1
|
Amrouche T, Lammi S, Drider D. Probiotics and Prebiotics Intervention in Respiratory and Digestive Infections Linked to Covid-19. Probiotics Antimicrob Proteins 2025; 17:1356-1367. [PMID: 39614066 DOI: 10.1007/s12602-024-10404-2] [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] [Accepted: 11/11/2024] [Indexed: 12/01/2024]
Abstract
Probiotics and prebiotics have been suggested as natural agents against viral infections and dysbiosis and may encourage clinical applications. This review aims to analyze the main and recent advances related to viral infections such as Covid-19 and its gastrointestinal complications, antiviral immunity generated and possible preventive role that probiotics and/or prebiotics can play in controlling and promoting antiviral immunity. The literature search was performed through a critical analysis of relevant publications reported in PubMed and Scopus databases on clinical trials and assays conducted in vitro on colon cells and in vivo on mice. Some studies using probiotics and prebiotics for the prevention of viral infection in different age groups are discussed. Covid-19 patients have been shown to suffer from gastrointestinal complications in addition to respiratory symptoms due to interactions between the respiratory system and the gastrointestinal tract infected with SARS-CoV-2. Unfortunately, therapies used to prevent (or treat) symptoms of Covid-19 have proven to be of limited effectiveness. In addition, the lack of access to coronavirus vaccines around the world and vaccine hesitancy continue to hamper control of Covid-19. It is therefore crucial to find alternative methods that can prevent disease symptoms. Evidence-based efficacy of certain probiotics (Lactobacillus and Bifidobacterium) that may be useful in viral infections was shown with immunomodulatory properties (pro-inflammatory mediators reduction), promoting antiviral immunity (antibodies production, virus titers) and controlling inflammation (anti-inflammatory effect), as well as viral clearance and antimicrobial potential against opportunistic bacteria (anti-dysbiosis effect). But, available data about clinical application of probiotics in Covid-19 context remain limited and relevant scientific investigation is still in its early stages. Also, evidence for prebiotics potential in this field is limited, since the exact mechanism involved in systemic immune modulation by these compounds is till now unknown. Thus, further research is necessary to explore in the viral infection context the mechanism by which gut and lung interact in the presence of probiotics and prebiotics through more animal and clinical experiments.
Collapse
Affiliation(s)
- Tahar Amrouche
- Laboratoire Qualité Et Sécurité Des Aliments, Faculté Des Sciences Biologiques Et Des Sciences Agronomiques, Université Mouloud Mammeri, 15 000, Tizi Ouzou, Algeria.
| | - Sarah Lammi
- Laboratoire Qualité Et Sécurité Des Aliments, Faculté Des Sciences Biologiques Et Des Sciences Agronomiques, Université Mouloud Mammeri, 15 000, Tizi Ouzou, Algeria
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro INRAE 1158, Université de Lille (ULille), 59000, Lille, France
| |
Collapse
|
2
|
Chen L, Li W, Zai W, Zheng X, Meng X, Yao Q, Li W, Liang Y, Ye M, Zhou K, Liu M, Yang Z, Mao Z, Wei H, Yang S, Shi G, Yuan Z, Yu W. HBV sequence integrated to enhancer acting as oncogenic driver epigenetically promotes hepatocellular carcinoma development. J Exp Clin Cancer Res 2025; 44:155. [PMID: 40405227 PMCID: PMC12096768 DOI: 10.1186/s13046-025-03413-8] [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: 03/08/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND HBV integration is considered as the main contributor to hepatocellular carcinoma (HCC). However, whether HBV integrated sequences determine genotype pathogenicity and how to block their function during HCC progression remains unclear. METHODS An in vitro HBV-infected PHH model and liver cancer cell lines were established to confirm the pathogenic potential of HBV-SITEs. The roles of HBV-SITE-1 in HCC development were analyzed using cellular phenotypic assays and molecular biology techniques, including the combined analysis of RNA-seq and ChIP-seq. Animal models were also used to evaluate the therapeutic effect of HBV-miR-2 inhibitors. RESULTS We identified nine fragments of HBV Sequences Integrated To Enhancer, termed as "HBV-SITEs". Particularly, a single nucleotide variation (T > G) was embedded at seed sequence of HBV-miR-2 in the highest integrated HBV-SITE-1 between genotypes B and H. Unexpectedly, B-HBV-SITE-1, not H-HBV-SITE-1, could abnormally activate oncogenic genes including TERT and accelerate HCC cell proliferation and migration. Meanwhile, HBV-miR-2 was gradually increased in HBV-infected cells and patient plasma with different HCC stages. Importantly, 227 genes upregulated by HBV, were also activated by HBV-miR-2 through triggering HBV-SITE-1 enhancer. Conversely, enhancer activities were particularly decreased by HBV-miR-2 inhibitors, and further downregulated activated oncogenic genes. Finally, HCC growth was dramatically restrained and HBV-induced transcripts were systematically reduced via injection of HBV-miR-2 inhibitors in animal models. CONCLUSION HBV-SITEs were identified as novel oncogenic elements for HCC, which provides an insightful perspective for the other cancers caused by oncogenic DNA viruses. We demonstrated that the integrated HBV sequence itself acted as oncogenic enhancers and nucleotide variations of HBV genotypes account for particular pathogenic progression, supporting that the viral nucleotide sequences are vital pathogenic substances beyond viral proteins. And modulation of their enhancer activities could be clinically achievable strategy for blocking DNA viruses-related cancer progression in the future.
Collapse
Affiliation(s)
- Lu Chen
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxuan Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangyi Zheng
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianlong Meng
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qunyan Yao
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Liang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mu Ye
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kaicheng Zhou
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengxing Liu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhicong Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhanrui Mao
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyan Wei
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
- Research and Development Department, Shanghai Epicurer Biotechnology Co., Ltd., Shanghai, China.
| | - Guoming Shi
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Kang H, Chen Y, Cheng M, Guo H, Zhang G, Shi Q, Zhou W, Zhao C, Zou B, Lv X, Yuan Z, Zeng G. State-Of-The-Art Structural Regulation Methods and Quantum Chemistry for Carbon-Based Single-Atom Catalysts in Advanced Oxidation Process: Critical Perspectives into Molecular Level. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2505128. [PMID: 40401577 DOI: 10.1002/adma.202505128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/24/2025] [Indexed: 05/23/2025]
Abstract
Advanced oxidation processes (AOPs) by carbon-based single-atom catalysts (SACs) are recognized as an attractive scientific frontier for water treatment, with the outstanding benefits of ultra-effective and anti-interference capability. However, most of the research has paid more attention to the performance of SACs, while the in-depth understanding of catalytic regulation by molecular interaction is relatively deficient. This critical review delves into deciphering the catalytic mechanism through a micro-level, which makes it more convenient to interpret apparent catalytic phenomena. It first summarizes basic theories of quantum chemistry, which provide mechanism interpretation and prediction for molecular-oxidation systems. Additionally, corresponding oxidation pathways of common oxidants are underscored. Following the oxidants, state-of-the-art regulation methods are discussed with special attention to involved molecular interactions and pollutants. Particularly, the preliminary insights into the "oxidant-catalyst-pollutants" internal relationships are provided to help construct the SAC-AOP system from a molecular standpoint. Meanwhile, some cutting-edge laboratory devices and pilot-scale engineering are presented to illustrate the ultimate purpose of scientific molecular exploration. Eventually, relative challenges of SACs-AOPs upon the design of catalytic systems and investigation methods are provided. This review aims to promote the large-scale potential of SACs-based AOPs in practical water treatment by emphasizing the pivotal role of micro-insights.
Collapse
Affiliation(s)
- Huayue Kang
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Yaoning Chen
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Min Cheng
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Huiqin Guo
- Key Laboratory of Jiangxi Province for Persistent Pollutants Control and Resources Recycle, Nanchang Hangkong University, Nanchang, 330063, China
- National-Local Joint Engineering Research Center of Heavy Metals Pollutants Control and Resource Utilization, Nanchang Hangkong University, Nanchang, 330063, China
| | - Gaoxia Zhang
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Qingkai Shi
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Wencheng Zhou
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Chen Zhao
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Bin Zou
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Xinyue Lv
- Key Laboratory of Jiangxi Province for Persistent Pollutants Control and Resources Recycle, Nanchang Hangkong University, Nanchang, 330063, China
- National-Local Joint Engineering Research Center of Heavy Metals Pollutants Control and Resource Utilization, Nanchang Hangkong University, Nanchang, 330063, China
| | - Ziyue Yuan
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| | - Guangming Zeng
- College of Environmental Science and Engineering, Hunan University and Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, China
| |
Collapse
|
4
|
Kılıç Ö, Tecer D, Kaya MN, Çınar M, Yılmaz S. Can the Early Warning Score (ANDC) Predict Tocilizumab Efficacy in Patients with COVID-19 Cytokine Storm? Eur J Rheumatol 2025; 12:1-6. [PMID: 40377410 PMCID: PMC12060182 DOI: 10.5152/eurjrheum.2025.24048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/22/2024] [Indexed: 05/18/2025] Open
Abstract
Background The aim of this study is to assess the effectiveness of the Early Warning Score [ANDC (age (A), neutrophil-to-lymphocyte ratio (NLR (N)), D-dimer (D), and CRP (C)] in predicting the treatment response in patients receiving tocilizumab for Coronavirus Disease 2019 (COVID-19)-related cytokine storm. Methods A retrospective review of medical records was conducted for patients treated with tocili- zumab for a cytokine storm related to COVID-19 between April 1, 2020, and April 1, 2021. Patient demographics, clinical characteristics, and laboratory parameters within 24 hours before tocilizumab were recorded. 1.14 × (age - 20) (years) + 1.63 × NLR + 5.00 × D-dimer (mg/L) + 0.14 × C-reactive protein (CRP) (mg/L) was used as the formula for the ANDC score. The study population was divided into 2 groups: those who died within 28 days of receiving tocilizumab and those who recovered. A comparative analysis was conducted. Results Within 28 days of tocilizumab treatment, 59 (35.32%) patients died. In comparison with living patients, deceased patients exhibited considerably higher levels of interleukin (IL)-6, lactate dehydro- genase (LDH), ANDC score, and CRP (P < .05). Lactate dehydrogenase was an independent predictor of response to tocilizumab treatment (P < .001) in a multivariate logistic regression analysis. In patients who did not receive steroid therapy before tocilizumab treatment, the ANDC score had the highest area under the curve (AUC). The optimal cut-off value was determined to be 92.56, with a sensitivity of 91.67% and a specificity of 60.61% (P < .001). In patients receiving steroids before tocilizumab, LDH had the highest AUC. The optimal cut-off value was 484.5 U/L (P < .001). Conclusion Lactate dehydrogenase was identified as an independent predictor of response to tocili- zumab treatment. The ANDC score showed the highest AUC value in steroid-naïve patients before tocilizumab, whereas LDH showed the highest AUC value in patients receiving steroids before tocili- zumab. Both the ANDC score and LDH levels show potential as valuable tools to guide treatment decisions.
Collapse
Affiliation(s)
- Özlem Kılıç
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Duygu Tecer
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Mehmet Nur Kaya
- Department of Rheumatology, Hakkari State Hospital, Hakkari, Türkiye
| | - Muhammet Çınar
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Sedat Yılmaz
- Department of Rheumatology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| |
Collapse
|
5
|
Tomos I, Grigoropoulos I, Kosti C, Chrysikos S, Digalaki A, Thomas K, Hillas G, Kazakou P, Antoniadou A, Kavatha D, Dimakou K. Comparison of effectiveness and safety between baricitinib and tocilizumab in severe COVID-19: a retrospective study. Expert Rev Respir Med 2025; 19:389-397. [PMID: 40017107 DOI: 10.1080/17476348.2025.2473486] [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: 12/02/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Immunomodulators tocilizumab and baricitinib have been used for the treatment of severe COVID-19, however, there are only few published studies comparing their efficacy. RESEARCH DESIGN AND METHODS All consecutive non-ICU hospitalized severe COVID-19 patients who received baricitinib or tocilizumab, were included retrospectively. Primary outcomes were mortality or intubation on day 14, time to oxygen therapy weaning and duration of hospitalization. Safety was measured as treatment-related adverse events. RESULTS 321 hospitalized patients with severe COVID-19 were included (mean age 62.4 years ± 14.7); 241 (75.1%) received baricitinib (mean age 64.2 years ± 15.2) and 80 (24.9%) tocilizumab (mean age 57.3 ± 11.7). Patients who received baricitinib presented significantly lower risk of mortality or intubation on day 14, compared to the tocilizumab group after adjusting for age, sex, vaccination, Charlson comorbidity index, body mass index, remdesivir administration and WHO ordinal scale at enrollment (OR: 0.42, 95% CI: 0.20-0.86). In the augmented inverse-probability weighting regression, the protective role of baricitinib remained statistically significant (OR: 0.76, 95% CI: 0.66-0.88). No difference in secondary bacterial infections was detected, but tocilizumab was associated with significant higher rate of liver injury (Odds Ratio, 95%CI, p < 0.001). CONCLUSIONS Our study suggests survival and safety are significantly better for baricitinib compared to tocilizumab in severe COVID-19. Clinical randomized trials are needed for confirmation.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Chrysavgi Kosti
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Serafeim Chrysikos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Antonia Digalaki
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Konstantinos Thomas
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Georgios Hillas
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Pinelopi Kazakou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Dimitra Kavatha
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Katerina Dimakou
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| |
Collapse
|
6
|
Yang X, Wang C, Xue Y, Zhang Y, Zheng M, Sun Q, Long S, Wang D, Yan J, Liao X, Zhang T, Cao L, Chen Y, Ju W, Zhang J, Gao M, Zhao Y, Luu LDW, Pan J, Wang Y, Wang G. Clinical outcomes of patients with coronavirus disease 2019 and active tuberculosis co-infection in Beijing China: A retrospective single-center descriptive study. INFECTIOUS MEDICINE 2025; 4:100169. [PMID: 40129441 PMCID: PMC11930588 DOI: 10.1016/j.imj.2025.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 03/26/2025]
Abstract
Background Coronavirus disease 2019 (COVID-19) and tuberculosis (TB) co-infection (COVID-19-TB) has the potential to exacerbate lung damage; however, information about the clinical features of COVID-19-TB is limited. This study aims to clarify the clinical characteristics and outcomes of patients with COVID-19-TB. Methods In this single-center retrospective study, the clinical features and outcomes of patients with COVID-19 with active TB who were admitted to Beijing Chest Hospital, Beijing, China, from 1 December 2022 to 18 January 2023 were collected. The severity of COVID-19 and TB was graded according to guidelines from the World Health Organization. The relationships of demographic and clinical variables with intensive care unit (ICU) admission were evaluated using univariable and multivariable logistic regression models. Results Overall, 102 patients with COVID-19-TB were enrolled. The mean age was 54.5 years (range 36.5-70 years). The most common clinical manifestations were cough (68.63%), sputum production (53.92%), fever (51.96%), and ground-glass opacities (35.29%). Complications included acute respiratory distress syndrome (11.76%), sepsis (9.8%), and respiratory failure (7.84%). Patients with COVID-19-TB had high concentrations of various proinflammatory cytokines, including interferon-γ, interleukin-1β, interferon-γ-inducible protein 10 kD, and monocyte chemoattractant protein-1. Sixteen of the 102 patients with COVID-19-TB (15.69%) were admitted to the ICU, and 10 (9.80%) died during hospitalization. The significant risk factors for ICU admission were respiratory failure, pulmonary fungal infection, and ventilation and oxygen therapy. Conclusions The mortality rate of COVID-19-TB was 9.80%. Several demographic and clinical characteristics were associated with adverse outcomes, indicating the importance of early recognition and treatment.
