenotyping characteristics and clinical outcomes of COVID-19 patients treated with azvudine during the Omicron variant surge. Methods This study enrolled patients diagnosed with COVID-19 from December 2022 to February 2023. The primary outcome was defined as all-cause mortality, along with a composite outcome reflecting disease progression. The enrolled patients were followed for a period of 60 days from their admission. Results A total of 268 COVID-19 patients treated with azvudine were enrolled in this retrospective study. The study found that the counts of lymphocyte subsets were significantly reduced in the composite outcome and all-cause mortality groups compared to the non-composite outcome and discharge groups (all p < 0.001). Correlation analysis revealed a negative association between lymphocyte subsets cell counts and inflammatory markers levels. The receiver operating characteristic (ROC) curve analysis identified low CD4+ T cell count as the most significant predictor of disease progression and all-cause mortality among the various lymphocyte subsets. Additionally, both the Kaplan-Meier curve and multivariate regression analysis demonstrated that low CD4+ T cell count level (< 156.00 cells/μl) was closely associated with all-cause mortality in COVID-19 patients treated with azvudine. Conclusions A low CD4+ T cell count may serve as a significant predictive indicator for identifying COVID-19 patients receiving azvudine treatment who are at an elevated risk of experiencing adverse outcomes. These findings may offer valuable insights for physicians in optimizing the administration of azvudine.
Collapse
Affiliation(s)
- Meihua Qiu
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaogang Song
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Qianqian Zhang
- Department of Imaging, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Shenchun Zou
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Lingling Pang
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Xueyuan Nian
- Department of Gastroenterology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
28
|
Abu-Helu RF, Zeer MJ, Adwan GM. The Association Between Diabetes and the Outcome of COVID-19 Infection in Bethlehem, Palestine: A Case-Control Study. J Pathog 2024; 2024:1051658. [PMID: 39995847 PMCID: PMC11850064 DOI: 10.1155/2024/1051658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/04/2024] [Indexed: 02/26/2025] Open
Abstract
The severity of COVID-19's outcomes has been positively correlated with an increased risk of respiratory failure and death, especially in patients with chronic illnesses. This case-control design study aims to examine the correlation in the Palestinian population in light of its impact on diabetic patients. The study was conducted from March 2020 to June 2021 on 417 patients admitted to the Palestinian National Center for Rehabilitation. Of them, 198 cases were tested positive for COVID-19 and had diabetes, whereas the remaining 219 were those who tested positive for COVID-19 but were not diagnosed with diabetes and acted as controls. Data from patient files were collected to address the study questions. Patients' ages ranged from 17 to 98 years, with a mean age of 58. Male participants represented 53.5% of the total. The results of the current study indicated that the case fatality rate (CFR) was 14.2% for all participants and 19.7% for patients with diabetes. In regard to patients' health conditions, 5.7% had cardiovascular diseases (CVD), 33.6% had hypertension, 6.5% had kidney diseases, and 47.4% had diabetes. According to the multivariate analysis, diabetic patients had a 1.63 times higher risk for COVID-19 infection compared to nondiabetic patients (adjustment of odds ratio of 1.63 and confidence interval [CI] = 0.354-1.856). This is the first study to investigate the relationship between the severity of COVID-19 outcomes in Palestine and the diabetes status, and the majority of its findings are consistent with other research studies that assessed the impact of COVID-19 on diabetic patients (with minor variations in the number and percentage). The study confirms that along with diabetes, age, hypertension, kidney diseases, shortness of breath, requirement of oxygen, D-dimer, C-reactive protein (CRP), kidney function test (KFT), and days of hospitalization were also associated with severe COVID-19 outcomes, whereas gender, CVDs, and liver function test (LFT) were not associated with severe COVID-19 outcomes.
Collapse
Affiliation(s)
- Rasmi Fayiz Abu-Helu
- Department of Medical Laboratory Sciences, Faculty of Health Professions, Al-Quds University, Jerusalem, State of Palestine
| | | | - Ghaleb Mohammad Adwan
- Department of Biology and Biotechnology, An-Najah National University, P.O. Box 7, Nablus, State of Palestine
| |
Collapse
|
29
|
Hu Z, Wang W, Lin Y, Guo H, Chen Y, Wang J, Yu F, Rao L, Fan Z. Extracellular Vesicle-Inspired Therapeutic Strategies for the COVID-19. Adv Healthc Mater 2024; 13:e2402103. [PMID: 38923772 DOI: 10.1002/adhm.202402103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Emerging infectious diseases like coronavirus pneumonia (COVID-19) present significant challenges to global health, extensively affecting both human society and the economy. Extracellular vesicles (EVs) have demonstrated remarkable potential as crucial biomedical tools for COVID-19 diagnosis and treatment. However, due to limitations in the performance and titer of natural vesicles, their clinical use remains limited. Nonetheless, EV-inspired strategies are gaining increasing attention. Notably, biomimetic vesicles, inspired by EVs, possess specific receptors that can act as "Trojan horses," preventing the virus from infecting host cells. Genetic engineering can enhance these vesicles by enabling them to carry more receptors, significantly increasing their specificity for absorbing the novel coronavirus. Additionally, biomimetic vesicles inherit numerous cytokine receptors from parent cells, allowing them to effectively mitigate the "cytokine storm" by adsorbing pro-inflammatory cytokines. Overall, this EV-inspired strategy offers new avenues for the treatment of emerging infectious diseases. Herein, this review systematically summarizes the current applications of EV-inspired strategies in the diagnosis and treatment of COVID-19. The current status and challenges associated with the clinical implementation of EV-inspired strategies are also discussed. The goal of this review is to provide new insights into the design of EV-inspired strategies and expand their application in combating emerging infectious diseases.