Collapse
Affiliation(s)
- Xinting Yang
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Chaohong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Yu Xue
- Department of Emergency, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Yun Zhang
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Maike Zheng
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Qing Sun
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Sibo Long
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Da Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Jun Yan
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Xinlei Liao
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Tiantian Zhang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Lei Cao
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Yan Chen
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Wenfu Ju
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Jing Zhang
- Department of Emergency, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Mengqiu Gao
- Tuberculosis Department, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Yan Zhao
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Laurence Don Wai Luu
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia
| | - Junhua Pan
- Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Yi Wang
- Experimental Research Center, Capital Institute of Pediatrics, Beijing 100020, China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| |
Collapse
|
7
|
Feng Z, Wang L, Yang J, Li T, Liao X, Kang Y, Xiao F, Zhang W. Sepsis: the evolution of molecular pathogenesis concepts and clinical management. MedComm (Beijing) 2025; 6:e70109. [PMID: 39991626 PMCID: PMC11847631 DOI: 10.1002/mco2.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/29/2024] [Accepted: 01/07/2025] [Indexed: 02/25/2025] Open
Abstract
The mortality rate of sepsis is approximately 22.5%, accounting for 19.7% of the total global mortality. Since Lewis Thomas proposed in 1972 that "it is our response that makes the disease (sepsis)" rather than the invading microorganisms, numerous drugs have been developed to suppress the "overwhelming" inflammatory response, but none of them has achieved the desired effect. Continued failure has led investigators to question whether deaths in septic patients are indeed caused by uncontrolled inflammation. Here, we review the history of clinical trials based on evolving concepts of sepsis pathogenesis over the past half century, summarize the factors that led to the failure of these historical drugs and the prerequisites for the success of future drugs, and propose the basic principles of preclinical research to ensure successful clinical translation. The strategy of targeting inflammatory factors are like attempting to eliminate invaders by suppressing the host's armed forces, which is logically untenable. Sepsis may not be that complex; rather, sepsis may be the result of a failure to fight microbes when the force of an invading pathogen overwhelms our defenses. Thus, strengthening the body's defense forces instead of suppressing them may be the correct strategy to overcome sepsis.
Collapse
Affiliation(s)
- Zhongxue Feng
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Lijun Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jing Yang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tingting Li
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xuelian Liao
- Department of Critical Care MedicineWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yan Kang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and ObstetricsWest China Second University Hospital, Sichuan UniversityChengduSichuanChina
| | - Wei Zhang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
8
|
Casetti R, Sacchi A, Mazzotta V, Cristofanelli F, Grassi G, Gili S, Cimini E, Notari S, Bordoni V, Mastrorosa I, Giancola ML, Vergori A, Tempestilli M, Vita S, Mariotti D, Rosati S, Lalle E, Meschi S, Colavita F, Garbuglia AR, Girardi E, Nicastri E, Antinori A, Agrati C. Innate and SARS-CoV-2 specific adaptive immune response kinetic in neutralizing monoclonal antibody successfully treated COVID-19 patients. Int Immunopharmacol 2025; 148:113934. [PMID: 39832460 DOI: 10.1016/j.intimp.2024.113934] [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: 07/22/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
The impact of anti-Spike monoclonal antibody (mAbs) treatment on the immune response of COVID19-patients is poorly explored. In particular, a comparison of the immunological influence of different therapeutic regimens has not yet been performed. Aim of the study was to compare the kinetic of innate and adaptive immune response as well as the SARS-CoV-2 specific humoral and T cell response in two groups of SARS-CoV-2-infected patients treated with two different mAbs regimens: Bamlanivimab/Etesevimab (BAM/ETE) or Casirivimab/Imdevimab (CAS/IMD). SARS-CoV-2-infected patients (n = 39) with mild/moderate disease were enrolled before (T0) and after 7 days (T7) and 30 day (T30) from mAbs infusion. Patients were divided in two groups on the basis of the mAb regimen: BAM/ETE (n = 15) and CAS/IMD (n = 24). The phenotype/function of immune cell subsets was evaluated by flow-cytometry and by ELISA. The Spike-specific T cell response (IFN-γ) and anti-Nucleocapside IgG were evaluated by chemiluminescence assay. SARS CoV-2 RNA in nasal swabs was evaluated by RT-PCR. Eleven out of the thirty-nine enrolled patients tested negative at T7, among which nine (81.8 %) had been treated with CAS/IMD regimen. A comparable increase in CD4 and CD8 T cells was observed in both treatment groups. Moreover, a reduction of CD38 expression on T (CD4, CD8 and Vδ2) and on NK cells was observed in both groups, as well as a reduction overtime of the perforin expression in T (CD8, Vδ2) and in NK cells reaching significance only in CAS/IMD-treated patients. The SARS-CoV-2-specific T cells response increased at T7 in BAM/ETE-treated patients and at T30 in CAS/IND group. Of note, at T30 SARS-CoV2-specific T cells was higher in CAS/IMD than in BAM/ETE group. Furthermore, the titre of anti-N IgG increased overtime in both groups with a faster kinetic in CAS/IMD group. The spontaneous production of inflammatory cytokines by monocytes and neutrophils was similar the two mAb regimens, as well as the level of plasmatic IL-6. Finally, patients were also analysed according to sex. The male group showed a higher frequency of activated CD4 T cells, NKG2A-expressing CD8 T cells and perforin-expressing Vδ2 T cells compared to female group. Moreover, a higher specific T cell response at T30 was observed in the male compared to female group. In conclusion, these results show similar effects of both mAb regimens in restoring T and NK cell homeostasis and in reducing inflammation. In contrast, CAS/IMD allows a better humoral and cellular SARS-CoV2 specific immunization.
Collapse
Affiliation(s)
- Rita Casetti
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Alessandra Sacchi
- Molecular Virology and Antimicrobic Immunity Laboratory, Department of Science, University of Rome Three, 00146 Rome, Italy.
| | | | - Flavia Cristofanelli
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Germana Grassi
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Simona Gili
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Eleonora Cimini
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Stefania Notari
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Veronica Bordoni
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, Rome 00146 Italy.
| | | | | | | | - Massimo Tempestilli
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Serena Vita
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Davide Mariotti
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Silvia Rosati
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Eleonora Lalle
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Silvia Meschi
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | | | | | - Enrico Girardi
- Scientific Directorate, INMI L. Spallanzani, 00149 Rome, Italy.
| | | | - Andrea Antinori
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Chiara Agrati
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, Rome 00146 Italy.
| |
Collapse
|
9
|
Peng Z, Zhou G. Progress on diagnosis and treatment of multisystem inflammatory syndrome in children. Front Immunol 2025; 16:1551122. [PMID: 40046058 PMCID: PMC11879827 DOI: 10.3389/fimmu.2025.1551122] [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: 12/24/2024] [Accepted: 01/24/2025] [Indexed: 05/13/2025] Open
Abstract
Since the emergence of COVID-19 in December 2019, the novel SARS-CoV-2 virus has primarily affected adults, with children representing a smaller proportion of cases. However, the escalation of the pandemic has led to a notable increase in pediatric cases of Multisystem Inflammatory Syndrome in Children (MIS-C). The pathogenesis of MIS-C is largely attributed to immune-mediated mechanisms, such as cytokine storms and endothelial damage, following SARS-CoV-2 infection. In this review, we comprehensively describe MIS-C, including its definitions as proposed by the CDC, WHO, and RCPCH, which emphasize persistent fever, excessive inflammatory responses, and multi-organ involvement. Additionally, we summarize current treatment approaches, prioritizing immunotherapy with intravenous immunoglobulin and corticosteroids, along with anticoagulation therapy, and monoclonal antibodies in severe cases.
Collapse
Affiliation(s)
| | - Gang Zhou
- Department of Pediatric Respiratory Diseases, Chongqing University Three Gorges Hospital, Chongqing, China
| |
Collapse
|
10
|
Ishikawa T, Matsumoto K, Hamada T, Koze H, Baba M, Okamoto M, Sudoh M. In Silico Discovery of SARS-CoV-2 Main Protease Inhibitors Using Docking, Molecular Dynamics, and Fragment Molecular Orbital Calculations. J Phys Chem B 2025; 129:1740-1749. [PMID: 39886917 DOI: 10.1021/acs.jpcb.4c07920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The 3C-like protease of severe acute respiratory syndrome coronavirus 2, known as the main protease (Mpro), is an attractive drug target for the treatment of coronavirus disease 2019. This study reports the discovery of novel Mpro inhibitors using several in silico techniques, including docking, molecular dynamics (MD), and fragment molecular orbital (FMO) calculations. We performed docking calculations on 5950 compounds with bioactivity, and 12 compounds were selected. An enzymatic assay was conducted, revealing that BP-1-102 exhibits significant Mpro inhibitory activity with an IC50 of 11.1 μM. The identification of seed compounds from the experiments on a few compounds demonstrates the effectiveness of our docking calculations. Furthermore, the detailed analyses using MD and FMO calculations suggested an interaction mechanism in which the hydroxyl group of BP-1-102 forms a hydrogen bond with E166 of Mpro. The Mpro inhibitory activity of SH-4-54, a derivative without the aforementioned hydroxyl group, was investigated and observed to be significantly reduced, with an IC50 of 81.5 μM. This result strongly supports the suggested interaction mechanism.
Collapse
Affiliation(s)
- Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Kenji Matsumoto
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Toshiyuki Hamada
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan
| | - Hinako Koze
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan
| | - Masanori Baba
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-8580, Japan
| | - Mika Okamoto
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-8580, Japan
| | - Masayuki Sudoh
- Department of Translational Research, Joint Research Center for Human Retrovirus Infection, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
11
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [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: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
12
|
Lokau J, Garbers Y, Vicente MM, Dittrich A, Meltendorf S, Lingel H, Münster-Kühnel AK, Brunner-Weinzierl M, Garbers C. Long-term increase in soluble interleukin-6 receptor levels in convalescents after mild COVID-19 infection. Front Immunol 2025; 15:1488745. [PMID: 39835136 PMCID: PMC11743636 DOI: 10.3389/fimmu.2024.1488745] [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: 12/12/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Serum levels of interleukin-6 (IL-6) are increased in COVID-19 patients. IL-6 is an effective therapeutic target in inflammatory diseases and tocilizumab, a monoclonal antibody that blocks signaling via the IL-6 receptor (IL-6R), is used to treat patients with severe COVID-19. However, the IL-6R exists in membrane-bound and soluble forms (sIL-6R), and the sIL-6R in combination with soluble glycoprotein 130 (sgp130) forms an IL-6-neutralizing buffer system capable of neutralizing small amounts of IL-6. Methods In this study, we analyzed serum levels of IL-6, sIL-6R and sgp130 in the serum of COVID-19 convalescent individuals with a history of mild COVID-19 disease and in acute severely ill COVID-19 patients compared to uninfected control subjects. Furthermore, we used single cell RNA sequencing data in order to determine which immune cell types are sources and targets of the individual cytokines and whether their expression is altered in severe COVID-19 patients. Results We find that sIL-6R levels are not only increased in acute severely ill patients, but also in convalescents after a mild COVID-19 infection. We show that this increase in sIL-6R results in an enhanced capacity of the sIL-6R/sgp130 buffer system, but that significantly enhanced free IL-6 is still present due to an overload of the buffer. Further, we identify IL-6 serum levels, age and the number of known pre-existing medical conditions as crucial determinants of disease outcome for the patients. We also show that IL-11 has no major systemic role in COVID-19 patients and that sCD25 is only increased in acute severely ill COVID-19 patients, but not in mild convalescent individuals. Discussion In conclusion, our study shows long-lasting alterations of the IL-6 system after COVID-19 disease, which might be relevant when applying anti-IL-6 or anti-IL-6R therapy.
Collapse
Affiliation(s)
- Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Yvonne Garbers
- Faculty of Management, Culture and Technology (Lingen campus), Osnabrück University of Applied Sciences, Lingen, Germany
| | - Manuel M. Vicente
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Anna Dittrich
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Stefan Meltendorf
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Holger Lingel
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Monika Brunner-Weinzierl
- Department of Experimental Pediatrics, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| |
Collapse
|
13
|
Liu JH, Wang Y, Dai SJ, Zhang DW, Yue XD. C 17-Labdane diterpenoid alkaloids bearing a rare skeleton with anti-inflammatory and anti-oxidant activities from Forsythia suspensa. Fitoterapia 2025; 180:106345. [PMID: 39667676 DOI: 10.1016/j.fitote.2024.106345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024]
Abstract
Two undescribed C17-Labdane diterpenoid alkaloids, named forsylinfenines A and B (1-2), attributable to a rare 4,4,10,13-tetramethyl-1(2),3(4),5(10),6(7)-octahydrobenzo[f]quinolin skeleton, along with three known β-carboline-type alkaloids (3-5), were isolated. The chemical structures including absolute configurations of two undescribed compounds were established by means of integrated spectroscopic techniques and electronic circular dichroism (ECD) calculations. In addition, a plausible biosynthetic pathway for the formation of compounds 1 and 2 was proposed. In vitro, five alkaloids (1-5), especially two undescribed alkaloids with rare skeleton (1-2), exhibited significant anti-inflammatory activities due to inhibiting the release of TNF-α, IL-6, and IL-1β, as well as effective anti-oxidant activities owing to preventing the production of ROS in the LPS-induced RAW264.7 cells.
Collapse
Affiliation(s)
- Jia-Huan Liu
- School of Pharmaceutical Science, Yantai University, Yantai 264005, China
| | - Yue Wang
- School of Pharmaceutical Science, Yantai University, Yantai 264005, China
| | - Sheng-Jun Dai
- School of Pharmaceutical Science, Yantai University, Yantai 264005, China.
| | - De-Wu Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Xi-Dian Yue
- College of Life Sciences, Yantai University, Yantai 264005, China.
| |
Collapse
|
14
|
Mori N, Nanki T, Hirakawa A, Yamato M, Kaneko Y, Shiokawa R, Ozaki R, Kawabata N, Ohmagari N. Tocilizumab in combination with standard of care in patients with severe COVID-19 pneumonia: Efficacy and safety from a phase 3 clinical trial in Japan. J Infect Chemother 2025; 31:102524. [PMID: 39326494 DOI: 10.1016/j.jiac.2024.09.013] [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: 12/15/2023] [Revised: 08/21/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is characterized by high interleukin-6 levels. Clinical data supporting tocilizumab, a monoclonal antibody that targets interleukin-6 receptor-alpha, for treating Japanese patients with severe COVID-19 pneumonia are needed. METHODS This single-arm phase 3 study investigated tocilizumab (8 mg/kg) plus standard of care (SOC) in Japanese patients hospitalized with severe COVID-19 pneumonia. Clinical status was assessed using a 7-category ordinal scale on day 28 (primary endpoint) and day 14 (secondary endpoint). Other secondary endpoints were time to improvement (≥2 category improvement) and time to hospital discharge. Safety was assessed as the incidence of adverse events (AEs). RESULTS Among 48 patients enrolled, 44 (91.7 %) scored ≥3 on the 7-category ordinal scale at baseline. At day 28, 35 patients (72.9 %) scored 1 and 5 (10.4 %) scored 7 on the 7-category ordinal scale; 36 (75.0 %, 95 % confidence interval [CI]: 60.40 %-86.36 %) and 39 (81.3 %, 95 % CI: 67.37 %-91.05 %) patients achieved ≥2- and ≥1-category improvement, respectively; 6 patients (12.5 %, 95 % CI: 4.73 %-25.25 %) demonstrated ≥1-category worsening. At day 14, 25 (52.1 %, 95 % CI: 37.19 %-66.71 %) and 33 patients (68.8 %, 95 % CI: 53.75 %-81.34 %) achieved ≥2- and ≥1-category improvement, respectively; 5 patients (10.4 %, 95 % CI: 3.47 %-22.66 %) demonstrated ≥1-category worsening. Median times (95 % CI) to improvement and hospital discharge were 11 (9-15) and 15 (11-18) days, respectively. Forty patients (83.3 %) experienced AEs; the incidence of ≥grade 3 AEs was 25 %. CONCLUSION Tocilizumab plus SOC may provide improved clinical status in Japanese patients with severe COVID-19 pneumonia; no new safety signals were identified.
Collapse
Affiliation(s)
- Nobuyoshi Mori
- Division of Infectious Diseases, Department of Medicine, St. Luke's International Hospital, Tokyo, Japan.
| | - Toshihiro Nanki
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaya Yamato
- Division of General Internal Medicine and Infectious Diseases, Rinku General Medical Center, Osaka, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | - Ryoto Ozaki
- Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Liu WL, Kampouri E, Bui JK, Sekhon MK, Tercero A, Finlay D, Asghedom LH, Romasanta GR, Rice NT, Ranjbaran F, Stoltzman C, Cook J, Blake J, Delaney CS, Hill JA. Off-the-shelf allogeneic natural killer cells for the treatment of COVID-19. Mol Ther Methods Clin Dev 2024; 32:101361. [PMID: 39624798 PMCID: PMC11609367 DOI: 10.1016/j.omtm.2024.101361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/24/2024] [Indexed: 02/14/2025]
Abstract
Low levels and function of natural killer (NK) cells are associated with increased coronavirus disease 2019 (COVID-19) severity. NK cell immunotherapy may improve immune function to reduce infection severity. We conducted a first-in-human, open-label, phase 1, dose-escalating (100 × 106, 300 × 106, or 900 × 106 cells) study of a single dose of DVX201, a cord-blood-derived allogeneic NK cell therapy, in hospitalized patients with COVID-19. Participants were followed for 28 days. The maximum allowed steroid dose for eligibility was up to 0.5 mg/kg prednisone (or equivalent) daily. We enrolled nine participants, 3 per dose level. Eight participants had ≥1 comorbidity associated with increased COVID-19 severity, three of whom had a hematologic malignancy. Infusions were well tolerated, with no treatment-related adverse events. There was no evidence of inflammatory complications related to infusions. Peripheral blood NK cells generally increased after infusion, peaking by day 7. The median time from infusion to discharge was 2 days (range: 1-13). Two patients (both with acute lymphoblastic leukemia) were readmitted with recurrent COVID-19. This trial demonstrates the safety of allogeneic NK cell immunotherapy as a potential antiviral. Larger controlled trials are needed to establish efficacy.
Collapse
Affiliation(s)
| | | | - John K. Bui
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | | | | | - Dan Finlay
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | | | | | | | - Jody Cook
- Deverra Therapeutics, Seattle, WA, USA
| | - Joe Blake
- Deverra Therapeutics, Seattle, WA, USA
| | | | - Joshua A. Hill
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
16
|
Maiti AK. MDA5 Is a Major Determinant of Developing Symptoms in Critically Ill COVID-19 Patients. Clin Rev Allergy Immunol 2024; 67:58-72. [PMID: 39460899 DOI: 10.1007/s12016-024-09008-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] [Accepted: 10/12/2024] [Indexed: 10/28/2024]
Abstract
Apart from the skin and mucosal immune barrier, the first line of defense of the human immune system includes MDA5 (ifih1 gene) which acts as a cellular sensor protein for certain viruses including SARS-CoV-2. Upon binding with viral RNA, MDA5 activates cell-intrinsic innate immunity, humoral responses, and MAVS (mitochondrial antiviral signaling). MAVS signaling induces type I and III interferon (IFN) expressions that further induce ISGs (interferon stimulatory genes) expressions to initiate human cell-mediated immune responses and attenuate viral replication. SARS-CoV-2 counteracts by producing NSP1, NSP2, NSP3, NSP5, NSP7, NSP12, ORF3A, ORF9, N, and M protein and directs anti-MDA5 antibody production presumably to antagonize IFN signaling. Furthermore, COVID-19 resembles several diseases that carry anti-MDA5 antibodies and the current COVID-19 vaccines induced anti-MDA5 phenotypes in healthy individuals. GWAS (genome-wide association studies) identified several polymorphisms (SNPs) in the ifih1-ifn pathway genes including rs1990760 in ifih1 that are strongly associated with COVID-19, and the associated risk allele is correlated with reduced IFN production. The genetic association of SNPs in ifih1 and ifih1-ifn pathway genes reinforces the molecular findings of the critical roles of MDA5 in sensing SARS-CoV-2 and subsequently the IFN responses to inhibit viral replication and host immune evasion. Thus, MDA5 or its pathway genes could be targeted for therapeutic development of COVID-19.
Collapse
Affiliation(s)
- Amit K Maiti
- Mydnavar, Department of Genetics and Genomics, 28475 Greenfield Rd, Southfield, MI, USA.
| |
Collapse
|
17
|
Liang P, Li Y, Meng L, Li Y, Mai H, Li T, Ma J, Ma J, Wang J, Zhuan B, Zhou W. Prognostic significance of serum interleukin-6 in severe/critical COVID-19 patients treated with tocilizumab: a detailed observational study analysis. Sci Rep 2024; 14:29634. [PMID: 39609511 PMCID: PMC11605089 DOI: 10.1038/s41598-024-81028-3] [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: 06/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
Baseline IL-6 levels have been found to be non-predictive of subsequent outcomes following tocilizumab treatment, highlighting the need for more reliable predictive markers. To address this, a retrospective analysis was conducted on the clinical profiles, diagnostic tests, and follow-up prognoses of 60 patients with severe or critical COVID-19, all of whom were identified as experiencing a cytokine storm and subsequently received tocilizumab treatment. Among the patients, the overall survival rate during follow-up was 80%, with further analysis revealing that advanced age was an independent risk factor for adverse outcomes. Following tocilizumab administration, a statistically significant increase in IL-6 and D-dimer levels was observed, while markers such as C-reactive protein (CRP), procalcitonin (PCT), and fibrinogen demonstrated reductions compared to pre-treatment values. Specifically, IL-6 levels initially surged briefly after tocilizumab intervention before gradually diminishing. To assess the prognostic utility of IL-6, the Receiver Operating Characteristic (ROC) curve was employed, which yielded an area under the curve (AUC) of 0.812, indicating strong predictive capability, with a sensitivity of 100% and a specificity of 53.49%. The optimal cut-off value for IL-6 was identified at 147.79 pg/mL. In conclusion, IL-6 levels tend to rise transiently following tocilizumab therapy, before gradually declining. These post-treatment IL-6 measurements may serve as a valuable biomarker for assessing prognosis in patients undergoing this treatment.
Collapse
Affiliation(s)
- Panpan Liang
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yan Li
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li'e Meng
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yuting Li
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Hailing Mai
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Tao Li
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jiarui Ma
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Junhui Ma
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jing Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Bing Zhuan
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| | - Wei Zhou
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
18
|
Rahmani D, Jafari A, Kesharwani P, Sahebkar A. Molecular targets in SARS-CoV-2 infection: An update on repurposed drug candidates. Pathol Res Pract 2024; 263:155589. [PMID: 39276508 DOI: 10.1016/j.prp.2024.155589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
The 2019 widespread contagion of the human coronavirus novel type (SARS-CoV-2) led to a pandemic declaration by the World Health Organization. A daily increase in patient numbers has formed an urgent necessity to find suitable targets and treatment options for the novel coronavirus (COVID-19). Despite scientists' struggles to discover quick treatment solutions, few effective specific drugs are approved to control SARS-CoV-2 infections thoroughly. Drug repositioning or Drug repurposing and target-based approaches are promising strategies for facilitating the drug discovery process. Here, we review current in silico, in vitro, in vivo, and clinical updates regarding proposed drugs for prospective treatment options for COVID-19. Drug targets that can direct pharmaceutical sciences efforts to discover new drugs against SARS-CoV-2 are divided into two categories: Virus-based targets, for example, Spike glycoprotein and Nucleocapsid Protein, and host-based targets, for instance, inflammatory cytokines and cell receptors through which the virus infects the cell. A broad spectrum of drugs has been found to show anti-SARS-CoV-2 potential, including antiviral drugs and monoclonal antibodies, statins, anti-inflammatory agents, and herbal products.
Collapse
Affiliation(s)
- Dibachehr Rahmani
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Michot JM, Dozio V, Rohmer J, Pommeret F, Roumier M, Yu H, Sklodowki K, Danlos FX, Ouali K, Kishazi E, Naigeon M, Griscelli F, Gachot B, Groh M, Bacciarello G, Stoclin A, Willekens C, Sakkal M, Bayle A, Zitvogel L, Silvin A, Soria JC, Barlesi F, Beeler K, André F, Vasse M, Chaput N, Ackermann F, Escher C, Marabelle A. Circulating Proteins Associated with Anti-IL6 Receptor Therapeutic Resistance in the Sera of Patients with Severe COVID-19. J Proteome Res 2024; 23:5001-5015. [PMID: 39352225 DOI: 10.1021/acs.jproteome.2c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Circulating proteomes provide a snapshot of the physiological state of a human organism responding to pathogenic challenges and drug interventions. The outcomes of patients with COVID-19 and acute respiratory distress syndrome triggered by the SARS-CoV2 virus remain uncertain. Tocilizumab is an anti-interleukin-6 treatment that exerts encouraging clinical activity by controlling the cytokine storm and improving respiratory distress in patients with COVID-19. We investigate the biological determinants of therapeutic outcomes after tocilizumab treatment. Overall, 28 patients hospitalized due to severe COVID-19 who were treated with tocilizumab intravenously were included in this study. Sera were collected before and after tocilizumab, and the patient's outcome was evaluated until day 30 post-tocilizumab infusion for favorable therapeutic response to tocilizumab and mortality. Hyperreaction monitoring measurements by liquid chromatography-mass spectrometry-based proteomic analysis with data-independent acquisition quantified 510 proteins and 7019 peptides in the serum of patients. Alterations in the serum proteome reflect COVID-19 outcomes in patients treated with tocilizumab. Our results suggested that circulating proteins associated with the most significant prognostic impact belonged to the complement system, platelet degranulation, acute-phase proteins, and the Fc-epsilon receptor signaling pathway. Among these, upregulation of the complement system by activation of the classical pathway was associated with poor response to tocilizumab, and upregulation of Fc-epsilon receptor signaling was associated with lower mortality.