Collapse
Affiliation(s)
- Ziwei Hu
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Wei Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Ying Lin
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Hui Guo
- Department of Dermatology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, P. R. China
| | - Yiwen Chen
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Junjie Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Feng Yu
- Institute of Otolaryngology Head and neck surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, 510282, P. R. China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, P. R. China
| | - Zhijin Fan
- Institute for Engineering Medicine, Kunming Medical University, Kunming, 650500, P. R. China
| |
Collapse
|
30
|
Almskog LM, Sjöström A, Sundén-Cullberg J, Taxiarchis A, Ågren A, Freyland S, Börjesson M, Wikman A, Wahlgren CM, Wanecek M, van der Linden J, Antovic J, Lampa J, Magnusson M. Tocilizumab reduces hypercoagulation in COVID-19 - Perspectives from the coagulation and immunomodulation Covid assessment (Coag-ImmCovA) clinical trial. Thromb Res 2024; 243:109135. [PMID: 39226747 DOI: 10.1016/j.thromres.2024.109135] [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: 06/17/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Despite medical interventions, COVID-19 continues to persist at pandemic proportions. A hypercoagulation state was rapidly observed in the severely ill, and the incidence of thromboembolic events remains elevated. Interleukin inhibitors have demonstrated positive effects on the hyperactivation of the immune system in COVID-19, with the interleukin-6 inhibitor tocilizumab showing promising results in reducing mortality. Nevertheless, the impact of interleukin inhibitors on the coagulation system remains incompletely understood. METHODS In this clinical trial conducted in Stockholm, Sweden, interleukin inhibitors, namely anakinra (ANA) or tocilizumab (TOCI), were randomly administered in addition to standard care (SC) to hospitalized patients with COVID-19. A control group received only SC. The primary outcome sought to measure effects on global hemostasis, as indicated by changes in functional coagulation tests, specifically Rotational Thromboelastometry (ROTEM) or Overall Hemostatic Potential (OHP), visualized through scanning electron microscopy images. Secondary outcomes included effects on conventional coagulation laboratory tests. RESULTS The study enrolled 74 patients who were randomized to receive either ANA or TOCI in addition to SC, or SC alone. In the TOCI group, ROTEM variables exhibited less hypercoagulation after 29 days compared with ANA or SC treatment groups, characterized by prolonged clot formation time and decreased clot firmness. OHP decreased, but there were no significant differences among the three treatment groups. Plasma fibrinogen levels, initially elevated, decreased significantly in TOCI recipients over time. CONCLUSION Tocilizumab treatment demonstrated a significant reduction of hypercoagulation in hospitalized COVID-19 patients, by improvements in both global coagulation tests and conventional laboratory tests, in comparison with anakinra or SC alone. This finding underscores the significance of tocilizumab as a viable treatment option in severe COVID-19 cases, with the potential to decrease thrombosis incidence.
Collapse
Affiliation(s)
- Lou M Almskog
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.
| | - Anna Sjöström
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Sundén-Cullberg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Apostolos Taxiarchis
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Ågren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Freyland
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Madeleine Börjesson
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Agneta Wikman
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Center for Hematology and Regenerative Medicine (HERM), Karolinska Institutet, Stockholm, Sweden
| | - Carl Magnus Wahlgren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Michael Wanecek
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Intensive Care Unit, Capio St Göran's Hospital, Stockholm, Sweden
| | - Jan van der Linden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jovan Antovic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
| | - Jon Lampa
- Rheumatology Division, Department of Medicine Solna, Center for Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Magnusson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Cong B, Dong X, Yang Z, Yu P, Chai Y, Liu J, Zhang M, Zang Y, Kang J, Feng Y, Liu Y, Feng W, Wang D, Deng W, Li F, Song Z, Wang Z, Chen X, Qin H, Yu Q, Li Z, Liu S, Xu X, Zhong N, Ren X, Qin C, Liu L, Wang J, Cao X. Single-cell spatiotemporal analysis of the lungs reveals Slamf9 + macrophages involved in viral clearance and inflammation resolution. Cell Discov 2024; 10:104. [PMID: 39414783 PMCID: PMC11484945 DOI: 10.1038/s41421-024-00734-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/08/2024] [Indexed: 10/18/2024] Open
Abstract
How the lung achieves immune homeostasis after a pulmonary infection is not fully understood. Here, we analyzed the spatiotemporal changes in the lungs over a 2-week natural recovery from severe pneumonia in a Syrian hamster model of SARS-CoV-2 infection. We find that SARS-CoV-2 infects multiple cell types and causes massive cell death at the early stage, including alveolar macrophages. We identify a group of monocyte-derived Slamf9+ macrophages, which are induced after SARS-CoV-2 infection and resistant to impairment caused by SARS-CoV-2. Slamf9+ macrophages contain SARS-CoV-2, recruit and interact with Isg12+Cst7+ neutrophils to clear the viruses. After viral clearance, Slamf9+ macrophages differentiate into Trem2+ and Fbp1+ macrophages, contributing to inflammation resolution at the late stage, and finally replenish alveolar macrophages. These findings are validated in a SARS-CoV-2-infected hACE2 mouse model and confirmed with publicly available human autopsy single-cell RNA-seq data, demonstrating the potential role of Slamf9+ macrophages and their coordination with neutrophils in post-injury tissue repair and inflammation resolution.
Collapse
Affiliation(s)
- Boyi Cong
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Dong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zongheng Yang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pin Yu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyang Chai
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaqi Liu
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Meihan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | | | | | - Yu Feng
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Dehe Wang
- Changping Laboratory, Beijing, China
| | - Wei Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fengdi Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqi Song
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqiao Wang
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosu Chen
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Qin
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China
| | - Qinyi Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhiqing Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Navy Medical University, Shanghai, China
- Guangzhou Laboratory, Guangzhou, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | | | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Xuetao Cao
- State Key Laboratory of Medicinal Chemical Biology, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, China.
- Department of Immunology, Center for Immunotherapy, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
32
|
Bispo ECI, Argañaraz ER, Neves FDAR, de Carvalho JL, Saldanha-Araujo F. Immunomodulatory effect of IFN-γ licensed adipose-mesenchymal stromal cells in an in vitro model of inflammation generated by SARS-CoV-2 antigens. Sci Rep 2024; 14:24235. [PMID: 39415027 PMCID: PMC11484699 DOI: 10.1038/s41598-024-75776-5] [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: 07/25/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
In recent years, clinical studies have shown positive results of the application of Mesenchymal Stromal Cells (MSCs) in severe cases of COVID-19. However, the mechanisms of immunomodulation of IFN-γ licensed MSCs in SARS-CoV-2 infection are only partially understood. In this study, we first tested the effect of IFN-γ licensing in the MSC immunomodulatory profile. Then, we established an in vitro model of inflammation by exposing Calu-3 lung cells to SARS-CoV-2 nucleocapsid and spike (NS) antigens, and determined the toxicity of SARS-CoV-2 NS antigen and/or IFN-γ stimulation to Calu-3. The conditioned medium (iCM) generated by Calu-3 cells exposed to IFN-γ and SARS-CoV-2 NS antigens was used to stimulate T-cells, which were then co-cultured with IFN-γ-licensed MSCs. The exposure to IFN-γ and SARS-CoV-2 NS antigens compromised the viability of Calu-3 cells and induced the expression of the inflammatory mediators ICAM-1, CXCL-10, and IFN-β by these cells. Importantly, despite initially stimulating T-cell activation, IFN-γ-licensed MSCs dramatically reduced IL-6 and IL-10 levels secreted by T-cells exposed to NS antigens and iCM. Moreover, IFN-γ-licensed MSCs were able to significantly inhibit T-cell apoptosis induced by SARS-CoV-2 NS antigens. Taken together, our data show that, in addition to reducing the level of critical cytokines in COVID-19, IFN-γ-licensed MSCs protect T-cells from SARS-CoV-2 antigen-induced apoptosis. Such observations suggest that MSCs may contribute to COVID-19 management by preventing the lymphopenia and immunodeficiency observed in critical cases of the disease.