Collapse
Affiliation(s)
- Jean-Marie Michot
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Vito Dozio
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Julien Rohmer
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Fanny Pommeret
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Haochen Yu
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | | | - François-Xavier Danlos
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Kaissa Ouali
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Edina Kishazi
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Marie Naigeon
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
| | - Franck Griscelli
- Département de biologie et pathologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Bertrand Gachot
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Giulia Bacciarello
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Annabelle Stoclin
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Christophe Willekens
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Madona Sakkal
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Arnaud Bayle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Jean-Charles Soria
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Fabrice Barlesi
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Fabrice André
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Unité INSERM U981, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Marc Vasse
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
- Service de biologie clinique, Hôpital Foch, Suresnes 92150, France
- Unité INSERM U1176, Le Kremlin-Bicêtre, Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Nathalie Chaput
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Felix Ackermann
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | | | - Aurélien Marabelle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Centre d'investigation clinique - biothérapie, INSERM CICBT1428, Villejuif 94800, France
| |
Collapse
|
20
|
Pan Y, Lin W, Huang Y, Pan J, Dong Y. Decoding the mechanism of Qingjie formula in the prevention of COVID-19 based on network pharmacology and molecular docking. Heliyon 2024; 10:e39167. [PMID: 39640673 PMCID: PMC11620151 DOI: 10.1016/j.heliyon.2024.e39167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Traditional Chinese medicine (TCM) has played a positive role in preventing and controlling the coronavirus disease 2019 (COVID-19) epidemic. Qingjie formula (QJF) developed to prevent COVID-19 is widely used in Wenzhou, Zhejiang province, China. However, the biological active ingredients of QJF and their specific mechanisms for preventing COVID-19 remain unclear. The study focused on exploring the pharmacological mechanism of QJF for the prevention of COVID-19 based on network pharmacology and molecular docking. The active ingredients of QJF were screened by TCMSP database. Databases such as Genecards and Swiss Target Prediction predicted potential targets of QJF against COVID-19. The "drug-active ingredient-potential target" network was constructed by Cytoscape software. We used STRING database to construct the protein-protein interaction (PPI) network. Enrichment of biological functions and signaling pathways were analyzed by using the DAVID database and R language. Then AutoDock Vina and Python software were used for molecular docking of hub targets and active ingredients. 147 active ingredients interacted with 316 potential targets of COVID-19. A PPI network consisting of 30 hub genes was constructed, and the top 10 hub genes were ALB, AKT1, TP53, TNF, IL6, VEGFA, IL1B, CASP3, JUN and STAT3. The results of GO analysis showed that these targets were mainly enriched in cell responses to oxidative stress, chemical stress, and other functions. KEGG analysis revealed that viral protein interactions with cytokines (e.g., human cytomegalovirus infection), endocrine resistance pathways (e.g., AGE-RAGE signaling pathway), PI3K-Akt signaling pathway, and lipid and atherosclerosis signaling pathway were the major signaling pathways. Moreover, the core active ingredients of QJF had good binding affinity with hub genes by molecular docking. QJF plays an important role in the prevention of COVID-19 by regulating host immune inflammatory response and oxidative stress response, inhibiting virus, improving immune function, regulating the hypoxia-cytokine storm, and inhibiting cell migration.
Collapse
Affiliation(s)
- Yu Pan
- Department of Pharmacy, Zhejiang Chinese Medical University Affiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, 325000, China
| | - Wanchun Lin
- Department of Pharmacy, Zhejiang Chinese Medical University Affiliated Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang, 325000, China
| | - Yueyue Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yihua Dong
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
21
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [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: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
22
|
Sarikaya ZT, Gucyetmez B, Tuzuner F, Dincer O, Sahan C, Dogan L, Yildirim SA, Zengin R, Kocagoz AS, Telci L, Akinci IO. The usage of immunosuppressant agents and secondary infections in patients with COVID-19 in the intensive care unit: a retrospective study. Sci Rep 2024; 14:20991. [PMID: 39251824 PMCID: PMC11385116 DOI: 10.1038/s41598-024-71912-3] [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: 05/24/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Although COVID-19 infection is an immunosuppressant disease, many immunosuppressant agents, such as pulse methylprednisolone (PMP), dexamethasone (DXM), and tocilizumab (TCZ), were used during the pandemic. Secondary infections in patients with COVID-19 have been reported recently. This study investigated these agents' effects on secondary infections and outcomes in patients with COVID-19 in intensive care units (ICUs). This study was designed retrospectively, and all data were collected from the tertiary intensive care units of six hospitals between March 2020 and October 2021. All patients were divided into three groups: Group I [GI, PMP (-), DXM (-) and TCZ (-)], Group II [GII, PMP (+), DXM (+)], and Group III [GIII, PMP (+), DXM (+), TCZ (+)]. Demographic data, PaO/FiO2 ratio, laboratory parameters, culture results, and outcomes were recorded. To compare GI-GII and GI-GIII, propensity score matching (PSM) was used by matching 14 parameters. Four hundred twelve patients with COVID-19 in the ICU were included in the study. The number of patients with microorganisms ≥ 2 was 279 (67.7%). After PSM, in GII and GIII, the number of (+) tracheal cultures and (+) bloodstream cultures detected different microorganisms ≥ 2 during the ICU period, neuropathy, tracheotomized patients, duration of IMV, and length of ICU stay were significantly higher than GI. The mortality rate was similar in GI and GII, whereas it was significantly higher in GIII than in GI. The use of immunosuppressant agents in COVID-19 patients may lead to an increase in secondary infections. In addition, increased secondary infections may lead to prolonged ICU stay, prolonged IMV duration, and increased mortality.
Collapse
Affiliation(s)
- Zeynep Tugce Sarikaya
- Department of Anesthesiology and Reanimation, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey.
- General Intensive Care Unit, Acibadem Altunizade Hospital, Istanbul, Turkey.
| | - Bulent Gucyetmez
- Department of Anesthesiology and Reanimation, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
- General Intensive Care Unit, Acibadem International Hospital, Istanbul, Turkey
| | - Filiz Tuzuner
- General Intensive Care Unit, Acibadem Taksim Hospital, Istanbul, Turkey
| | - Ozlem Dincer
- General Intensive Care Unit, Acibadem Atakent Hospital, Istanbul, Turkey
- General Intensive Care Unit, Acibadem Bakırköy Hospital, Istanbul, Turkey
| | - Cenk Sahan
- General Intensive Care Unit, Acibadem Atakent Hospital, Istanbul, Turkey
- General Intensive Care Unit, Acibadem Maslak Hospital, Istanbul, Turkey
| | - Lerzan Dogan
- General Intensive Care Unit, Acibadem Altunizade Hospital, Istanbul, Turkey
| | - Serap Aktas Yildirim
- Department of Anesthesiology and Reanimation, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Rehile Zengin
- Department of Infectious Diseases and Clinical Microbiology, Acibadem Altunizade Hospital, Istanbul, Turkey
| | - Ayse Sesin Kocagoz
- Department of Infectious Disease and Clinical Microbiology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Lutfi Telci
- General Intensive Care Unit, Acibadem International Hospital, Istanbul, Turkey
| | | |
Collapse
|
23
|
Griffin DO. Postacute Sequelae of COVID (PASC or Long COVID): An Evidenced-Based Approach. Open Forum Infect Dis 2024; 11:ofae462. [PMID: 39220656 PMCID: PMC11363684 DOI: 10.1093/ofid/ofae462] [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: 06/14/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
While the acute manifestations of infectious diseases are well known, in some individuals, symptoms can either persist or appear after the acute period. Postviral fatigue syndromes are recognized with other viral infections and are described after coronavirus disease 2019 (COVID-19). We have a growing number of individuals with symptoms that persist for weeks, months, and years. Here, we share the evidence regarding the abnormalities associated with postacute sequelae of COVID-19 (PASC) and therapeutics. We describe physiological and biochemical abnormalities seen in individuals reporting PASC. We describe the several evidence-based interventions to offer patients. It is expected that this growing understanding of the mechanisms driving PASC and the benefits seen with certain therapeutics may not only lead to better outcomes for those with PASC but may also have the potential for understanding and treating other postinfectious sequelae.
Collapse
Affiliation(s)
- Daniel O Griffin
- Division of Infectious Diseases, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
24
|
Kumar S, Ramaraju K, Kakarla MS, Eranezhath SS, Chenthamarakshan C, Alagesan M, Satheesan B, Unniappan I, Wilhalme H, Pīrāgs V, Furst DE. Evaluating Personalized Add-On Ayurveda Therapy in Oxygen-Dependent Diabetic COVID-19 Patients: A 60-Day Study of Symptoms, Inflammation, and Radiological Changes. Cureus 2024; 16:e68392. [PMID: 39355453 PMCID: PMC11444340 DOI: 10.7759/cureus.68392] [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] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
Background Effective management of both acute and post-acute sequelae of SARS-CoV-2 is essential, particularly for type 2 diabetes mellitus (T2DM) patients, who are at increased risk of severe pro-inflammatory responses and complications. Persistent symptoms and residual lung and cardiovascular damage in post-coronavirus disease (COVID-19) individuals highlight the need for comprehensive long-term treatment strategies. Conventional treatments, including Remdesivir and glucocorticoids, have limitations, suggesting that further investigation into Ayurvedic therapies could be beneficial, though controlled trials are currently limited. Objectives Evaluate the effectiveness and safety of Ayurveda with the standard of care (SOC) versus SOC in improving symptoms, moderating immune responses (interleukin-6 (IL-6), C-reactive protein (CRP), neutrophil-lymphocyte ratio (NLR), and radiological outcomes in oxygen-dependent, high-risk, non-vaccinated type 2 diabetes COVID-19 patients over 60 days, and thus addressing their heightened vulnerability to severe infections. Methods A controlled trial with 50 diabetic COVID-19 patients, aged 18-80, with an NLR of >= 4, primarily on Remdesivir, was assigned to Group 1 (Add-on Ayurveda+SOC, n=30) or Group 2 (SOC, n=20) based on their voluntary choice with follow-up on days 14, 28, and 60. Parametric outcomes in group analysis were assessed with robust regression and non-parametric outcomes with Cochran-Mantel-Haenszel, log-rank test, and chi-square tests at 95% confidence interval (CI). Results Group 1 exhibited statistically significant improvements in fever, cough, diarrhea, as well as NLR, IL-6, and CRP by 14 days, and in anosmia, loss of taste, shortness of breath, general weakness, and headache by 60 days. Though the sample size is small, notable improvements can be seen in troponin levels in Group 1 at 28 and 60 days. High-resolution computer tomography COVID-19 reporting and data system (HRCT CO-RADS) scores improved more slowly in Group 2 than in Group 1. Survival rates were 96.4% for Group 1 and 90% for Group 2. Numbers were too small for reliable comparisons at 60 days. Conclusion The add-on Ayurveda group showed a better symptomatic response, and faster normalization in inflammatory markers, including IL-6 and NLR by 14 days, and cardiac markers by 28 days. Minimal clinical and no laboratory adverse events were observed. This study supports the need for a randomized, double-blind trial.
Collapse
Affiliation(s)
- Somit Kumar
- Clinical Research, AVP Research Foundation, Coimbatore, IND
- Research and Development, The Arya Vaidya Pharmacy, Coimbatore, IND
| | - Karthikeyan Ramaraju
- Respiratory Medicine, PSG Institute of Medical Sciences and Research, Coimbatore, IND
| | | | | | | | - Murali Alagesan
- General Medicine, PSG Institute of Medical Sciences and Research, Coimbatore, IND
| | - Balagopal Satheesan
- Ayurveda and Integrative Medicine, Saranya Ayurveda Hospital, Coimbatore, IND
| | - Indulal Unniappan
- Ayurveda and Integrative Medicine, AVP Research Foundation, Coimbatore, IND
| | - Holly Wilhalme
- Statistics, University of California Los Angeles, Los Angeles, USA
| | | | - Daniel E Furst
- Rheumatology, University of California Los Angeles, Los Angeles, USA
- Rheumatology, University of Washington, Seattle, USA
- Rheumatology, University of Florence, Florence, ITA
| |
Collapse
|
25
|
Silawal S, Gögele C, Pelikan P, Werner C, Levidou G, Mahato R, Schulze-Tanzil G. A Histological Analysis and Detection of Complement Regulatory Protein CD55 in SARS-CoV-2 Infected Lungs. Life (Basel) 2024; 14:1058. [PMID: 39337843 PMCID: PMC11432792 DOI: 10.3390/life14091058] [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: 07/05/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND A complement imbalance in lung alveolar tissue can play a deteriorating role in COVID-19, leading to acute respiratory distress syndrome (ARDS). CD55 is a transmembrane glycoprotein that inhibits the activation of the complement system at the intermediate cascade level, blocking the activity of the C3 convertase. OBJECTIVE In our study, lung specimens from COVID-19 and ARDS-positive COVID+/ARDS+ patients were compared with COVID-19 and ARDS-negative COVID-/ARDS- as well as COVID-/ARDS+ patients. METHODS Histochemical staining and immunolabeling of CD55 protein were performed. RESULTS The COVID-/ARDS- specimen showed higher expression and homogeneous distribution of glycosaminoglycans as well as compactly arranged elastic and collagen fibers of the alveolar walls in comparison to ARDS-affected lungs. In addition, COVID-/ARDS- lung tissues revealed stronger and homogenously distributed CD55 expression on the alveolar walls in comparison to the disrupted COVID-/ARDS+ lung tissues. CONCLUSIONS Even though the collapse of the alveolar linings and the accumulation of cellular components in the alveolar spaces were characteristic of COVID+/ARDS+ lung tissues, evaluating CD55 expression could be relevant to understand its relation to the disease. Furthermore, targeting CD55 upregulation as a potential therapy could be an option for post-infectious complications of COVID-19 and other inflammatory lung diseases in the future.
Collapse
Affiliation(s)
- Sandeep Silawal
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Petr Pelikan
- Institute for Pathology, Paracelsus Medical University, Nuremberg, General Hospital, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Christian Werner
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Georgia Levidou
- Institute for Pathology, Paracelsus Medical University, Nuremberg, General Hospital, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Raman Mahato
- Department of Emergency and Intensive Care Medicine, Klinikum Ernst von Bergmann, Charlottenstraße 72, 14467 Potsdam, Germany
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| |
Collapse
|
26
|
Kempuraj D, Aenlle KK, Cohen J, Mathew A, Isler D, Pangeni RP, Nathanson L, Theoharides TC, Klimas NG. COVID-19 and Long COVID: Disruption of the Neurovascular Unit, Blood-Brain Barrier, and Tight Junctions. Neuroscientist 2024; 30:421-439. [PMID: 37694571 DOI: 10.1177/10738584231194927] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), could affect brain structure and function. SARS-CoV-2 can enter the brain through different routes, including the olfactory, trigeminal, and vagus nerves, and through blood and immunocytes. SARS-CoV-2 may also enter the brain from the peripheral blood through a disrupted blood-brain barrier (BBB). The neurovascular unit in the brain, composed of neurons, astrocytes, endothelial cells, and pericytes, protects brain parenchyma by regulating the entry of substances from the blood. The endothelial cells, pericytes, and astrocytes highly express angiotensin converting enzyme 2 (ACE2), indicating that the BBB can be disturbed by SARS-CoV-2 and lead to derangements of tight junction and adherens junction proteins. This leads to increased BBB permeability, leakage of blood components, and movement of immune cells into the brain parenchyma. SARS-CoV-2 may also cross microvascular endothelial cells through an ACE2 receptor-associated pathway. The exact mechanism of BBB dysregulation in COVID-19/neuro-COVID is not clearly known, nor is the development of long COVID. Various blood biomarkers could indicate disease severity and neurologic complications in COVID-19 and help objectively diagnose those developing long COVID. This review highlights the importance of neurovascular and BBB disruption, as well as some potentially useful biomarkers in COVID-19, and long COVID/neuro-COVID.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Kristina K Aenlle
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
- Miami Veterans Affairs Healthcare System, Miami, FL, USA
| | - Jessica Cohen
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Annette Mathew
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Dylan Isler
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Rajendra P Pangeni
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Lubov Nathanson
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, School of Medicine, Tufts University, Boston, MA, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL, USA
- Miami Veterans Affairs Healthcare System, Miami, FL, USA
| |
Collapse
|
27
|
Zheng HY, Song TZ, Zheng YT. Immunobiology of COVID-19: Mechanistic and therapeutic insights from animal models. Zool Res 2024; 45:747-766. [PMID: 38894519 PMCID: PMC11298684 DOI: 10.24272/j.issn.2095-8137.2024.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/22/2024] [Indexed: 06/21/2024] Open
Abstract
The distribution of the immune system throughout the body complicates in vitro assessments of coronavirus disease 2019 (COVID-19) immunobiology, often resulting in a lack of reproducibility when extrapolated to the whole organism. Consequently, developing animal models is imperative for a comprehensive understanding of the pathology and immunology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This review summarizes current progress related to COVID-19 animal models, including non-human primates (NHPs), mice, and hamsters, with a focus on their roles in exploring the mechanisms of immunopathology, immune protection, and long-term effects of SARS-CoV-2 infection, as well as their application in immunoprevention and immunotherapy of SARS-CoV-2 infection. Differences among these animal models and their specific applications are also highlighted, as no single model can fully encapsulate all aspects of COVID-19. To effectively address the challenges posed by COVID-19, it is essential to select appropriate animal models that can accurately replicate both fatal and non-fatal infections with varying courses and severities. Optimizing animal model libraries and associated research tools is key to resolving the global COVID-19 pandemic, serving as a robust resource for future emerging infectious diseases.
Collapse
Affiliation(s)
- Hong-Yi Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Tian-Zhang Song
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650107, China. E-mail:
| |
Collapse
|
28
|
Low M, Suresh H, Zhou X, Bhuyan DJ, Alsherbiny MA, Khoo C, Münch G, Li CG. The wide spectrum anti-inflammatory activity of andrographolide in comparison to NSAIDs: A promising therapeutic compound against the cytokine storm. PLoS One 2024; 19:e0299965. [PMID: 39018291 PMCID: PMC11253928 DOI: 10.1371/journal.pone.0299965] [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: 02/19/2024] [Accepted: 06/26/2024] [Indexed: 07/19/2024] Open
Abstract
The challenges of the COVID-19 pandemic have highlighted an increasing clinical demand for safe and effective treatment options against an overzealous immune defence response, also known as the "cytokine storm". Andrographolide is a naturally derived bioactive compound with promising anti-inflammatory activity in many clinical studies. However, its cytokine-inhibiting activity, in direct comparison to commonly used nonsteroidal anti-inflammatory drugs (NSAIDs), has not been extensively investigated in existing literature. The anti-inflammatory activities of andrographolide and common NSAIDs, such as diclofenac, aspirin, paracetamol and ibuprofen were measured on lipopolysaccharide (LPS) and interferon-γ induced RAW264.7 cells. The levels of PGE2, nitric oxide (NO), TNF-α & LPS-induced release of pro-inflammatory cytokines on differentiated human macrophage THP-1 cells were measured against increasing concentrations of andrographolide and aforementioned NSAIDs. The associated mechanistic pathway was examined on NFκB using flow cytometry on the human endothelial-leukocyte adhesion molecule (ELAM9) (E-selectin) transfected RAW264.7 cells with green fluorescent protein (GFP). Andrographolide exhibited broad and potent anti-inflammatory and cytokine-inhibiting activity in both cell lines by inhibiting the release of IL-6, TNF-α and IFN-γ, which are known to play a key role in the etiology of cytokine storm and the pathogenesis of inflammation. In comparison, the tested NSAIDs demonstrated weak or no activity against proinflammatory mediators except for PGE2, where the activity of andrographolide (IC50 = 8.8 μM, 95% CI = 7.4 to 10.4 μM) was comparable to that of paracetamol (IC50 = 7.73 μM, 95% CI = 6.14 to 9.73 μM). The anti-inflammatory action of andrographolide was associated with its potent downregulation of NFκB. The wide-spectrum anti-inflammatory activity of andrographolide demonstrates its therapeutic potential against cytokine storms as an alternative to NSAIDs.
Collapse
Affiliation(s)
- Mitchell Low
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| | - Harsha Suresh
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
- School of Medicine, Western Sydney University, Campbelltown, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| | | | - Cheang Khoo
- Wentworth Institute of Higher Education, Surry Hills, Sydney, Australia
| | - Gerald Münch
- School of Medicine, Western Sydney University, Campbelltown, Australia
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| |
Collapse
|
29
|
Bhavaraju K, Dhiman MK, Desai H, Brien KO, Gadgil SS, Mohapatra S, Kumar V. Mitigating target interference challenges in bridging immunogenicity assay to detect anti-tocilizumab antibodies. Bioanalysis 2024; 16:587-602. [PMID: 39010827 PMCID: PMC11352699 DOI: 10.1080/17576180.2024.2349417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/26/2024] [Indexed: 07/17/2024] Open
Abstract
Aim: An assay to detect anti-tocilizumab antibodies in the presence of high levels of circulating target and drug is needed for immunogenicity assessment in comparative clinical studies.Methods: An assay was developed and validated using a combination of blocking agents and dilutions to overcome target interference challenges.Results: No false-positive signal was detected in serum samples spiked with 350-500 ng/ml of IL-6 receptor. As low as 50 ng/ml of positive control antibodies could be detected in the presence of either 500 ng/ml of IL-6 or 250 μg/ml of the drug product. Assay also demonstrated high sensitivity, selectivity and precision.Conclusion: A robust, easy to perform immunogenicity assay was developed and validated for detecting anti-tocilizumab antibodies.
Collapse
Affiliation(s)
- Kamala Bhavaraju
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Mamta Kumari Dhiman
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Hema Desai
- Clinical Pharmacology and Bioanalysis, Syneos Health, Princeton, NJ08540, USA
| | - Kyla O’ Brien
- Clinical Pharmacology and Bioanalysis, Syneos Health, Princeton, NJ08540, USA
| | - Sagarika Sunil Gadgil
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Soumyaranjan Mohapatra
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| | - Vikas Kumar
- Clinical Bioanalytics, Biological Sciences, Biologics, Dr. Reddy's Laboratories Ltd., 8-2-337, Road No.3, Banjara Hills, Hyderabad500034, Telangana, India
| |
Collapse
|
30
|
Nasir N, Khanum I, Habib K, Wagley A, Arshad A, Majeed A. Insight into COVID-19 associated liver injury: Mechanisms, evaluation, and clinical implications. HEPATOLOGY FORUM 2024; 5:139-149. [PMID: 39006140 PMCID: PMC11237249 DOI: 10.14744/hf.2023.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/25/2023] [Accepted: 11/02/2023] [Indexed: 07/16/2024]
Abstract
COVID-19 has affected millions worldwide, causing significant morbidity and mortality. While predominantly involving the respiratory tract, SARS-CoV-2 has also caused systemic illnesses involving other sites. Liver injury due to COVID-19 has been variably reported in observational studies. It has been postulated that liver damage may be due to direct damage by the SARS-CoV-2 virus or multifactorial secondary to hepatotoxic therapeutic options, as well as cytokine release syndrome and sepsis-induced multiorgan dysfunction. The approach to a COVID-19 patient with liver injury requires a thorough evaluation of the pattern of hepatocellular injury, along with the presence of underlying chronic liver disease and concurrent medications which may cause drug-induced liver injury. While studies have shown uneventful recovery in the majority of mildly affected patients, severe COVID-19 associated liver injury has been associated with higher mortality, prolonged hospitalization, and greater morbidity in survivors. Furthermore, its impact on long-term outcomes remains to be ascertained as recent studies report an association with metabolic-fatty liver disease. This present review provides insight into the subject by describing the postulated mechanism of liver injury, its impact in the presence of pre-existing liver disease, and its short- and long-term clinical implications.
Collapse
Affiliation(s)
- Nosheen Nasir
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Kiren Habib
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Abdullah Wagley
- Research Facilitation Office, Medical College, Aga Khan University, Karachi, Pakistan
| | - Aleena Arshad
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Atif Majeed
- Section of Gastroenterology, Department of Medicine, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
31
|
Li Y, Gao Z, Zheng X, Pan Y, Xu J, Li Y, Chen H. Interventional Removal of Travelling Microthrombi Using Targeted Magnetic Microbubble. Adv Healthc Mater 2024:e2401631. [PMID: 38938195 DOI: 10.1002/adhm.202401631] [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: 05/04/2024] [Revised: 06/17/2024] [Indexed: 06/29/2024]
Abstract
Microthrombus is one of the major causes of the sequelae of Corona Virus Disease 2019 (COVID-19 and leads to subsequent embolism and necrosis. Due to their small size and irregular movements, the early detection and efficient removal of microthrombi in vivo remain a great challenge. In this work, an interventional method is developed to identify and remove the traveling microthrombi using targeted-magnetic-microbubbles (TMMBs) and an interventional magnetic catheter. The thrombus-targeted drugs are coated on the TMMBs and magnetic nanoparticles are shelled inside, which allow not only targeted adhesion onto the traveling microthrombi, but also the effective capture by the magnetic catheter in the vessel. In the proof-of-concept experiments in the rat models, the concentration of microthrombus is reduced by more than 60% in 3 min, without damaging the organs. It is a promising method for treating microthrombus issues.
Collapse
Affiliation(s)
- Yongjian Li
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Zujie Gao
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Xiaobing Zheng
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Yunfan Pan
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Jinlong Xu
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Yan Li
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Haosheng Chen
- The State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
32
|
Fernández JJ, Marín A, Rosales R, Penrice-Randal R, Mlcochova P, Alvarez Y, Villalón-Letelier F, Yildiz S, Pérez E, Rathnasinghe R, Cupic A, Kehrer T, Uccellini MB, Alonso S, Martínez F, McGovern BL, Clark JJ, Sharma P, Bayón Y, Alonso A, Albrecht RA, White KM, Schotsaert M, Miorin L, Stewart JP, Hiscox JA, Gupta RK, Irigoyen N, García-Sastre A, Crespo MS, Fernández N. The IRE1α-XBP1 arm of the unfolded protein response is a host factor activated in SARS-CoV-2 infection. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167193. [PMID: 38648902 DOI: 10.1016/j.bbadis.2024.167193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 03/30/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
SARS-CoV-2 infection can cause severe pneumonia, wherein exacerbated inflammation plays a major role. This is reminiscent of the process commonly termed cytokine storm, a condition dependent on a disproportionated production of cytokines. This state involves the activation of the innate immune response by viral patterns and coincides with the biosynthesis of the biomass required for viral replication, which may overwhelm the capacity of the endoplasmic reticulum and drive the unfolded protein response (UPR). The UPR is a signal transduction pathway composed of three branches that is initiated by a set of sensors: inositol-requiring protein 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and activating transcription factor 6 (ATF6). These sensors control adaptive processes, including the transcriptional regulation of proinflammatory cytokines. Based on this background, the role of the UPR in SARS-CoV-2 replication and the ensuing inflammatory response was investigated using in vivo and in vitro models of infection. Mice and Syrian hamsters infected with SARS-CoV-2 showed a sole activation of the Ire1α-Xbp1 arm of the UPR associated with a robust production of proinflammatory cytokines. Human lung epithelial cells showed the dependence of viral replication on the expression of UPR-target proteins branching on the IRE1α-XBP1 arm and to a lower extent on the PERK route. Likewise, activation of the IRE1α-XBP1 branch by Spike (S) proteins from different variants of concern was a uniform finding. These results show that the IRE1α-XBP1 system enhances viral replication and cytokine expression and may represent a potential therapeutic target in SARS-CoV-2 severe pneumonia.