Collapse
Affiliation(s)
- Elizabete Cristina Iseke Bispo
- Laboratory of Hematology and Stem Cells (LHCT), Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular NeuroVirology, Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil
| | | | - Juliana Lott de Carvalho
- Interdisciplinary Laboratory of Bioscience, Faculty of Medicine, University of Brasília, Brasília, 70910-900, Brazil
| | - Felipe Saldanha-Araujo
- Laboratory of Hematology and Stem Cells (LHCT), Faculty of Health Sciences, University of Brasília, Brasília, 70910-900, Brazil.
| |
Collapse
|
33
|
Wu K, Yin L, Han J, Cai Q, Guo Y, Jin X, Wu J, Cheng Y. Case-control study on risk factors for in-hospital mortality in patients with severe COVID-19. Front Public Health 2024; 12:1424720. [PMID: 39440172 PMCID: PMC11493594 DOI: 10.3389/fpubh.2024.1424720] [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: 04/28/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Objective The purpose of this study was to identify independent risk factors affecting patient survival and explore predictors of severe cases of coronavirus disease 2019 (COVID-19). Methods We conducted a retrospective, observational, case-control study on adult patients with severe COVID-19 who were admitted to affiliated hospitals in Tianjin between December 18, 2022, and January 31, 2023. We used univariate and multifactorial logistic regression analyses to analyze demographic indicators, comorbidity profiles, and laboratory parameters in two groups of patients (deceased and surviving) to identify independent risk factors for death in patients with severe COVID-19. Results Patients in the deceased group were older than those in the survival group (p = 0.018), and there were more cases of coexisting respiratory insufficiency in the deceased group (p = 0.002). Additionally, laboratory test results for white blood cell count (WBC) and creatine kinase (CK) showed significantly higher values in the deceased group (p = 0.047 and p = 0.029, respectively), while arterial oxygen partial pressure (PAO2) showed significantly lower values compared to the survival group (p = 0.021). Age, respiratory insufficiency, WBCH (highest WBC value), CKH (highest CK value), and PAO2F (first PAO2 value) had area under curve (AUC) values of 0.698, 0.838, 0.721, 0.744, and 0.633, respectively. Conclusion The main risk factors for mortality in patients with severe COVID-19 that we identified in this study were the advanced age of patients, coexisting respiratory insufficiency, elevated levels of WBC and CK, and decreased levels of PAO2. Elevated WBC and CK laboratory parameters, in particular, demonstrated good predictive value for in-hospital mortality risk.
Collapse
Affiliation(s)
- Kemei Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lili Yin
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiangqin Han
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuhan Cai
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Guo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xin Jin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinling Wu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yupei Cheng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
34
|
Pinto S, Al Lawati H, Al Raisi M, Maawali BA. Association Between the Use of Proton Pump Inhibitors and Severe Clinical Outcomes in COVID-19 Patients: A Retrospective Observational Study. Cureus 2024; 16:e72385. [PMID: 39463908 PMCID: PMC11510648 DOI: 10.7759/cureus.72385] [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: 10/25/2024] [Indexed: 10/29/2024] Open
Abstract
Background Proton pump inhibitors (PPIs) increase the risk of pneumonia secondary to PPI-induced hypochlorhydria. We aim to investigate the association between PPI and disease severity in coronavirus disease 2019 (COVID-19)-positive patients and the risk of hospitalizations in Muscat, Oman. Methodology COVID-19-positive patients aged 18 years and above at the time of diagnosis were included in this retrospective observational study. The details of the patients were retrieved from the electronic health records of the Al Shifa Hospital Information Management System and Tarassud. The composite primary endpoint was COVID-19 admission to a government tertiary hospital ward or intensive care within 14 days of diagnosis. Results A total of 506 COVID-19-positive patients were identified during the specified period. The mean age was 44 ± 15 years. The majority of the patients were Omani, and a female preponderance was observed. Overall, 104 (20.4%) patients were current PPI users. Admission due to COVID-19 was significantly associated with the presence of comorbid conditions such as diabetes mellitus (p = 0.001), hypertension (p = 0.001), and chronic kidney disease (p < 0.001). However, current PPI use (p = 0.140) was not significantly associated with an increased risk of hospitalization. Conclusions This data suggests that the use of PPIs during COVID-19 infection did not increase the risk of severe COVID-19 infection and poor outcomes leading to hospitalization in Muscat, Oman. However, the presence of other medical comorbidities, such as diabetes and hypertension, was associated with a higher risk of adverse clinical symptoms that resulted in hospitalization.
Collapse
Affiliation(s)
- Sharon Pinto
- Primary Healthcare, Ministry of Health, Muscat, OMN
- Medicine, James Cook University Hospital, Middlesbrough, GBR
| | | | | | | |
Collapse
|
35
|
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
|
36
|
Krisht AAH, Grapin K, de Beauchene RC, Bonnet B, Cassagnes L, Evrard B, Adda M, Souweine B, Dupuis C. SARS-CoV2 pneumonia patients admitted to the ICU: Analysis according to clinical and biological parameters and the extent of lung parenchymal lesions on chest CT scan, a monocentric observational study. PLoS One 2024; 19:e0308014. [PMID: 39298399 PMCID: PMC11412649 DOI: 10.1371/journal.pone.0308014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/16/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND CT-scan and inflammatory and coagulation biomarkers could help in prognostication of COVID-19 in patients on ICU admission. OBJECTIVE The objectives of this study were to measure the prognostic value of the extent of lung parenchymal lesions on computed tomography (CT) and of several coagulation and inflammatory biomarkers, and to explore the characteristics of the patients depending on the extent of lung parenchymal lesions. DESIGN Retrospective monocentric observational study achieved on a dataset collected prospectively. SETTING Medical ICU of the university hospital of Clermont-Ferrand, France. PATIENTS All consecutive adult patients aged ≥18 years admitted between 20 March, 2020 and 31 August, 2021 for COVID-19 pneumonia. INTERVENTIONS Characteristics at baseline and during ICU stay, and outcomes at day 60 were recorded. The extent of lung parenchyma lesions observed on the chest CT performed on admission was established by artificial intelligence software. MEASUREMENTS Several clinical characteristics and laboratory features were collected on admission including plasma interleukin-6, HLA-DR monocytic-expression rate (mHLA-DR), and the extent of lung parenchymal lesions. Factors associated with day-60 mortality were investigated by uni- and multivariate survival analyses. RESULTS 270 patients were included. Inflammation biomarkers including the levels of neutrophils, CRP, ferritin and Il10 were the indices the most associated with the severity of the extent of the lung lesions. Patients with more extensive lung parenchymal lesions (≥ 75%) on admission had higher CRP serum levels. The extent of lung parenchymal lesions was associated with a decrease in the PaO2/FiO2 ratio(p<0.01), fewer ventilatory-free days (p = 0.03), and a higher death rate at day 60(p = 0.01). Extent of the lesion of more than 75% was independently associated with day-60 mortality (aHR = 1.72[1.06; 2.78], p = 0.03). The prediction of death at day 60 was improved when considering simultaneously biological and radiological markers obtained on ICU admission (AUC = 0.78). CONCLUSIONS The extent of lung parenchyma lesions on CT was associated with inflammation, and the combination of coagulation and inflammatory biomarkers and the extent of the lesions predicted the poorest outcomes.
Collapse
Affiliation(s)
- Abed al Hadi Krisht
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Kévin Grapin
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | | | - Benjamin Bonnet
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mireille Adda
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, CNRS, LMGE, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| |
Collapse
|
37
|
Davidson CB, El Sabbagh DES, Machado AK, Pappis L, Sagrillo MR, Somacal S, Emanuelli T, Schultz JV, Augusto Pereira da Rocha J, Santos AFD, Fagan SB, Silva IZD, Andreazza AC, Machado AK. Euterpe oleracea Mart. Bioactive Molecules: Promising Agents to Modulate the NLRP3 Inflammasome. BIOLOGY 2024; 13:729. [PMID: 39336156 PMCID: PMC11428631 DOI: 10.3390/biology13090729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024]
Abstract
Inflammation is a vital mechanism that defends the organism against infections and restores homeostasis. However, when inflammation becomes uncontrolled, it leads to chronic inflammation. The NLRP3 inflammasome is crucial in chronic inflammatory responses and has become a focal point in research for new anti-inflammatory therapies. Flavonoids like catechin, apigenin, and epicatechin are known for their bioactive properties (antioxidant, anti-inflammatory, etc.), but the mechanisms behind their anti-inflammatory actions remain unclear. This study aimed to explore the ability of various flavonoids (isolated and combined) to modulate the NLRP3 inflammasome using in silico and in vitro models. Computer simulations, such as molecular docking, molecular dynamics, and MM/GBSA calculations examined the interactions between bioactive molecules and NLRP3 PYD. THP1 cells were treated with LPS + nigericin to activate NLRP3, followed by flavonoid treatment at different concentrations. THP1-derived macrophages were also treated following NLRP3 activation protocols. The assays included colorimetric, fluorometric, microscopic, and molecular techniques. The results showed that catechin, apigenin, and epicatechin had high binding affinity to NLRP3 PYD, similar to the known NLRP3 inhibitor MCC950. These flavonoids, particularly at 1 µg/mL, 0.1 µg/mL, and 0.01 µg/mL, respectively, significantly reduced LPS + nigericin effects in both cell types and decreased pro-inflammatory cytokine, caspase-1, and NLRP3 gene expression, suggesting their potential as anti-inflammatory agents through NLRP3 modulation.
Collapse
Affiliation(s)
- Carolina Bordin Davidson
- Graduate Program in Nanosciences, Franciscan University, Santa Maria 97010-030, RS, Brazil
- Laboratory of Cell Culture and Bioactive Effects, Franciscan University, Santa Maria 97010-030, RS, Brazil
| | | | - Amanda Kolinski Machado
- Laboratory of Cell Culture and Bioactive Effects, Franciscan University, Santa Maria 97010-030, RS, Brazil
| | - Lauren Pappis
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
| | | | - Sabrina Somacal
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Tatiana Emanuelli
- Department of Technology and Food Science, Federal University of Santa Maria, Santa Maria 97105-900, RS, Brazil
| | - Júlia Vaz Schultz
- Graduate Program in Nanosciences, Franciscan University, Santa Maria 97010-030, RS, Brazil
| | - João Augusto Pereira da Rocha
- Federal Institute of Pará, Bragança Campus, Computational Chemistry and Modeling Laboratory, Bragança 68600-000, PA, Brazil
| | | | - Solange Binotto Fagan
- Graduate Program in Nanosciences, Franciscan University, Santa Maria 97010-030, RS, Brazil
| | - Ivana Zanella da Silva
- Graduate Program in Nanosciences, Franciscan University, Santa Maria 97010-030, RS, Brazil
| | - Ana Cristina Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada
| | - Alencar Kolinski Machado
- Graduate Program in Nanosciences, Franciscan University, Santa Maria 97010-030, RS, Brazil
- Laboratory of Cell Culture and Bioactive Effects, Franciscan University, Santa Maria 97010-030, RS, Brazil
| |
Collapse
|
38
|
Li J, Lyu C, An R, Wang D. Interaction Between SARS-CoV-2 Spike Protein S1 Subunit and Oyster Heat Shock Protein 70. FOOD AND ENVIRONMENTAL VIROLOGY 2024; 16:380-390. [PMID: 38635140 DOI: 10.1007/s12560-024-09599-y] [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: 10/17/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
There is growing evidence that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) contaminates the marine environment and is bioaccumulated in filter-feeding shellfish. Previous study shows the Pacific oyster tissues can bioaccumulate the SARS-CoV-2, and the oyster heat shock protein 70 (oHSP70) may play as the primary attachment receptor to bind SARS-CoV-2's recombinant spike protein S1 subunit (rS1). However, detailed information about the interaction between rS1 and oHSP70 is still unknown. In this study, we confirmed that the affinity of recombinant oHSP70 (roHSP70) for rS1 (KD = 20.4 nM) is comparable to the receptor-binding affinity of rACE2 for rS1 (KD = 16.7 nM) by surface plasmon resonance (SPR)-based Biacore and further validated by enzyme-linked immunosorbent assay (ELISA). Three truncated proteins (roHSP70-N/C/M) and five mutated proteins (p.I229del, p.D457del, p.V491_K495del, p.K556I, and p.ΣroHSP70) were constructed according to the molecular docking results. All three truncated proteins have significantly lower affinity for rS1 than the full-length roHSP70, indicating that all three segments of roHSP70 are involved in binding to rS1. Further, the results of SPR and ELISA showed that all five mutant proteins had significantly lower affinity for rS1 than roHSP70, suggesting that amino acids at these sites are involved in binding to rS1. This study provides a preliminary theoretical basis for the bioaccumulation of SARS-CoV-2 in oyster tissues or using roHSP70 as the capture unit to selectively enrich virus particles for detection.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chenang Lyu
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Ran An
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Dapeng Wang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
39
|
Groves AM, Paris ND, Johnston CJ, Hernady E, Finkelstein J, Lawrence P, Marples B. Mitigating Viral Impact on the Radiation Response of the Lung. Radiat Res 2024; 202:552-564. [PMID: 39048109 PMCID: PMC11610374 DOI: 10.1667/rade-24-00103.1] [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: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024]
Abstract
Inflammation is a key factor in both influenza and radiation-induced lung pathophysiology. This implies a commonality of response to pulmonary damage from these insults and suggests exacerbated pathology may occur after combined exposure. We therefore tested the hypothesis that past inflammation from viral infection alters the lung microenvironment and lowers tolerance for radiation injury. Mice were inoculated with influenza A virus (IAV) and three weeks later, after virus clearance, mice received total-body irradiation (TBI). Survival as well as systemic and local lung inflammation were assessed, and strategies to mitigate pulmonary injury were investigated. After IAV infection alone, body condition recovered within 3 weeks, however inflammatory pathways remained active for 15 weeks. IAV infection exacerbated subsequent TBI responses, evident by increased lethality, enhanced histologically evident lung injury and an altered lung macrophage phenotype. To mitigate this enhanced sensitivity, captopril [an angiotensin converting enzyme inhibitor (ACEi)] was administered to limit tissue inflammation, or inflammatory monocyte-derived macrophage recruitment was blocked with a C-C chemokine receptor type 2 (CCR2) inhibitor. Both treatments abrogated the changes in circulating immune cells observed 4 weeks after TBI, and attenuated pro-inflammatory phenotypes in lung alveolar macrophages, appearing to shift immune cell dynamics towards recovery. Histologically apparent lung injury was not improved by either treatment. We show that latent lung injury from viral infection exacerbates radiation morbidity and mortality. Although strategies that attenuate proinflammatory immune cell phenotypes can normalize macrophage dynamics, this does not fully mitigate lung injury. Recognizing that past viral infections can enhance lung radiosensitivity is of critical importance for patients receiving TBI, as it could increase the incidence of adverse outcomes.