Collapse
Affiliation(s)
- Jose Javier Fernández
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arturo Marín
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Romel Rosales
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebekah Penrice-Randal
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Yolanda Alvarez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| | | | - Soner Yildiz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Enrique Pérez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Ciencias de la Salud, Universidad Europea Miguel de Cervantes (UEMC), 47012 Valladolid, Spain
| | - Raveen Rathnasinghe
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas Kehrer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa B Uccellini
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sara Alonso
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Fernando Martínez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Briana Lynn McGovern
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jordan J Clark
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Parul Sharma
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Yolanda Bayón
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| | - Andrés Alonso
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris M White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James P Stewart
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Department of Infectious Diseases, University of Georgia, GA 30602, USA
| | - Julian A Hiscox
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Infectious Diseases Horizontal Technology Centre (ID HTC), A*STAR, Singapore, Singapore; Department of Preventive Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Mariano Sánchez Crespo
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain.
| | - Nieves Fernández
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular, CSIC-Universidad de Valladolid, 47003 Valladolid, Spain; Departamento de Bioquímica, Biología Molecular y Fisiología, Universidad de Valladolid, 47003 Valladolid, Spain
| |
Collapse
|
33
|
Li L, Zhang X, Wu Y, Xing C, Du H. Challenges of mesenchymal stem cells in the clinical treatment of COVID-19. Cell Tissue Res 2024; 396:293-312. [PMID: 38512548 DOI: 10.1007/s00441-024-03881-y] [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: 06/17/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024]
Abstract
The 2019 coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has brought an enormous public health burden to the global society. The duration of the epidemic, the number of infected people, and the widespread of the epidemic are extremely rare in modern society. In the initial stage of infection, people generally show fever, cough, and dyspnea, which can lead to pneumonia, acute respiratory syndrome, kidney failure, and even death in severe cases. The strong infectivity and pathogenicity of SARS-CoV-2 make it more urgent to find an effective treatment. Mesenchymal stem cells (MSCs) are a kind of pluripotent stem cells with the potential for self-renewal and multi-directional differentiation. They are widely used in clinical experiments because of their low immunogenicity and immunomodulatory function. Mesenchymal stem cell-derived exosomes (MSC-Exo) can play a physiological role similar to that of stem cells. Since the COVID-19 pandemic, a series of clinical trials based on MSC therapy have been carried out. The results show that MSCs are safe and can significantly improve patients' respiratory function and prognosis of COVID-19. Here, the effects of MSCs and MSC-Exo in the treatment of COVID-19 are reviewed, and the clinical challenges that may be faced in the future are clarified.
Collapse
Affiliation(s)
- Luping Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing, 100083, China
- Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China
| | - Xiaoshuang Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing, 100083, China
- Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China
| | - Yawen Wu
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing, 100083, China
- Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China
| | - Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, No. 30 XueYuan Road, Haidian District, Beijing, 100083, China.
- Daxing Research Institute, University of Science and Technology Beijing, Beijing, 100083, China.
| |
Collapse
|
34
|
Toczyłowski K, Lewandowski D, Martonik D, Moniuszko-Malinowska A, Kruszewska E, Parfieniuk-Kowerda A, Flisiak R, Sulik A. Differential Inflammatory Responses in Adult and Pediatric COVID-19 Patients: Implications for Long-Term Consequences and Anti-Inflammatory Treatment. Med Sci Monit 2024; 30:e944052. [PMID: 38816982 PMCID: PMC11149468 DOI: 10.12659/msm.944052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 03/21/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND COVID-19 manifests with varying degrees of severity across different age groups; adults typically experience more severe symptoms than children. Matrix metalloproteinases (MMPs), known for their role in tissue remodeling and immune responses, may contribute to the pathophysiological disparities observed between these groups. We sought to delineate differences in serum MMP profiles between adult and pediatric COVID-19 patients, assess the influence of anti-inflammatory treatment on MMP levels, and examine potential implications for long-term consequences. MATERIAL AND METHODS Serum samples from adult and pediatric COVID-19 patients, alongside controls, were analyzed for MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-10, MMP-12, MMP-13, EMMPRIN, TNF-alpha, TIMP-1, TIMP-2, TIMP-3, and TIMP-4. A subset of adult patients received treatment with glucocorticoids, tocilizumab, and convalescent plasma, and MMP levels were compared with those of untreated patients. RESULTS Elevated levels of MMP-1, MMP-7, TIMP-1, and TIMP-2 were observed in adult and pediatric patients. Adult patients displayed higher concentrations of MMP-3, MMP-8, MMP-9, TNF-alpha, and TIMP-4 than children. Post-treatment reduction in MMP-1, MMP-8, MMP-9 levels was observed, with median decreases from 21% to 70%. MMP-3 and MMP-7 remained largely unchanged, and MMP-2 concentrations increased after treatment. Notably, anti-inflammatory treatment correlated with reduced post-treatment MMP levels, suggesting potential therapeutic benefit. CONCLUSIONS Distinctive inflammatory responses in COVID-19 were evident between adults and children. While certain MMPs exhibited post-treatment reduction, the persistence of elevated levels raises concerns about potential long-term consequences, including lung fibrosis. Our findings emphasize the need for personalized treatment strategies and further investigation into the dynamics of MMP regulation in COVID-19.
Collapse
Affiliation(s)
- Kacper Toczyłowski
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| | - Dawid Lewandowski
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| | - Diana Martonik
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, Białystok, Poland
| | - Ewelina Kruszewska
- Department of Infectious Diseases and Neuroinfections, Medical University of Białystok, Białystok, Poland
| | - Anna Parfieniuk-Kowerda
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland
| | - Artur Sulik
- Department of Pediatric Infectious Diseases, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
35
|
Trisyani Y, Mahendra D, Nuraeni A. Lessons Learned from the Lived Experiences of COVID-19 ICU Survivors Who are Struggling Through Critical Conditions and Surviving to Champion Life: A Qualitative Study. J Multidiscip Healthc 2024; 17:2659-2669. [PMID: 38828267 PMCID: PMC11141573 DOI: 10.2147/jmdh.s380389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
Background Due to the coronavirus disease 2019 (COVID-19) pandemic, millions of lives were lost globally, including in Indonesia. Some patients with COVID-19 may experience severe symptoms of hypoxia, while some may be critically ill and admitted to the intensive care unit (ICU) for survival. Purpose This study aimed to understand the lived experiences of COVID-19 ICU survivors who were in a critical condition. Methods This phenomenological study used semistructured interviews with nine participants who were COVID-19 ICU survivors. Data analysis was performed using the Colaizzi approach. Results The phenomenon of the lived experiences of COVID-19 ICU survivors was presented in seven subthemes and four main themes: struggling in a state of helplessness, fostering a positive spirit from within, amplifying the support from nurses and doctors, and strengthening the connection with family and the Almighty. These themes indicated the essential aspects of psychosocial support needed to boost strength and energy and elevate the body's immune system, which is crucial to champion life through critical conditions. Conclusion The new insight resulting from the study is shown in the four main themes, which play a significant role in elevating the healing process and enabling patients to survive critical conditions. Therefore, this study recommends the importance of psychosocial support for patients with critical conditions, which involves family and their significant others, and facilitating the connection between the patient and God.
Collapse
Affiliation(s)
- Yanny Trisyani
- Department of Critical Care and Emergency Nursing, Faculty of Nursing, Universitas Padjadjaran, Sumedang, Jawa Barat, Indonesia
| | - Donny Mahendra
- Vocational Faculty, Universitas Kristen Indonesia, Jakarta, Indonesia
| | - Aan Nuraeni
- Department of Critical Care and Emergency Nursing, Faculty of Nursing, Universitas Padjadjaran, Sumedang, Jawa Barat, Indonesia
| |
Collapse
|
36
|
Żegleń S, Nagajewski A, Górski D, Wojarski J, Karlsen W, Akily L, Sunesson F, Sawczuk M, Nojek R, Lipka K, Gallas M, Dukat-Mazurek A, Karolak W. Treatment of Patients After Lung Transplantation With Covid Infection During Long-Term Follow-Up. Transplant Proc 2024; 56:881-884. [PMID: 38714369 DOI: 10.1016/j.transproceed.2024.03.010] [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: 12/30/2023] [Accepted: 03/26/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Patients undergoing lung transplantation are routinely managed with lifelong immunosuppression, which is associated with a heightened risk for infections. This study delves into the therapeutic challenges and strategies for managing lung transplant recipients (LTRs) infected with COVID-19 during long-term follow-up. METHODS The was a case series analysis, among which nonstandard therapies consisting of targeted antibody treatment, antiviral drugs, or anti-interleukin-6 drugs were applied in patients after lung transplantation. Additional analysis of laboratory test results for systemic inflammation and imaging studies was also carried out. The study was limited to a dedicated COVID-19 center, commonly known as a temporary hospital, and included patients infected with COVID-19 in the late post-lung transplant period (home-related infection). RESULTS Fifteen post-lung transplantation patients with current COVID-19 infection were treated with antibodies such as tocilizumab, casirivimab, imdevimab, and regdanvimab. Of these patients, 1 was given tocilizumab (7%), 8 casirivimab and imdevimab (53%), and 2 regdanvimab (13%). Of the 15 lung transplant recipients studied, 8 presented COVID-19-associated lung changes in computed tomography scans (53%). Common clinical manifestations included dyspnea, fever, and fatigue. Antiviral agents, like remdesivir, were employed in the remaining 4 cases (27%), and adjunctive therapies, such as corticosteroids and anticoagulants, were used selectively. All treated patients survived the infection without complications; the treatment proved effective and safe.
Collapse
Affiliation(s)
- Sławomir Żegleń
- Department of Pneumonology and Allergology, Medical University of Gdansk, Poland.
| | - Adam Nagajewski
- Temporary Hospital - Szczytno, Department of Internal Diseases and Infectious Diseases, Szczytno, Poland
| | - Dominik Górski
- Temporary Hospital - Szczytno, Department of Internal Diseases and Infectious Diseases, Szczytno, Poland
| | - Jacek Wojarski
- Depatment of Cardiac Surgery, Medical University of Gdańsk, Poland
| | - William Karlsen
- Scientific Circle of Lung Transplantology, Department of Lung Transplantation, Medical University of Gdansk, Poland
| | - Lin Akily
- Scientific Circle of Lung Transplantology, Department of Lung Transplantation, Medical University of Gdansk, Poland
| | - Fanny Sunesson
- Emergency Department of Surgery (KAVA), Kristianstad, Sweden
| | - Marcin Sawczuk
- Depatment of Cardiac Surgery, Medical University of Gdańsk, Poland
| | - Rafał Nojek
- Department of Applied Computer Science, AGH University of Science and Technology in Krakow, Poland
| | - Karolina Lipka
- Depatment of Cardiac Surgery, Medical University of Gdańsk, Poland
| | - Marta Gallas
- Institute of Nursing and Midwifery, Department of Nursing Management, Medical University of Gdansk, Poland
| | | | - Wojtek Karolak
- Depatment of Cardiac Surgery, Medical University of Gdańsk, Poland
| |
Collapse
|
37
|
Chávez-Pacheco JL, Castillejos-López M, Hernández-Regino LM, Velasco-Hidalgo L, Zapata-Tarres M, Correa-Carranza V, Rosario-Méndez G, Barrientos-Ríos R, Aquino-Gálvez A, Torres-Espíndola LM. Challenges in Treating Pediatric Cancer Patients during the COVID-19 Pandemic: Balancing Risks and Care. Viruses 2024; 16:690. [PMID: 38793571 PMCID: PMC11125850 DOI: 10.3390/v16050690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
The COVID-19 pandemic has resulted in millions of fatalities worldwide. The case of pediatric cancer patients stands out since, despite being considered a population at risk, few studies have been carried out concerning symptom detection or the description of the mechanisms capable of modifying the course of the COVID-19 disease, such as the interaction and response between the virus and the treatment given to cancer patients. By synthesizing existing studies, this paper aims to expose the treatment challenges for pediatric patients with COVID-19 in an oncology context. Additionally, this updated review includes studies that utilized the antiviral agents Remdesivir and PaxlovidTM in pediatric cancer patients. There is no specific treatment designed exclusively for pediatric cancer patients dealing with COVID-19, and it is advisable to avoid self-medication to prevent potential side effects. Managing COVID-19 in pediatric cancer patients is indeed a substantial challenge. New strategies, such as chemotherapy application rooms, have been implemented for children with cancer who were positive for COVID-19 but asymptomatic since the risk of disease progression is greater than the risk of complications from SARS-CoV-2.
Collapse
Affiliation(s)
- Juan Luis Chávez-Pacheco
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Manuel Castillejos-López
- Epidemiology and Infectology, National Institute of Respiratory Diseases, Mexico City 14080, Mexico;
| | - Laura M. Hernández-Regino
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | | | - Marta Zapata-Tarres
- Head of Research Coordination at Mexican Social Security Institute Foundation, Mexico City 06600, Mexico;
| | - Valeria Correa-Carranza
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Guillermo Rosario-Méndez
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| | - Rehotbevely Barrientos-Ríos
- Cytogenetics Laboratory, Department of Human Genetics, National Institute of Pediatrics, Mexico City 04530, Mexico;
| | - Arnoldo Aquino-Gálvez
- Molecular Biology Laboratory, Pulmonary Fibrosis Department, National Institute of Respiratory Diseases, Mexico City 14080, Mexico
| | - Luz María Torres-Espíndola
- Pharmacology Laboratory, National Institute of Pediatrics, Mexico City 04530, Mexico; (J.L.C.-P.); (L.M.H.-R.); (V.C.-C.); (G.R.-M.)
| |
Collapse
|
38
|
Wu S, Liao G, Mao J, Yan H, Chen J, Peng J. Factors Associated with Mortality Among Severe Omicron Patients for COVID-19. Infect Drug Resist 2024; 17:1309-1319. [PMID: 38585415 PMCID: PMC10999197 DOI: 10.2147/idr.s450504] [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: 12/17/2023] [Accepted: 03/13/2024] [Indexed: 04/09/2024] Open
Abstract
Purpose The purpose of the study was to explore the potential risk factors of mortality in patients with severe pneumonia during the omicron pandemic in South China in 2022. Methods Clinical data was collected from patients hospitalized with omicron COVID-19. Then, patients were categorized into the non-survival and survival groups. A comprehensive analysis was conducted to analyze the factors associated with negative outcome in individuals suffering from severe omicron COVID-19. Results In this study, 155 severe COVID-19 patients were included, comprising 55 non-survivors and 100 survivors. Non-survivors, in comparison to survivors, exhibited elevated levels of various biomarkers including neutrophil count, hypersensitive troponin T, urea, creatinine, C-reactive protein, procalcitonin, interleukin-6, plasma D-dimer, and derived neutrophil-to-lymphocyte ratio (dNLR) (P < 0.05). They also displayed reduced lymphocyte count, platelet count, and albumin levels (P < 0.05) and were more prone to developing comorbidities, including shock, acute cardiac and renal injury, acute respiratory distress syndrome, coagulation disorders, and secondary infections. Platelet count (PLT) <100 × 10^/L, interleukin-6 (IL-6) >100 pg/mL, and dNLR >5.0 independently contributed to the risk of death in patients suffering from severe COVID-19. Conclusion PLT, IL-6, and dNRL independently contributed to the risk of mortality in patients with severe pneumonia during the 2022 omicron pandemic in South China.