Collapse
Affiliation(s)
- Angela M. Groves
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Nicole D. Paris
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Carl J. Johnston
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Jacob Finkelstein
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Paige Lawrence
- Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| |
Collapse
|
40
|
Compeer B, Neijzen TR, van Lelyveld SFL, Martina BEE, Russell CA, Goeijenbier M. Uncovering the Contrasts and Connections in PASC: Viral Load and Cytokine Signatures in Acute COVID-19 versus Post-Acute Sequelae of SARS-CoV-2 (PASC). Biomedicines 2024; 12:1941. [PMID: 39335455 PMCID: PMC11428903 DOI: 10.3390/biomedicines12091941] [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: 07/09/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
The recent global COVID-19 pandemic has had a profound and enduring impact, resulting in substantial loss of life. The scientific community has responded unprecedentedly by investigating various aspects of the crisis, particularly focusing on the acute phase of COVID-19. The roles of the viral load, cytokines, and chemokines during the acute phase and in the context of patients who experienced enduring symptoms upon infection, so called Post-Acute Sequelae of COVID-19 or PASC, have been studied extensively. Here, in this review, we offer a virologist's perspective on PASC, highlighting the dynamics of SARS-CoV-2 viral loads, cytokines, and chemokines in different organs of patients across the full clinical spectrum of acute-phase disease. We underline that the probability of severe or critical disease progression correlates with increased viral load levels detected in the upper respiratory tract (URT), lower respiratory tract (LRT), and plasma. Acute-phase viremia is a clear, although not unambiguous, predictor of PASC development. Moreover, both the quantity and diversity of functions of cytokines and chemokines increase with acute-phase disease severity. Specific cytokines remain or become elevated in the PASC phase, although the driving factor of ongoing inflammation found in patients with PASC remains to be investigated. The key findings highlighted in this review contribute to a further understanding of PASC and their differences and overlap with acute disease.
Collapse
Affiliation(s)
- Brandon Compeer
- Artemis Bioservices B.V., 2629 JD Delft, The Netherlands
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Tobias R Neijzen
- Department of Intensive Care Medicine, Spaarne Gasthuis, 2035 RC Haarlem, The Netherlands
| | | | | | - Colin A Russell
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
| | - Marco Goeijenbier
- Department of Medical Microbiology, University Medical Center Amsterdam (UMC, Amsterdam), 1105 AZ Amsterdam, The Netherlands
- Department of Intensive Care, Erasmus MC University Medical Centre, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
41
|
Yao L, Wang L, Liu S, Qu H, Mao Y, Li Y, Zheng L. Evolution of a bispecific G-quadruplex-forming circular aptamer to block IL-6/sIL-6R interaction for inflammation inhibition. Chem Sci 2024; 15:13011-13020. [PMID: 39148786 PMCID: PMC11323322 DOI: 10.1039/d4sc02183e] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
IL-6 (interleukin-6) is an essential cytokine that participates in many inflammatory and immune responses, and disrupting the interaction between IL-6 and its receptor sIL-6R (soluble form of IL-6 receptor) represents a promising treatment strategy for inflammation and related diseases. Herein we report the first-ever effort of evolving a bispecific circular aptamer, named CIL-6A6-1, that is capable of binding both IL-6 and sIL-6R with nanomolar affinities and is stable in serum for more than 48 hours. CIL-6A6-1 can effectively block the IL-6/sIL-6R interaction and significantly inhibit cell inflammation. Most importantly, this bispecific aptamer is much more effective than aptamers that bind IL-6 and sIL-6R alone as well as tocilizumab, a commercially available humanized monoclonal antibody against sIL-6R, highlighting the advantage of selecting bispecific circular aptamers as molecular tools for anti-inflammation therapy. Interestingly, CIL-6A6-1 is predicted to adopt a unique structural fold with two G-quadruplex motifs capped by a long single-stranded region, which differs from all known DNA aptamers. This unique structural fold may also contribute to its excellent functionality and high stability in biological complex media. We anticipate that our study will represent a significant step forward towards demonstrating the practical utility of bispecific DNA aptamers for therapeutic applications.
Collapse
Affiliation(s)
- Lili Yao
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| | - Lei Wang
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| | - Shuai Liu
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| | - Hao Qu
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| | - Yu Mao
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University Hamilton L8S4K1 Canada
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology Hefei 230009 China
| |
Collapse
|
42
|
Antar SA, Ashour NA, Hamouda AO, Noreddin AM, Al-Karmalawy AA. Recent advances in COVID-19-induced liver injury: causes, diagnosis, and management. Inflammopharmacology 2024:10.1007/s10787-024-01535-7. [PMID: 39126569 DOI: 10.1007/s10787-024-01535-7] [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: 10/14/2023] [Accepted: 03/29/2024] [Indexed: 08/12/2024]
Abstract
Since the start of the pandemic, considerable advancements have been made in our understanding of the effects of SARS-CoV-2 infection and the associated COVID-19 on the hepatic system. There is a broad range of clinical symptoms for COVID-19. It affects multiple systems and has a dominant lung illness depending on complications. The progression of COVID-19 in people with pre-existing chronic liver disease (CLD) has also been studied in large multinational groups. Notably, SARS-CoV-2 infection is associated with a higher risk of hepatic decompensation and death in patients with cirrhosis. In this review, the source, composition, mechanisms, transmission characteristics, clinical characteristics, therapy, and prevention of SARS-CoV-2 were clarified and discussed, as well as the evolution and variations of the virus. This review briefly discusses the causes and effects of SARS-CoV-2 infection in patients with CLD. As part of COVID-19, In addition, we assess the potential of liver biochemistry as a diagnostic tool examine the data on direct viral infection of liver cells, and investigate potential pathways driving SARS-CoV-2-related liver damage. Finally, we explore how the pandemic has had a significant impact on patient behaviors and hepatology services, which may increase the prevalence and severity of liver disease in the future. The topics encompassed in this review encompass the intricate relationships between SARS-CoV-2, liver health, and broader health management strategies, providing valuable insights for both current clinical practice and future research directions.
Collapse
Affiliation(s)
- Samar A Antar
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, 24016, USA
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Nada A Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Amir O Hamouda
- Department of Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Ayman M Noreddin
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, 12566, Egypt
- Department of Internal Medicine, School of Medicine, University of California -Irvine, Irvine, USA
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, New Damietta, 34518, Egypt.
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6Th of October City, Giza, 12566, Egypt.
| |
Collapse
|
43
|
Smaani B, Nafa F, Benlatrech MS, Mahdi I, Akroum H, walid Azizi M, Harrar K, Kanungo S. Recent progress on field-effect transistor-based biosensors: device perspective. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:977-994. [PMID: 39136041 PMCID: PMC11318611 DOI: 10.3762/bjnano.15.80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024]
Abstract
Over the last few decades, field-effect transistor (FET)-based biosensors have demonstrated great potential across various industries, including medical, food, agriculture, environmental, and military sectors. These biosensors leverage the electrical properties of transistors to detect a wide range of biomolecules, such as proteins, DNA, and antibodies. This article presents a comprehensive review of advancements in the architectures of FET-based biosensors aiming to enhance device performance in terms of sensitivity, detection time, and selectivity. The review encompasses an overview of emerging FET-based biosensors and useful guidelines to reach the best device dimensions, favorable design, and realization of FET-based biosensors. Consequently, it furnishes researchers with a detailed perspective on design considerations and applications for future generations of FET-based biosensors. Finally, this article proposes intriguing avenues for further research on the topology of FET-based biosensors.
Collapse
Affiliation(s)
- Billel Smaani
- Abdelhafid Boussouf University Centre of Mila, Mila, Algeria
| | - Fares Nafa
- University of Jijel, Automation Department, Jijel, Algeria
| | | | - Ismahan Mahdi
- Laboratoire de Recherche Electrification des Entreprises Industrilles (LREEI), Faculté des Hydrocarbures et de la Chimie, Université M’Hamed Bougara Boumerdes, Algeria
| | - Hamza Akroum
- LIST Laboratory, University M’Hamed Bougara, Boumerdes, Algeria
| | | | - Khaled Harrar
- LIST Laboratory, University M’Hamed Bougara, Boumerdes, Algeria
| | - Sayan Kanungo
- Department of Electrical and Electronics Engineering Birla Institute of Technology and Science Pilani, Hyderabad, India
| |
Collapse
|
44
|
Liu X, Zheng M, Zhang H, Feng B, Li J, Zhang Y, Zhang J, Zhao N, Li C, Song N, Song B, Yang D, Chen J, Qi A, Zhao L, Luo C, Zang Y, Liu H, Li J, Zhang B, Zhou Y, Zheng J. Characterization and noncovalent inhibition of the K63-deubiquitinase activity of SARS-cov-2 PLpro. Antiviral Res 2024; 228:105944. [PMID: 38914283 DOI: 10.1016/j.antiviral.2024.105944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/26/2024]
Abstract
SARS-CoV-2 papain-like protease (PLpro) could facilitate viral replication and host immune evasion by respectively hydrolyzing viral polyprotein and host ubiquitin conjugates, thereby rendering itself as an important antiviral target. Yet few noncovalent PLpro inhibitors of SARS-CoV-2 have been reported with improved directed towards pathogenic deubiquitinating activities inhibition. Herein, we report that coronavirus PLpro proteases have distinctive substrate bias and are conserved to deubiquitylate K63-linked polyubiquitination, thereby attenuating host type I interferon response. We identify a noncovalent compound specifically optimized towards halting the K63-deubiquitinase activity of SARS-CoV-2 PLpro, but not other coronavirus (CoV) counterparts or host deubiquitinase. Contrasting with GRL-0617, a SARS-CoV-1 PLpro inhibitor, SIMM-036 is 50-fold and 7-fold (half maximal inhibitory concentration (IC50)) more potent to inhibit viral replication during SARS-CoV-2 infection and restore the host interferon-β (IFN-β) response in human angiotensin-converting enzyme 2 (hACE2)-HeLa cells, respectively. Structure-activity relationship (SAR) analysis further reveals the importance of BL2 groove of PLpro, which could determine the selectivity of K63-deubiquitinase activity of the enzyme.
Collapse
Affiliation(s)
- Xin Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Miao Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hongqing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bo Feng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiaqi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yanan Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Ji Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Na Zhao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chaoqiang Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ning Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bin Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dongyuan Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jin Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ao Qi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linxiang Zhao
- Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Cheng Luo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Zang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hong Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yu Zhou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Shenyang Pharmaceutical University, Shenyang, 110016, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 310024, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shanghai Institute of Virology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
45
|
Du Q, Liang R, Wu M, Yang M, Xie Y, Liu Q, Tang K, Lin X, Yuan S, Shen J. Alisol B 23-acetate broadly inhibits coronavirus through blocking virus entry and suppresses proinflammatory T cells responses for the treatment of COVID-19. J Adv Res 2024; 62:273-290. [PMID: 37802148 PMCID: PMC11331179 DOI: 10.1016/j.jare.2023.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/11/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023] Open
Abstract
INTRODUCTION Emerging severe acute respiratory syndrome (SARS) coronavirus (CoV)-2 causes a global health disaster and pandemic. Seeking effective anti-pan-CoVs drugs benefit critical illness patients of coronavirus disease 2019 (COVID-19) but also may play a role in emerging CoVs of the future. OBJECTIVES This study tested the hypothesis that alisol B 23-acetate could be a viral entry inhibitor and would have proinflammatory inhibition for COVID-19 treatment. METHODS SARS-CoV-2 and its variants infected several cell lines were applied to evaluate the anti-CoVs activities of alisol B 23-aceate in vitro. The effects of alisol B 23-acetate on in vivo models were assessed by using SARS-CoV-2 and its variants challenged hamster and human angiotensin-converting enzyme 2 (ACE2) transgenic mice. The target of alisol B 23-acetate to ACE2 was analyzed using hydrogen/deuterium exchange (HDX) mass spectrometry (MS). RESULTS Alisol B 23-acetate had inhibitory effects on different species of coronavirus. By using HDX-MS, we found that alisol B 23-acetate had inhibition potency toward ACE2. In vivo experiments showed that alisol B 23-acetate treatment remarkably decreased viral copy, reduced CD4+ T lymphocytes and CD11b+ macrophages infiltration and ameliorated lung damages in the hamster model. In Omicron variant infected human ACE2 transgenic mice, alisol B 23-acetate effectively alleviated viral load in nasal turbinate and reduced proinflammatory cytokines interleukin 17 (IL17) and interferon γ (IFNγ) in peripheral blood. The prophylactic treatment of alisol B 23-acetate by intranasal administration significantly attenuated Omicron viral load in the hamster lung tissues. Moreover, alisol B 23-acetate treatment remarkably inhibited proinflammatory responses through mitigating the secretions of IFNγ and IL17 in the cultured human and mice lymphocytes in vitro. CONCLUSION Alisol B 23-acetate could be a promising therapeutic agent for COVID-19 treatment and its underlying mechanisms might be attributed to viral entry inhibition and anti-inflammatory activities.