Collapse
Affiliation(s)
- Shuting Wu
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Guichan Liao
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jingchun Mao
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Haiming Yan
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Juanjuan Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
39
|
Steuber TD, Rosandich T, Cadwallader T, Steil L, Belk M, Yendrapalli U, Hassoun A, Edwards J. Dosing and Administration Strategies of Tocilizumab in Patients With COVID-19: A Retrospective Cohort Analysis. Ann Pharmacother 2024; 58:391-397. [PMID: 37522616 DOI: 10.1177/10600280231190401] [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: 08/01/2023] Open
Abstract
BACKGROUND Tocilizumab may reduce the risk of death, length of stay, and time of mechanical ventilation in patients hospitalized with COVID-19. Limited data are available evaluating low-dose subcutaneous administration of tocilizumab in this setting. OBJECTIVE To compare outcomes of 2 tocilizumab dosing and administration strategies in patients hospitalized with COVID-19. METHODS A retrospective, observational cohort study was conducted to compare clinical outcomes in patients hospitalized with COVID-19 receiving tocilizumab 400 mg intravenously (400 mg IV) or 162 mg subcutaneously (162 mg SC). Hospitalized patients receiving a single dose of tocilizumab were eligible for inclusion and grouped by dosing and administration strategy. The primary endpoint was ventilator-free days at day 28. Secondary endpoints included length of stay (LOS), intensive care unit (ICU) LOS, mechanical ventilation required after dose, 28-day readmission, 28-day mortality, and change in inflammatory markers. RESULTS A total of 303 patients were included, with 147 who received tocilizumab 400 mg IV and 156 who received 162 mg SC. There was no significant difference in average ventilator-free days at day 28 in patients receiving 400 mg IV compared with 162 mg SC (26.4 ± 5.3 vs 25.6 ± 6.8 days, respectively; P = 0.812). There was also no difference in LOS (10.4 ± 12.6 vs 10.5 ± 14.0 days; P = 0.637), ICU LOS (3.9 ± 9.0 vs 3.5 ± 8.3 days; P = 0.679), mechanical ventilation after dose (15.6% vs 19.2%; P = 0.412), 28-day readmission (6.1% vs 9.6%; P = 0.268), or 28-day mortality (23.1% vs 25.6%; P = 0.611). Finally, there was no difference regarding change in inflammatory markers at 48 hours (P > 0.05 for all interactions). CONCLUSION AND RELEVANCE In this retrospective study involving hospitalized patients with COVID-19, there was no difference between tocilizumab 162 mg SC and 400 mg IV in terms of efficacy. The 162 mg SC dose may be a reasonable alternative to traditional doses.
Collapse
Affiliation(s)
- Taylor D Steuber
- School of Pharmacy, University of Missouri-Kansas City, Columbia, MO, USA
- Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Thomas Rosandich
- Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | | | - Lauren Steil
- Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Madeline Belk
- Department of Pharmacy, Huntsville Hospital, Huntsville, AL, USA
| | - Usha Yendrapalli
- Department of Internal Medicine, Huntsville Hospital, Huntsville, AL, USA
| | - Ali Hassoun
- Alabama Infectious Disease Center, Huntsville, AL, USA
| | - Jonathan Edwards
- Department of Pharmacy, Huntsville Hospital, Huntsville, AL, USA
| |
Collapse
|
40
|
Tong T, Jin YH, Wang M, Gong FQ. Treatment of multisystem inflammatory syndrome in children. World J Pediatr 2024; 20:325-339. [PMID: 38509432 DOI: 10.1007/s12519-024-00798-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/29/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Multisystem inflammatory syndrome in children (MIS-C), a relatively uncommon but severe pediatric complication, is associated with coronavirus disease 2019 (COVID-19). A variety of treatment approaches, including intravenous immunoglobulins (IVIGs), glucocorticoids (GCs) and biologic agents, such as anakinra and infliximab, have been described for the management of COVID-19-related MIS-C. Anticoagulant therapy is also important. However, a well-developed treatment system has not been established, and many issues remain controversial. Several recently published articles related to the treatment of MIS-C have been released. Hence, in this review, we identified relevant articles published recently and summarized the treatment of MIS-C more comprehensively and systematically. DATA SOURCES We reviewed the literature on the treatment of MIS-C through 20 September 2023. The PubMed/Medline, Web of Science, EMBASE, and Cochrane Library databases were searched with the combination of the terms "multisystem inflammatory syndrome", "MIS-C", "PIMS-TS", "therapy", "treatment", "drug", "IVIG", "GCs", "intravenous immunoglobulin", "corticosteroids", "biological agent", and "aspirin". RESULTS The severity of MIS-C varies, and different treatment schemes should be used according to the specific condition. Ongoing research and data collection are vital to better understand the pathophysiology and optimal management of MIS-C. CONCLUSIONS MIS-C is a disease involving multiple systems and has great heterogeneity. With the accumulation of additional experience, we have garnered fresh insights into its treatment strategies. However, there remains a critical need for greater standardization in treatment protocols, alongside the pressing necessity for more robust and meticulously conducted studies to deepen our understanding of these protocols. Supplementary file1 (MP4 208044 kb).
Collapse
Affiliation(s)
- Tong Tong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China
| | - Yi-Hua Jin
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No. 3333 Binsheng Road, Hangzhou, 310052, China
| | - Min 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, China
| | - Fang-Qi 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, China.
| |
Collapse
|
41
|
Shang W, Hu X, Lin X, Li S, Xiong S, Huang B, Wang X. Iterative In Silico Screening for Optimizing Stable Conformation of Anti-SARS-CoV-2 Nanobodies. Pharmaceuticals (Basel) 2024; 17:424. [PMID: 38675386 PMCID: PMC11054880 DOI: 10.3390/ph17040424] [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: 03/01/2024] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
Nanobodies (Nbs or VHHs) are single-domain antibodies (sdAbs) derived from camelid heavy-chain antibodies. Nbs have special and unique characteristics, such as small size, good tissue penetration, and cost-effective production, making Nbs a good candidate for the diagnosis and treatment of viruses and other pathologies. Identifying effective Nbs against COVID-19 would help us control this dangerous virus or other unknown variants in the future. Herein, we introduce an in silico screening strategy for optimizing stable conformation of anti-SARS-CoV-2 Nbs. Firstly, various complexes containing nanobodies were downloaded from the RCSB database, which were identified from immunized llamas. The primary docking between Nbs and the SARS-CoV-2 spike protein receptor-binding domain was performed through the ClusPro program, with the manual screening leaving the reasonable conformation to the next step. Then, the binding distances of atoms between the antigen-antibody interfaces were measured through the NeighborSearch algorithm. Finally, filtered nanobodies were acquired according to HADDOCK scores through HADDOCK docking the COVID-19 spike protein with nanobodies under restrictions of calculated molecular distance between active residues and antigenic epitopes less than 4.5 Å. In this way, those nanobodies with more reasonable conformation and stronger neutralizing efficacy were acquired. To validate the efficacy ranking of the nanobodies we obtained, we calculated the binding affinities (∆G) and dissociation constants (Kd) of all screened nanobodies using the PRODIGY web tool and predicted the stability changes induced by all possible point mutations in nanobodies using the MAESTROWeb server. Furthermore, we examined the performance of the relationship between nanobodies' ranking and their number of mutation-sensitive sites (Spearman correlation > 0.68); the results revealed a robust correlation, indicating that the superior nanobodies identified through our screening process exhibited fewer mutation hotspots and higher stability. This correlation analysis demonstrates the validity of our screening criteria, underscoring the suitability of these nanobodies for future development and practical implementation. In conclusion, this three-step screening strategy iteratively in silico greatly improved the accuracy of screening desired nanobodies compared to using only ClusPro docking or default HADDOCK docking settings. It provides new ideas for the screening of novel antibodies and computer-aided screening methods.
Collapse
Affiliation(s)
| | | | | | | | | | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China; (W.S.); (X.H.); (X.L.); (S.L.); (S.X.)
| | - Xin Wang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen 518118, China; (W.S.); (X.H.); (X.L.); (S.L.); (S.X.)
| |
Collapse
|
42
|
Wang M, Chen L, He J, Xia W, Ye Z, She J. Structural insights into IL-6 signaling inhibition by therapeutic antibodies. Cell Rep 2024; 43:113819. [PMID: 38393945 DOI: 10.1016/j.celrep.2024.113819] [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: 08/22/2023] [Revised: 12/14/2023] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Antibody inhibitors of the interleukin-6 (IL-6) signaling pathway, such as tocilizumab and sarilumab, have been used to treat rheumatoid arthritis, chimeric antigen receptor T cell-induced cytokine storm, and severe COVID-19 pneumonia. Here, we solve the cryogenic electron microscopy structures of sarilumab and tocilizumab in complex with IL-6R to resolutions of 3.2 and 3.3 Å, respectively. These structures reveal that both tocilizumab and sarilumab bind to the D3 domain of IL-6R. The binding surfaces of the two antibodies largely overlap, but the detailed interactions are different. Functional studies of various mutants show results consistent with our structural analysis of the antibodies and IL-6R interactions. Structural comparisons with the IL-6/IL-6R/gp130 complex indicate that sarilumab and tocilizumab probably inhibit IL-6/IL-6R signaling by competing for the IL-6 binding site. In summary, this work reveals the antibody-blocking mechanism of the IL-6 signaling pathway and paves the way for future antibody discovery.
Collapse
Affiliation(s)
- Mingxing Wang
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Long Chen
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Jin He
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Wenqiang Xia
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, Zhejiang, China; College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zihong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, Zhejiang, China.
| | - Ji She
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China.
| |
Collapse
|
43
|
Huang W, Liu W, Yu T, Zhang Z, Zhai L, Huang P, Lu Y. Effect of anti-COVID-19 drugs on patients with cancer. Eur J Med Chem 2024; 268:116214. [PMID: 38367490 DOI: 10.1016/j.ejmech.2024.116214] [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: 07/31/2023] [Revised: 01/11/2024] [Accepted: 02/01/2024] [Indexed: 02/19/2024]
Abstract
The clinical treatment of patients with cancer who are also diagnosed with coronavirus disease (COVID-19) has been a challenging issue since the outbreak of COVID-19. Therefore, it is crucial to understand the effects of commonly used drugs for treating COVID-19 in patients with cancer. Hence, this review aims to provide a reference for the clinical treatment of patients with cancer to minimize the losses caused by the COVID-19 pandemic. In this study, we also focused on the relationship between COVID-19, commonly used drugs for treating COVID-19, and cancer. We specifically investigated the effect of these drugs on tumor cell proliferation, migration, invasion, and apoptosis. The potential mechanisms of action of these drugs were discussed and evaluated. We found that most of these drugs showed inhibitory effects on tumors, and only in a few cases had cancer-promoting effects. Furthermore, inappropriate usage of these drugs may lead to irreversible kidney and heart damage. Finally, we have clarified the use of different drugs, which can provide useful guidance for the clinical treatment of cancer patients diagnosed with COVID-19.
Collapse
Affiliation(s)
- Weicai Huang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Wenyu Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Tingting Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Zhaoyang Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Lingyun Zhai
- Gynecology Department, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Panpan Huang
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China.
| | - Yao Lu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
44
|
Handa Y, Okuwaki K, Kawashima Y, Hatada R, Mochizuki Y, Komeiji Y, Tanaka S, Furuishi T, Yonemochi E, Honma T, Fukuzawa K. Prediction of Binding Pose and Affinity of Nelfinavir, a SARS-CoV-2 Main Protease Repositioned Drug, by Combining Docking, Molecular Dynamics, and Fragment Molecular Orbital Calculations. J Phys Chem B 2024; 128:2249-2265. [PMID: 38437183 PMCID: PMC10946393 DOI: 10.1021/acs.jpcb.3c05564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 03/06/2024]
Abstract
A novel in silico drug design procedure is described targeting the Main protease (Mpro) of the SARS-CoV-2 virus. The procedure combines molecular docking, molecular dynamics (MD), and fragment molecular orbital (FMO) calculations. The binding structure and properties of Mpro were predicted for Nelfinavir (NFV), which had been identified as a candidate compound through drug repositioning, targeting Mpro. Several poses of the Mpro and NFV complexes were generated by docking, from which four docking poses were selected by scoring with FMO energy. Then, each pose was subjected to MD simulation, 100 snapshot structures were sampled from each of the generated MD trajectories, and the structures were evaluated by FMO calculations to rank the pose based on binding energy. Several residues were found to be important in ligand recognition, including Glu47, Asp48, Glu166, Asp187, and Gln189, all of which interacted strongly with NFV. Asn142 is presumably regarded to form hydrogen bonds or CH/π interaction with NFV; however, in the present calculation, their interactions were transient. Moreover, the tert-butyl group of NFV had no interaction with Mpro. Identifying such strong and weak interactions provides candidates for maintaining and substituting ligand functional groups and important suggestions for drug discovery using drug repositioning. Besides the interaction between NFV and the amino acid residues of Mpro, the desolvation effect of the binding pocket also affected the ranking order. A similar procedure of drug design was applied to Lopinavir, and the calculated interaction energy and experimental inhibitory activity value trends were consistent. Our approach provides a new guideline for structure-based drug design starting from a candidate compound whose complex crystal structure has not been obtained.