Collapse
Affiliation(s)
- Qiaohui Du
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, Hong Kong Special Administrative Region
| | - Ronghui Liang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region
| | - Minxiao Yang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region
| | - Yubin Xie
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Qing Liu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region
| | - Kaiming Tang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Xiang Lin
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China.
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, Hong Kong Special Administrative Region; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
46
|
Einollahi B, Javanbakht M, Ebrahimi M, Ahmadi M, Izadi M, Ghasemi S, Einollahi Z, Beyram B, Mirani A, Kianfar E. Surveying haemoperfusion impact on COVID-19 from machine learning using Shapley values. Inflammopharmacology 2024; 32:2285-2294. [PMID: 38762840 DOI: 10.1007/s10787-024-01494-z] [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: 04/02/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Haemoperfusion (HP) is an innovative extracorporeal therapy that utilizes special cartridges to filter the blood, effectively removing pro-inflammatory cytokines, toxins, and pathogens in COVID-19 patients. This retrospective cohort study aimed to assess the clinical benefits of HP for severe COVID-19 cases using Shapley values for machine learning models. METHODS The research involved 578 inpatients (≥ 20 years old) admitted to Baqiyatallah hospital (Tehran, Iran). The control group (359 patients) received standard treatment, including high doses of corticosteroids (a single 500 mg methylprednisolone pulse, followed by 250 mg for 2 days), categorized as regimen (I). On the other hand, the HP group (219 patients) received regimen II, consisting of the same corticosteroid treatment (regimen I) along with haemoperfusion using Cytosorb H300. The frequency of haemoperfusion sessions varied based on the type of lung involvement determined by chest CT scans. In addition, the value function v defines the Shapley value of the i th feature for the query point x , where the input matrix features represent individual characteristics, drugs, and history and clinical conditions of the patient. RESULTS Our data showed a favorable clinical response in the HP group compared to the control group. Notably, one-to-three sessions of HP using the CytoSorb® 300 cartridge led to reduced ventilation requirements and mortality rates in severe COVID-19 patients. Shapley values were calculated to evaluate the contribution of haemoperfusion among other factors, such as side effects, medications, and individual characteristics, to COVID-19 patient outcomes. In addition, there is a significant difference between the two groups among the treatments and medications used remdesivir, adalimumab, tocilizumab, favipiravir, Interferon beta-1a, enoxaparin prophylaxis, enoxaparin full dose, heparin prophylaxis, and heparin full dose (P < 0.05). It seems that haemoperfusion has a positive impact on the reduction of inflammation markers and renal functional such as ferritin and creatinine, respectively, as well as D-dimer and WBC levels in the HP group were significantly lower than the control group. CONCLUSION The findings indicated that haemoperfusion played a crucial role in predicting patient survival, making it a significant feature in classifying patients' prognoses.
Collapse
Affiliation(s)
- Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Ebrahimi
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ahmadi
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Morteza Izadi
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sholeh Ghasemi
- Department of Nephrology, Shahid Hasheminejad Kidney Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Einollahi
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bentolhoda Beyram
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Mirani
- Biomedical Engineering Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ehsan Kianfar
- Biomedical Engineering Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Liu X, Zhou M, Fang M, Xie Y, Chen P, Chen R, Wu K, Ye J, Liu C, Zhu H, Cheng T, Yuan L, Zhao H, Guan Y, Xia N. Decisive reversal of lethal coronavirus disease 2019 in senescent hamster by synchronic antiviral and immunoregulatory intervention. MedComm (Beijing) 2024; 5:e642. [PMID: 39036342 PMCID: PMC11258460 DOI: 10.1002/mco2.642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/23/2024] Open
Abstract
The poor prognosis observed in elderly individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a serious clinical burden and the underlying mechanism is unclear, which necessities detailed investigation of disease characteristics and research for efficient countermeasures. To simulate lethal coronavirus disease 2019 (COVID-19) in senescent human patients, 80-week-old male hamsters are intranasally inoculated with different doses of SARS-CoV-2 Omicron BA.5 variant. Exposure to a low dose of the Omicron BA.5 variant results in early activation of the innate immune response, followed by rapid viral clearance and minimal lung damage. However, a high dose of BA.5 results in impaired interferon signaling, cytokine storm, uncontrolled viral replication, and severe lung injury. To decrease viral load and reverse the deterioration of COVID-19, a new bio-mimic decoy called CoVR-MV is used as a preventive or therapeutic agent. Administration of CoVR-MV as a preventive or therapeutic intervention in the early stages of infection can effectively suppress viral load, regulate the immune response, and rescue animals from death and critical illness. These findings underscore the risk associated with SARS-CoV-2 Omicron BA.5 exposure in senescent hamsters and highlight the importance of early intervention to prevent disease progression.
Collapse
Affiliation(s)
- Xuan Liu
- Clinical Center for Bio‐TherapyZhongshan HospitalFudan University (Xiamen Branch)XiamenFujianChina
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Ming Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Mujing Fang
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Ying Xie
- National Institute for Food and Drug ControlBeijingChina
- Institute of Medical BiologyChinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
| | - Peiwen Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- Guangdong‐Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU)Shantou UniversityShantouGuangdongChina
| | - Rirong Chen
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- Guangdong‐Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU)Shantou UniversityShantouGuangdongChina
| | - Kun Wu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Jianghui Ye
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Che Liu
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Huachen Zhu
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- Guangdong‐Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU)Shantou UniversityShantouGuangdongChina
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Hui Zhao
- National Institute for Food and Drug ControlBeijingChina
| | - Yi Guan
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- Guangdong‐Hong Kong Joint Laboratory of Emerging Infectious Diseases/Joint Laboratory for International Collaboration in Virology and Emerging Infectious Diseases, Joint Institute of Virology (STU/HKU)Shantou UniversityShantouGuangdongChina
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, School of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| |
Collapse
|
48
|
Aminsobahni E, Hosseini M, Gholizadeh N, Soltani-Zangbar MS, Savari G, Motlagh Asghari K, Pourlak T, Zolfaghari M, Chakari-Khiavi F, Motavalli R, Chakari-Khiavi A, Shekarchi AA, Mahmoodpoor A, Ahmadian Heris J, Pouya K, Mehdizadeh A, Babalou Z, Yousefi M. T Lymphocyte Characteristic Changes Under Serum Cytokine Deviations and Prognostic Factors of COVID-19 in Pregnant Women. Appl Biochem Biotechnol 2024; 196:4366-4381. [PMID: 37947946 DOI: 10.1007/s12010-023-04775-5] [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] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Physiological changes during pregnancy make the individuals more susceptible to severe respiratory diseases. Hence, pregnant women with coronavirus disease 2019 (COVID-19) are likely at a higher risk. We investigated the effects of COVID-19 on T cell response and serum cytokine profile in pregnant patients. Peripheral blood mononuclear cells (PBMCs) of women with COVID-19 were collected during the first trimester of pregnancy, and the percentage of total lymphocytes, as well as CD4 + and CD8 + T cells, was assessed using flow cytometry. The expression of the programmed death-1 (PD-1) marker for exhausted T cells was evaluated. Additionally, the serum samples were provided to evaluate the levels of antiviral and proinflammatory cytokines, as well as laboratory serological tests. Pregnant women with COVID-19 presented lymphopenia with diminished CD4 + and CD8 + T cells. Besides, high expression levels of the PD-1 gene and protein were observed on PBMCs and T cells, respectively, when compared with normal pregnant individuals. Moreover, serum levels of TNF-α, IL-6, IL-1β, and IL-2 receptor were notably enhanced, while IFN-I α/β values were significantly decreased in the patients when compared with controls. Furthermore, hyperlipidemia, hyperglycemia, and hypertension were directly correlated with the disease although serum albumin and vitamin D3 levels adversely affected the viral infection. Our study showed extreme lymphopenia and poor T cell response while elevated values of serum inflammatory cytokines in infected pregnant women. Moreover, a hypertension background or metabolic changes, including hyperlipidemia, hyperglycemia, and vitamin D3 or albumin deficiency, might be promising prognostic factors in pregnant women with COVID-19.
Collapse
Affiliation(s)
- Ehsan Aminsobahni
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hosseini
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Golaleh Savari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Tannaz Pourlak
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali Zolfaghari
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Roza Motavalli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aref Chakari-Khiavi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Pouya
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babalou
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Pena NM, Santana LC, Hunter JR, Blum VF, Vergara T, Gouvea C, Leal E, Bellei N, Schechter M, Diaz RS. T cell-mediated Immune response and correlates of inflammation and their relationship with COVID-19 clinical severity: not an intuitive guess. BMC Infect Dis 2024; 24:612. [PMID: 38902613 PMCID: PMC11191252 DOI: 10.1186/s12879-024-09490-y] [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: 05/01/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Predictors of the outcome of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection remain to be fully determined. We evaluated selected viral characteristics and immunological responses that might predict and/or correlate to the clinical outcome of COVID-19. METHODS For individuals developing divergent clinical outcomes, the magnitude and breadth of T cell-mediated responses were measured within 36 h of symptom onset. Peripheral Blood Mononuclear Cells (PBMCs) were subjected to in vitro stimulation with SARS-CoV-2-based peptides. In addition, SARS-CoV-2 sequences were generated by metagenome, and HLA typing was performed using Luminex technology. FINDINGS CD4+ T cell activation was negatively correlated with SARS-CoV-2 basal viral load in patients with severe COVID-19 (p = 0·043). The overall cellular immune response, as inferred by the IFN-γ signal, was higher at baseline for patients who progressed to mild disease compared to patients who progressed to severe disease (p = 0·0044). Subjects with milder disease developed higher T cell responses for MHC class I and II-restricted peptides (p = 0·033). INTERPRETATION Mounting specific cellular immune responses in the first days after symptom onset, as inferred by IFN-γ magnitude in the ELISPOT assay, may efficiently favor a positive outcome. In contrast, progression to severe COVID-19 was accompanied by stronger cellular immune responses, higher CD4 + T cell activation, and a higher number of in silico predicted high-affinity class I HLA alleles.
Collapse
Affiliation(s)
- Nathalia Mantovani Pena
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Weill Cornell Medicine, New York, United States of America
| | - Luiz Claudio Santana
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - James R Hunter
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Vinicius Fontanesi Blum
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Tania Vergara
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Oncohiv, Rio de Janeiro, Brazil
| | - Celso Gouvea
- Centro de Hematologia e Hemoterapia do Ceará, Fortaleza, CE, Brazil
| | - Elcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belem, Pará, Brazil
| | - Nancy Bellei
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Mauro Schechter
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
50
|
Berger L, Wolf J, Kalbitz S, Kellner N, Lübbert C, Borte S. Comparative Analysis of Lymphocyte Populations in Post-COVID-19 Condition and COVID-19 Convalescent Individuals. Diagnostics (Basel) 2024; 14:1286. [PMID: 38928701 PMCID: PMC11202600 DOI: 10.3390/diagnostics14121286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Reduced lymphocyte counts in peripheral blood are one of the most common observations in acute phases of viral infections. Although many studies have already examined the impact of immune (dys)regulation during SARS-CoV-2 infection, there are still uncertainties about the long-term consequences for lymphocyte homeostasis. Furthermore, as persistent cellular aberrations have been described following other viral infections, patients with "Post-COVID-19 Condition" (PCC) may present similarly. In order to investigate cellular changes in the adaptive immune system, we performed a retrospective analysis of flow cytometric data from lymphocyte subpopulations in 106 patients with confirmed SARS-CoV-2 infection who received medical care at our institution. The patients were divided into three groups according to the follow-up date; laboratory analyses of COVID-19 patients were compared with 28 unexposed healthy controls. Regarding B lymphocyte subsets, levels of IgA + CD27+, IgG + CD27+, IgM + CD27- and switched B cells were significantly reduced at the last follow-up compared to unexposed healthy controls (UHC). Of the 106 COVID-19 patients, 56 were clinically classified as featuring PCC. Significant differences between PCC and COVID-19 convalescents compared to UHC were observed in T helper cells and class-switched B cells. However, we did not detect specific or long-lasting immune cellular changes in PCC compared to the non-post-COVID-19 condition.
Collapse
Affiliation(s)
- Luisa Berger
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Johannes Wolf
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Sven Kalbitz
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
| | - Nils Kellner
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| | - Christoph Lübbert
- Department of Infectious Diseases and Tropical Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Stephan Borte
- Department of Laboratory Medicine, Hospital St. Georg, 04129 Leipzig, Germany
- ImmunoDeficiencyCenter Leipzig, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiency Diseases, Hospital St. Georg, 04139 Leipzig, Germany
| |
Collapse
|