Collapse
Affiliation(s)
- Yuma Handa
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
- Graduate
School of Pharmaceutical Sciences, Osaka
University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koji Okuwaki
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
- Department
of Chemistry and Research Center for Smart Molecules, Faculty of Science, Rikkyo University, 3-34-1 Nishi-ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
| | - Yusuke Kawashima
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Ryo Hatada
- Department
of Chemistry and Research Center for Smart Molecules, Faculty of Science, Rikkyo University, 3-34-1 Nishi-ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
| | - Yuji Mochizuki
- Department
of Chemistry and Research Center for Smart Molecules, Faculty of Science, Rikkyo University, 3-34-1 Nishi-ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
- Institute
of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Yuto Komeiji
- Graduate
School of Pharmaceutical Sciences, Osaka
University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department
of Chemistry and Research Center for Smart Molecules, Faculty of Science, Rikkyo University, 3-34-1 Nishi-ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
- Health
and Medical Research Institute, AIST, Tsukuba Central 6, Tsukuba, Ibaraki 305-8566, Japan
- RIKEN
Center
for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Shigenori Tanaka
- Graduate
School of System Informatics, Department of Computational Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Takayuki Furuishi
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Etsuo Yonemochi
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Teruki Honma
- RIKEN
Center
for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Kaori Fukuzawa
- Department
of Physical Chemistry, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan
- Graduate
School of Pharmaceutical Sciences, Osaka
University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department
of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, 6-6-11 Aoba, Aramaki, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
45
|
Samarelli F, Graziano G, Gambacorta N, Graps EA, Leonetti F, Nicolotti O, Altomare CD. Small Molecules for the Treatment of Long-COVID-Related Vascular Damage and Abnormal Blood Clotting: A Patent-Based Appraisal. Viruses 2024; 16:450. [PMID: 38543815 PMCID: PMC10976273 DOI: 10.3390/v16030450] [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: 01/07/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 05/23/2024] Open
Abstract
People affected by COVID-19 are exposed to, among others, abnormal clotting and endothelial dysfunction, which may result in deep vein thrombosis, cerebrovascular disorders, and ischemic and non-ischemic heart diseases, to mention a few. Treatments for COVID-19 include antiplatelet (e.g., aspirin, clopidogrel) and anticoagulant agents, but their impact on morbidity and mortality has not been proven. In addition, due to viremia-associated interconnected prothrombotic and proinflammatory events, anti-inflammatory drugs have also been investigated for their ability to mitigate against immune dysregulation due to the cytokine storm. By retrieving patent literature published in the last two years, small molecules patented for long-COVID-related blood clotting and hematological complications are herein examined, along with supporting evidence from preclinical and clinical studies. An overview of the main features and therapeutic potentials of small molecules is provided for the thromboxane receptor antagonist ramatroban, the pan-caspase inhibitor emricasan, and the sodium-hydrogen antiporter 1 (NHE-1) inhibitor rimeporide, as well as natural polyphenolic compounds.
Collapse
Affiliation(s)
- Francesco Samarelli
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, I-70125 Bari, Italy; (F.S.); (G.G.); (N.G.); (F.L.); (O.N.)
| | - Giovanni Graziano
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, I-70125 Bari, Italy; (F.S.); (G.G.); (N.G.); (F.L.); (O.N.)
| | - Nicola Gambacorta
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, I-70125 Bari, Italy; (F.S.); (G.G.); (N.G.); (F.L.); (O.N.)
| | - Elisabetta Anna Graps
- ARESS Puglia—Agenzia Regionale Strategica per la Salute ed il Sociale, I-70121 Bari, Italy;
| | - Francesco Leonetti
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, I-70125 Bari, Italy; (F.S.); (G.G.); (N.G.); (F.L.); (O.N.)
| | - Orazio Nicolotti
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, I-70125 Bari, Italy; (F.S.); (G.G.); (N.G.); (F.L.); (O.N.)
| | | |
Collapse
|
46
|
Nasiri Z, Soleimanjahi H, Baheiraei N, Hashemi SM, Pourkarim MR. The impact understanding of exosome therapy in COVID-19 and preparations for the future approaches in dealing with infectious diseases and inflammation. Sci Rep 2024; 14:5724. [PMID: 38459174 PMCID: PMC10924089 DOI: 10.1038/s41598-024-56334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/05/2024] [Indexed: 03/10/2024] Open
Abstract
Cytokine storms, which result from an abrupt, acute surge in the circulating levels of different pro-inflammatory cytokines, are one of the complications associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This study aimed to assess the effect of exosomes on the release of pro-inflammatory cytokines in patients with coronavirus disease 2019 (COVID-19) and compare it with a control group. The cytokines evaluated in this study were TNF-α, IL-6, IL-17, and IFN-γ. The study compared the levels of these pro-inflammatory cytokines in the peripheral blood mononuclear cells (PBMCs) of five COVID-19 patients in the intensive care unit, who were subjected to both inactivated SARS-CoV-2 and exosome therapy, with those of five healthy controls. The cytokine levels were quantified using the ELISA method. The collected data was analyzed in SPSS Version 26.0 and GraphPad Prism Version 9. According to the study findings, when PBMCs were exposed to inactivated SARS-CoV-2, pro-inflammatory cytokines increased in both patients and healthy controls. Notably, the cytokine levels were significantly elevated in the COVID-19 patients compared to the control group P-values were < 0.001, 0.001, 0.008, and 0.008 for TNF-α, IL-6, IL-17, and IFN-γ, respectively. Conversely, when both groups were exposed to exosomes, there was a marked reduction in the levels of pro-inflammatory cytokines. This suggests that exosome administration can effectively mitigate the hyperinflammation induced by COVID-19 by suppressing the production of pro-inflammatory cytokines in patients. These findings underscore the potential safety and efficacy of exosomes as a therapeutic strategy for COVID-19.
Collapse
Affiliation(s)
- Zeynab Nasiri
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Nafiseh Baheiraei
- Department of Anatomical Science, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Pourkarim
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
47
|
Su S, Hu W, Chen X, Ren Y, Lu Y, Shi J, Zhang T, Zhang H, Wang M, Wang Y, Zhao F, Jin R, Liu Y, Zhang H, Liu G. Cardiac injury progression in children with multisystem inflammatory syndrome associated with SARS-CoV-2 infection: a review. Front Pediatr 2024; 12:1348016. [PMID: 38510081 PMCID: PMC10950994 DOI: 10.3389/fped.2024.1348016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
The symptoms and signs of infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are milder in children than in adults. However, in April 2020, British pediatricians first reported that coronavirus disease 2019 (COVID-19) may present as multisystem inflammatory syndrome in children and adolescents (MIS-C), similar to that observed in Kawasaki disease. MIS-C can be associated with multiple systemic injuries and even death in children. In addition to digestive system involvement, cardiac injury is prominent. This article reviews the pathogenesis, clinical manifestations, and treatment of cardiac injury caused by MIS-C, which may help clinicians in early diagnosis and timely commencement of treatment.
Collapse
Affiliation(s)
- Song Su
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Wandong Hu
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Xiao Chen
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Ying Ren
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Yi Lu
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Jianguo Shi
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Tong Zhang
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Huan Zhang
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Meng Wang
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Yaping Wang
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Fen Zhao
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Ruifeng Jin
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Yong Liu
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Hongwei Zhang
- Epilepsy Center, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Epilepsy Center, Jinan Children's Hospital, Jinan, Shandong, China
| | - Guohua Liu
- Department of Ophthalmology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Department of Ophthalmology, Jinan Children's Hospital, Jinan, Shandong, China
| |
Collapse
|
48
|
Ferrer MD, Reynés C, Jiménez L, Malagraba G, Monserrat-Mesquida M, Bouzas C, Sureda A, Tur JA, Pons A. Nitrite Attenuates the In Vitro Inflammatory Response of Immune Cells to the SARS-CoV-2 S Protein without Interfering in the Antioxidant Enzyme Activation. Int J Mol Sci 2024; 25:3001. [PMID: 38474248 DOI: 10.3390/ijms25053001] [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: 01/18/2024] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
SARS-CoV-2 induces a hyperinflammatory reaction due to the excessive release of cytokines during the immune response. The bacterial endotoxin lipopolysaccharide (LPS) contributes to the low-grade inflammation associated with the metabolic syndrome, enhancing the hyperinflammatory reaction induced by the SARS-CoV-2 infection. The intake of sodium nitrate, a precursor of nitrite and nitric oxide, influences the antioxidant and pro-inflammatory gene expression profile after immune stimulation with LPS in peripheral blood mononuclear cells from metabolic syndrome patients. We aimed to assess the inflammatory and antioxidant responses of immune cells from metabolic syndrome patients to exposure to the SARS-CoV-2 spike protein (S protein) together with LPS and the effect of nitrite in these responses. Whole blood samples obtained from six metabolic syndrome patients were cultured for 16 h at 37 °C with four different media: control medium, control medium plus LPS (100 ng/mL), control medium plus LPS (100 ng/mL) plus S protein (10 ng/mL), and control medium plus LPS (100 ng/mL) plus S protein (10 ng/mL) plus nitrite (5 µM). Immune stimulation with the LPS/S protein enhanced nitrate biosynthesis from nitrite oxidation and probably from additional organic precursors. In vitro incubations with the LPS/S protein enhanced the expression and/or release of pro-inflammatory TNFα, IL-6, IL-1β, and TLR4, as well as the expression of the anti-inflammatory IL-1ra and IL-10 and antioxidant enzymes. Nitrite attenuated the pro- and anti-inflammatory response induced by the S protein without interfering with the activation of TLR4 and antioxidant enzyme expression, raising the possibility that nitrite could have potential as a coadjutant in the treatment of COVID-19.
Collapse
Affiliation(s)
- Miguel D Ferrer
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Clara Reynés
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
| | - Laura Jiménez
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
| | - Gianluca Malagraba
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
| | - Margalida Monserrat-Mesquida
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Cristina Bouzas
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Josep A Tur
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antoni Pons
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
49
|
Barosa M, Jamrozik E, Prasad V. The Ethical Obligation for Research During Public Health Emergencies: Insights From the COVID-19 Pandemic. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2024; 27:49-70. [PMID: 38153559 PMCID: PMC10904511 DOI: 10.1007/s11019-023-10184-6] [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] [Accepted: 11/06/2023] [Indexed: 12/29/2023]
Abstract
In times of crises, public health leaders may claim that trials of public health interventions are unethical. One reason for this claim can be that equipoise-i.e. a situation of uncertainty and/or disagreement among experts about the evidence regarding an intervention-has been disturbed by a change of collective expert views. Some might claim that equipoise is disturbed if the majority of experts believe that emergency public health interventions are likely to be more beneficial than harmful. However, such beliefs are not always justified: where high quality research has not been conducted, there is often considerable residual uncertainty about whether interventions offer net benefits. In this essay we argue that high-quality research, namely by means of well-designed randomized trials, is ethically obligatory before, during, and after implementing policies in public health emergencies (PHEs). We contend that this standard applies to both pharmaceutical and non-pharmaceutical interventions, and we elaborate an account of equipoise that captures key features of debates in the recent pandemic. We build our case by analyzing research strategies employed during the COVID-19 pandemic regarding drugs, vaccines, and non-pharmaceutical interventions; and by providing responses to possible objections. Finally, we propose a public health policy reform: whenever a policy implemented during a PHE is not grounded in high-quality evidence that expected benefits outweigh harms, there should be a planned approach to generate high-quality evidence, with review of emerging data at preset time points. These preset timepoints guarantee that policymakers pause to review emerging evidence and consider ceasing ineffective or even harmful policies, thereby improving transparency and accountability, as well as permitting the redirection of resources to more effective or beneficial interventions.
Collapse
Affiliation(s)
- Mariana Barosa
- Nova Medical School, Nova University of Lisbon, Lisbon, Portugal
- Science and Technologies Studies (MSc student), University College London, London, UK
| | - Euzebiusz Jamrozik
- Ethox and Pandemic Sciences Institute, University of Oxford, Oxford, UK
- Royal Melbourne Hospital Department of Medicine, University of Melbourne, Melbourne, Australia
- Monash Bioethics Centre, Monash University, Melbourne, Australia
| | - Vinay Prasad
- University of California, San Francisco, 550 16th St, San Francisco, CA, 94158, USA.
| |
Collapse
|
50
|
Ruchiwit P, Pongtongkam K, Saiphoklang N. dCROX and ROX Indices Predict Clinical Outcomes in Patients with COVID-19 Pneumonia Treated with High-Flow Nasal Cannula Oxygen Therapy. Crit Care Res Pract 2024; 2024:8880259. [PMID: 38450049 PMCID: PMC10917475 DOI: 10.1155/2024/8880259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 01/29/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
Background High-flow nasal cannula (HFNC) therapy is a common respiratory support in patients with COVID-19 pneumonia. Predictive tools for the evaluation of successful weaning from HFNC therapy for COVID-19 pneumonia have been limited. This study aimed to develop a new predictor for weaning success from HFNC treatment in patients with COVID-19 pneumonia. Methods We conducted a retrospective cohort study at Thammasat University Hospital, Thailand. Patients with COVID-19 pneumonia requiring HFNC therapy from April 2020 to September 2021 were included. The ROX index was defined as the ratio of oxygen saturation (SpO2)/fraction of inspired oxygen (FiO2) to respiratory rate. The CROX index was defined as the ratio of C-reactive protein (CRP) to the ROX index. dCROX was defined as the difference in CROX index between 24 hours and 72 hours. Weaning success was defined as the ability to sustain spontaneous breathing after separation from HFNC without any invasive or noninvasive ventilatory support for ≥48 hours or death. Results A total of 106 patients (49.1% male) were included. The mean age was 62.1 ± 16.2 years. Baseline SpO2/FiO2 was 276.1 ± 124.8. The rate of HFNC weaning success within 14 days was 61.3%. The best cutoff value of the dCROX index to predict HFNC weaning success was 3.15 with 66.2% sensitivity, 70.7% specificity, and an area under the ROC curve (AUC) of 0.71 (95% CI: 0.59-0.81, p < 0.001). The best cutoff value of the ROX index was 9.13, with 75.4% sensitivity, 78.0% specificity, and an AUC of 0.79 (95% CI: 0.69-0.88, p < 0.001). Conclusions ROX index has the highest accuracy for predicting successful weaning from HFNC in patients with COVID-19 pneumonia. dCROX index is the alternative tool for this setting. However, a larger prospective cohort study is needed to verify these indices for determining separation from HFNC therapy. This trial is registered with TCTR20221107004.
Collapse
Affiliation(s)
- Pitchayapa Ruchiwit
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Kanpisut Pongtongkam
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Narongkorn Saiphoklang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| |
Collapse
|